首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 93 毫秒
1.
Riboflavin significantly enhanced the efficacy of simulated solar disinfection (SODIS) at 150 watts per square meter (W m−2) against a variety of microorganisms, including Escherichia coli, Fusarium solani, Candida albicans, and Acanthamoeba polyphaga trophozoites (>3 to 4 log10 after 2 to 6 h; P < 0.001). With A. polyphaga cysts, the kill (3.5 log10 after 6 h) was obtained only in the presence of riboflavin and 250 W m−2 irradiance.Solar disinfection (SODIS) is an established and proven technique for the generation of safer drinking water (11). Water is collected into transparent plastic polyethylene terephthalate (PET) bottles and placed in direct sunlight for 6 to 8 h prior to consumption (14). The application of SODIS has been shown to be a simple and cost-effective method for reducing the incidence of gastrointestinal infection in communities where potable water is not available (2-4). Under laboratory conditions using simulated sunlight, SODIS has been shown to inactivate pathogenic bacteria, fungi, viruses, and protozoa (6, 12, 15). Although SODIS is not fully understood, it is believed to achieve microbial killing through a combination of DNA-damaging effects of ultraviolet (UV) radiation and thermal inactivation from solar heating (21).The combination of UVA radiation and riboflavin (vitamin B2) has recently been reported to have therapeutic application in the treatment of bacterial and fungal ocular pathogens (13, 17) and has also been proposed as a method for decontaminating donor blood products prior to transfusion (1). In the present study, we report that the addition of riboflavin significantly enhances the disinfectant efficacy of simulated SODIS against bacterial, fungal, and protozoan pathogens.Chemicals and media were obtained from Sigma (Dorset, United Kingdom), Oxoid (Basingstoke, United Kingdom), and BD (Oxford, United Kingdom). Pseudomonas aeruginosa (ATCC 9027), Staphylococcus aureus (ATCC 6538), Bacillus subtilis (ATCC 6633), Candida albicans (ATCC 10231), and Fusarium solani (ATCC 36031) were obtained from ATCC (through LGC Standards, United Kingdom). Escherichia coli (JM101) was obtained in house, and the Legionella pneumophila strain used was a recent environmental isolate.B. subtilis spores were produced from culture on a previously published defined sporulation medium (19). L. pneumophila was grown on buffered charcoal-yeast extract agar (5). All other bacteria were cultured on tryptone soy agar, and C. albicans was cultured on Sabouraud dextrose agar as described previously (9). Fusarium solani was cultured on potato dextrose agar, and conidia were prepared as reported previously (7). Acanthamoeba polyphaga (Ros) was isolated from an unpublished keratitis case at Moorfields Eye Hospital, London, United Kingdom, in 1991. Trophozoites were maintained and cysts prepared as described previously (8, 18).Assays were conducted in transparent 12-well tissue culture microtiter plates with UV-transparent lids (Helena Biosciences, United Kingdom). Test organisms (1 × 106/ml) were suspended in 3 ml of one-quarter-strength Ringer''s solution or natural freshwater (as pretreated water from a reservoir in United Kingdom) with or without riboflavin (250 μM). The plates were exposed to simulated sunlight at an optical output irradiance of 150 watts per square meter (W m−2) delivered from an HPR125 W quartz mercury arc lamp (Philips, Guildford, United Kingdom). Optical irradiances were measured using a calibrated broadband optical power meter (Melles Griot, Netherlands). Test plates were maintained at 30°C by partial submersion in a water bath.At timed intervals for bacteria and fungi, the aliquots were plated out by using a WASP spiral plater and colonies subsequently counted by using a ProtoCOL automated colony counter (Don Whitley, West Yorkshire, United Kingdom). Acanthamoeba trophozoite and cyst viabilities were determined as described previously (6). Statistical analysis was performed using a one-way analysis of variance (ANOVA) of data from triplicate experiments via the InStat statistical software package (GraphPad, La Jolla, CA).The efficacies of simulated sunlight at an optical output irradiance of 150 W m−2 alone (SODIS) and in the presence of 250 μM riboflavin (SODIS-R) against the test organisms are shown in Table Table1.1. With the exception of B. subtilis spores and A. polyphaga cysts, SODIS-R resulted in a significant increase in microbial killing compared to SODIS alone (P < 0.001). In most instances, SODIS-R achieved total inactivation by 2 h, compared to 6 h for SODIS alone (Table (Table1).1). For F. solani, C. albicans, ands A. polyphaga trophozoites, only SODIS-R achieved a complete organism kill after 4 to 6 h (P < 0.001). All control experiments in which the experiments were protected from the light source showed no reduction in organism viability over the time course (results not shown).

TABLE 1.

Efficacies of simulated SODIS for 6 h alone and with 250 μM riboflavin (SODIS-R)
OrganismConditionaLog10 reduction in viability at indicated h of exposureb
1246
E. coliSODIS0.0 ± 0.00.2 ± 0.15.7 ± 0.05.7 ± 0.0
SODIS-R1.1 ± 0.05.7 ± 0.05.7 ± 0.05.7 ± 0.0
L. pneumophilaSODIS0.7 ± 0.21.3 ± 0.34.8 ± 0.24.8 ± 0.2
SODIS-R4.4 ± 0.04.4 ± 0.04.4 ± 0.04.4 ± 0.0
P. aeruginosaSODIS0.7 ± 0.01.8 ± 0.04.9 ± 0.04.9 ± 0.0
SODIS-R5.0 ± 0.05.0 ± 0.05.0 ± 0.05.0 ± 0.0
S. aureusSODIS0.0 ± 0.00.0 ± 0.06.2 ± 0.06.2 ± 0.0
SODIS-R0.2 ± 0.16.3 ± 0.06.3 ± 0.06.3 ± 0.0
C. albicansSODIS0.2 ± 0.00.4 ± 0.10.5 ± 0.11.0 ± 0.1
SODIS-R0.1 ± 0.00.7 ± 0.15.3 ± 0.05.3 ± 0.0
F. solani conidiaSODIS0.2 ± 0.10.3 ± 0.00.2 ± 0.00.7 ± 0.1
SODIS-R0.3 ± 0.10.8 ± 0.11.3 ± 0.14.4 ± 0.0
B. subtilis sporesSODIS0.3 ± 0.00.2 ± 0.00.0 ± 0.00.1 ± 0.0
SODIS-R0.1 ± 0.10.2 ± 0.10.3 ± 0.30.1 ± 0.0
SODIS (250 W m−2)0.1 ± 0.00.1 ± 0.10.1 ± 0.10.0 ± 0.0
SODIS-R (250 W m−2)0.0 ± 0.00.0 ± 0.00.2 ± 0.00.4 ± 0.0
SODIS (320 W m−2)0.1 ± 0.10.1 ± 0.00.0 ± 0.14.3 ± 0.0
SODIS-R (320 W m−2)0.1 ± 0.00.1 ± 0.10.9 ± 0.04.3 ± 0.0
A. polyphaga trophozoitesSODIS0.4 ± 0.20.6 ± 0.10.6 ± 0.20.4 ± 0.1
SODIS-R0.3 ± 0.11.3 ± 0.12.3 ± 0.43.1 ± 0.2
SODIS, naturalc0.3 ± 0.10.4 ± 0.10.5 ± 0.20.3 ± 0.2
SODIS-R, naturalc0.2 ± 0.11.0 ± 0.22.2 ± 0.32.9 ± 0.3
A. polyphaga cystsSODIS0.4 ± 0.10.1 ± 0.30.3 ± 0.10.4 ± 0.2
SODIS-R0.4 ± 0.20.3 ± 0.20.5 ± 0.10.8 ± 0.3
SODIS (250 W m−2)0.0 ± 0.10.2 ± 0.30.2 ± 0.10.1 ± 0.2
SODIS-R (250 W m−2)0.4 ± 0.20.3 ± 0.20.8 ± 0.13.5 ± 0.3
SODIS (250 W m−2), naturalc0.0 ± 0.30.2 ± 0.10.1 ± 0.10.2 ± 0.1
SODIS-R (250 W m−2), naturalc0.1 ± 0.10.2 ± 0.20.6 ± 0.13.4 ± 0.2
Open in a separate windowaConditions are at an intensity of 150 W m−2 unless otherwise indicated.bThe values reported are means ± standard errors of the means from triplicate experiments.cAdditional experiments for this condition were performed using natural freshwater.The highly resistant A. polyphaga cysts and B. subtilis spores were unaffected by SODIS or SODIS-R at an optical irradiance of 150 W m−2. However, a significant reduction in cyst viability was observed at 6 h when the optical irradiance was increased to 250 W m−2 for SODIS-R only (P < 0.001; Table Table1).1). For spores, a kill was obtained only at 320 W m−2 after 6-h exposure, and no difference between SODIS and SODIS-R was observed (Table (Table1).1). Previously, we reported a >2-log kill at 6 h for Acanthamoeba cysts by using SODIS at the higher optical irradiance of 850 W m−2, compared to the 0.1-log10 kill observed here using the lower intensity of 250 W m−2 or the 3.5-log10 kill with SODIS-R.Inactivation experiments performed with Acanthamoeba cysts and trophozoites suspended in natural freshwater gave results comparable to those obtained with Ringer''s solution (P > 0.05; Table Table1).1). However, it is acknowledged that the findings of this study are based on laboratory-grade water and freshwater and that differences in water quality through changes in turbidity, pH, and mineral composition may significantly affect the performance of SODIS (20). Accordingly, further studies are indicated to evaluate the enhanced efficacy of SODIS-R by using natural waters of varying composition in the areas where SODIS is to be employed.Previous studies with SODIS under laboratory conditions have employed lamps delivering an optical irradiance of 850 W m−2 to reflect typical natural sunlight conditions (6, 11, 12, 15, 16). Here, we used an optical irradiance of 150 to 320 W m−2 to obtain slower organism inactivation and, hence, determine the potential enhancing effect of riboflavin on SODIS.In conclusion, this study has shown that the addition of riboflavin significantly enhances the efficacy of simulated SODIS against a range of microorganisms. The precise mechanism by which photoactivated riboflavin enhances antimicrobial activity is unknown, but studies have indicated that the process may be due, in part, to the generation of singlet oxygen, H2O2, superoxide, and hydroxyl free radicals (10). Further studies are warranted to assess the potential benefits from riboflavin-enhanced SODIS in reducing the incidence of gastrointestinal infection in communities where potable water is not available.  相似文献   

2.
Feeding high levels of zinc oxide to piglets significantly increased the relative abundance of ileal Weissella spp., Leuconostoc spp., and Streptococcus spp., reduced the occurrence of Sarcina spp. and Neisseria spp., and led to numerical increases of all Gram-negative facultative anaerobic genera. High dietary zinc oxide intake has a major impact on the porcine ileal bacterial composition.Zinc oxide (ZnO) is used as a feed additive for diarrhea prophylaxis in piglets (23). However, the mode of action of ZnO is not fully understood. Besides its effects on the host (10, 30, 31), high dietary zinc levels may affect the diversity of intestinal microbial communities (2, 11, 20). The prevention of postweaning diarrhea in piglets due to high dietary ZnO intake may not be directly related to a reduction of pathogenic E. coli (8) but, rather, to the diversity of the coliform community (15). Studies on the impact of high ZnO levels on the porcine ileal bacterial community are scarce but nevertheless important, as bacterial diarrhea is initiated in the small intestine (9, 17). The small intestine is a very complex habitat with many different factors shaping the bacterial community. Studies on the ecophysiology (22) and maturation of the porcine ileal microbiota (13, 27) indicate a drastic impact directly after weaning and a gradual decline of modifications during the following 2 weeks. Thus, the time point for analysis chosen in this study (14 days postweaning) does reflect a more stable period of the ileal porcine microbiota. In this study, we used bar-coded pyrosequencing of 16S rRNA genes to gain further insight into the mode of action of pharmacological levels of ZnO in the gastrointestinal tract of young pigs.Total DNA was extracted from the ileal digesta of 40- to 42-day-old piglets using a commercial kit (Qiagen stool kit; Qiagen, Hilden, Germany) and PCR amplified with unique bar-coded primer sets targeting the V1-to-V3 and the V6-to-V8 hypervariable regions (see the supplemental material for detailed methods). The rationale behind this approach was derived from the fact that no single “universal” primer pair can completely cover a complex bacterial habitat (4, 24, 32, 33). Furthermore, these studies also show that in silico information on the coverage of selected primer sets diverges from empirical results, and hence, two hypervariable regions were chosen in this study to maximize the detection of phylogenetically diverse bacterial groups.Equimolar dilutions of all samples were combined into one master sample. Pyrosequencing was performed by Agowa (Berlin, Germany) on a Roche genome sequencer FLX system using a Titanium series PicoTiterPlate. The resulting data files were uploaded to the MG-RAST server (http://metagenomics.nmpdr.org/) (19) and processed with its SEED software tool using the RDP database (5) as the reference database. After automated sequence analysis, all sequences with less than five identical reads per sample were deleted in order to increase the confidence of sequence reads and reduce bias from possible sequencing errors (12, 16). Thus, 0.43% of all sequences were not considered (1,882 of 433,302 sequences). These sequences were assigned to a total of 238 genera, of which most only occurred in a few samples (see the supplemental material). Furthermore, all unclassified sequences were removed (8.7%; 41,467 of 474,769 sequences). Due to the use of the RDP reference database, the SEED software incorrectly assigned the majority of unclassified sequences as unclassified Deferribacterales (83%; 34,393 sequences), which were actually identified as 16S soybean or wheat chloroplasts by BLAST or as cyanobacterial chloroplasts by the RDP II seqmatch tool.The pyrosequencing results for the two primer combinations were merged by taking only sequences from the primer combination that yielded the higher number of reads for a specific sequence assignment in a sample. The remaining reads were used to calculate the relative contribution of assigned sequences to total sequence reads in a sample.The Firmicutes phylum dominated the small intestinal bacterial communities in both the control group and the group with high dietary ZnO intake, with 98.3% and 97.0% of total sequence reads, respectively. No significant influence of high dietary ZnO intake was found for the main phyla Proteobacteria (0.92% versus 1.84%), Actinobacteria (0.61% versus 0.75%), Bacteroidetes (0.15% versus 0.17%), and Fusobacteria (0.09% versus 0.12%).On the order level, a total of 20 bacterial orders were detected (data not shown). Lactobacillales dominated bacterial communities in the control and high-dietary-ZnO-intake groups, with 83.37% and 93.24% of total reads. Lactic acid bacteria are well known to dominate the bacterial community in the ileum of piglets (11, 22). No significant difference between the control group and the group with high dietary ZnO intake was observed on the order level, although high dietary ZnO intake led to a strong numerical decrease for Clostridiales (14.4 ± 24.0% [mean ± standard deviation] versus 2.8 ± 1.7%), as well as to numerical increases for Pseudomonadales (0.3 ± 0.3% versus 0.6 ± 0.6%) and Enterobacteriales (0.2 ± 0.2% versus 0.5 ± 0.6%).On the genus level, a total of 103 genera were detected. Table Table11 summarizes the main 31 genera which exceeded 0.05% of total reads (see the supplemental material for a complete list). Lactobacilli clearly dominated the bacterial communities in both trial groups, but they also were numerically lower due to high dietary ZnO intake.

TABLE 1.

Bacterial genera in the ileum of piglets fed diets supplemented with 200 or 3,000 ppm ZnO
GenusProportion (% ± SD) of ileal microbiota in groupa receiving:
200 ppm ZnO3,000 ppm ZnO
Lactobacillus59.3 ± 30.640.7 ± 19.1
Weissella11.6 ± 7.8 A24.1 ± 8.3 B
Sarcina11.4 ± 20.5 A0.84 ± 1.2 B
Leuconostoc4.7 ± 3.2 A9.4 ± 3.1 B
Streptococcus1.8 ± 1.6 A5.7 ± 5.1 B
Lactococcus1.6 ± 1.52.6 ± 3.1
Veillonella0.57 ± 0.630.34 ± 0.30
Gemella0.34 ± 0.67 A0.45 ± 0.25 B
Acinetobacter0.25 ± 0.210.44 ± 0.50
Clostridium0.25 ± 0.400.22 ± 0.21
Enterococcus0.19 ± 0.150.26 ± 0.24
Acidovorax0.14 ± 0.040.16 ± 0.19
Arcobacter0.14 ± 0.150.16 ± 0.17
Neisseria0.14b0.03 ± 0.01
Enterobacter0.13 ± 0.090.29 ± 0.34
Lachnospira0.12 ± 0.130.13 ± 0.03
Peptostreptococcus0.11 ± 0.100.07 ± 0.09
Chryseobacterium0.10 ± 0.070.15 ± 0.16
Actinomyces0.09 ± 0.040.15 ± 0.16
Anaerobacter0.07 ± 0.080.02 ± 0.01
Aerococcus0.07 ± 0.040.07 ± 0.04
Dorea0.07b0.05 ± 0.05
Fusobacterium0.06 ± 0.090.08 ± 0.11
Microbacterium0.06 ± 0.010.07 ± 0.04
Carnobacterium0.06 ± 0.020.08 ± 0.13
Granulicatella0.06 ± 0.020.09 ± 0.10
Staphylococcus0.06 ± 0.040.05 ± 0.02
Facklamia0.05 ± 0.060.03 ± 0.01
Comamonas0.05 ± 0.030.04 ± 0.02
Citrobacter0.05 ± 0.020.07 ± 0.08
Erysipelothrix0.05 ± 0.010.22 ± 0.40
Open in a separate windowan = 6 piglets per trial group. A,B, results are significantly different by Kruskal-Wallis test.bSingle sample.Significant changes due to high dietary ZnO intake were observed for other lactic acid bacteria, including Weissella spp., Leuconostoc spp., and Streptococcus spp. A significant and strong decrease was observed for Sarcina spp., which is a genus of acid-tolerant strictly anaerobic species found in the intestinal tract of piglets and other mammals (6, 28, 29). This genus thus appeared to be very sensitive to modifications induced by high dietary ZnO intake.An interesting result was observed for Gram-negative Proteobacteria, (i.e., enterobacteria and relatives). Although not statistically significant, virtually all detected proteobacteria increased numerically due to high dietary ZnO intake (Enterobacter spp., Microbacterium spp., Citrobacter spp., Neisseria spp., and Acinetobacter spp.). Apparently, enterobacteria gained colonization potential by high dietary ZnO intake. This is in good agreement with the results of studies by Hojberg et al. (11), Amezcua et al. (1), and Castillo et al. (3). Therefore, the frequently observed diarrhea-reducing effect of zinc oxide may not be directly related to a reduction of pathogenic E. coli strains. Considering a possible antagonistic activity of lactobacilli against enterobacteria (25), it can be speculated that a numerical decrease of dominant lactobacilli may lead to increased colonization with Gram-negative enterobacteria. On the other hand, specific plasmid-borne genes for resistance against heavy metals have been reported for both Gram-positive and Gram-negative bacteria present in the intestine (21, 26), and an increased resistance against Zn ions may exist for Gram-negative enterobacteria. Zinc oxide is an amphoteric molecule and shows a high solubility at acid pH. The low pH in the stomach of piglets (pH 3.5 to 4.5) transforms a considerable amount of insoluble ZnO into zinc ions (54 to 84% free Zn2+ at 150 ppm and 24 ppm ZnO, respectively) (7), and thus, high concentrations of toxic zinc ions exist in the stomach. The stomach of piglets harbors large numbers of lactic acid bacteria, especially lactobacilli. Zn ions may thus lead to a modification of the lactic acid bacterial community in the stomach, and the changes observed in the ileum could have been created in the stomach. A reduction of dominant lactobacilli may thus point to an increased adaptation potential of Gram-negative facultative anaerobes and a generally increased bacterial diversity.Additionally, the direct effects of dietary ZnO on intestinal tissues include altered expression of genes responsible for glutathione metabolism and apoptosis (30), enhanced gastric ghrelin secretion, which increases feed intake (31), and increased production of digestive enzymes (10). An analysis of the intestinal morphology was beyond the scope of this study, but although ZnO concentrations are markedly increased in intestinal tissue, the influence of ZnO on morphology is apparently not always observed (10, 14, 18). Consequently, any changes in epithelial cell turnover, feed intake, or digestive capacity may influence the composition of bacterial communities in the small intestine.In conclusion, this study has shown that high dietary zinc oxide has a major impact on ileal bacterial communities in piglets. Future studies on the impact of zinc oxide in pigs should include a detailed analysis of host responses in order to identify the cause for the observed modifications of intestinal bacterial communities.  相似文献   

3.
4.
At present there is little quantitative information on the identity and composition of bacterial populations in the rumen microbial community. Quantitative fluorescence in situ hybridization using newly designed oligonucleotide probes was applied to identify the microbial populations in liquid and solid fractions of rumen digesta from cows fed barley silage or grass hay diets with or without flaxseed. Bacteroidetes, Firmicutes, and Proteobacteria were abundant in both fractions, constituting 31.8 to 87.3% of the total cell numbers. They belong mainly to the order Bacteroidales (0.1 to 19.2%), hybridizing with probe BAC1080; the families Lachnospiraceae (9.3 to 25.5%) and Ruminococcaceae (5.5 to 23.8%), hybridizing with LAC435 and RUM831, respectively; and the classes Deltaproteobacteria (5.8 to 28.3%) and Gammaproteobacteria (1.2 to 8.2%). All were more abundant in the rumen communities of cows fed diets containing silage (75.2 to 87.3%) than in those of cows fed diets containing hay (31.8 to 49.5%). The addition of flaxseed reduced their abundance in the rumens of cows fed silage-based diets (to 45.2 to 58.7%) but did not change markedly their abundance in the rumens of cows fed hay-based diets (31.8 to 49.5%). Fibrolytic species, including Fibrobacter succinogenes and Ruminococcus spp., and archaeal methanogens accounted for only a small proportion (0.4 to 2.1% and 0.2 to 0.6%, respectively) of total cell numbers. Depending on diet, between 37.0 and 91.6% of microbial cells specifically hybridized with the probes used in this study, allowing them to be identified in situ. The identities of other microbial populations (8.4 to 63.0%) remain unknown.The rumen is an anaerobic ecosystem used by herbivores to convert fibrous plant material into fermentation products that are in turn used as energy by the host. Fibrolytic degradation is accomplished by a complex microbial community which includes specialized fungi, protozoa, and bacteria (14). More than 200 bacterial species (5) have been isolated from rumen, and many of these have been phylogenetically and physiologically characterized. Several of these, including Fibrobacter succinogenes, Ruminococcus albus, and Ruminococcus flavefaciens, have the ability to hydrolyze cellulose in axenic culture (24). Despite the presence of these fibrolytic populations, a large portion of the fiber in low-quality forage diets passes through the rumen undigested. In the rumen, fibrolytic bacteria do not digest plant cell walls in isolation but rather interact with a consortium of bacteria (18). Although culture-dependent studies have improved our understanding of rumen microbiology, the importance of the isolates to the structure and function of the rumen microbial community, with the possible exception of the fibrolytic strains, is still unknown. Expanding our knowledge of the structure and function of the rumen microbial community may provide insights into approaches to improve the efficiency of fiber digestion and biofuel production (14).To provide a high-resolution view of the population structure of the rumen bacterial community, we used quantitative fluorescence in situ hybridization (qFISH) to investigate the composition and distribution of bacterial populations associated with the liquid and solid rumen contents from 12 ruminally cannulated Holstein dairy cows (3 cows were used for each diet) fed (for at least 21 days) grass hay or barley silage diets with or without flaxseed (Table (Table1).1). Six new 16S rRNA-targeted FISH probes (Table (Table2)2) for not only the fibrolytic groups but also other unclassified bacterial groups in the rumen were designed, using ARB software (17), against the rumen 16S rRNA gene sequences (data not shown) retrieved from the Ribosomal Database Project (RDP) database (6). The new probes target Bacteroidales-related clones (probe BAC1080) (phylum Bacteroidetes), Lachnospiraceae- and Ruminococcaceae-related clones (probes LAC435 and RUM831, respectively) (phylum Firmicutes), Butyrivibrio fibrisolvens-related clones (probe BFI826), and R. albus- and R. flavefaciens-related clones (probes RAL1436 and RFL155, respectively).

TABLE 1.

Composition of diets used in this study
IngredientDiet composition (% dry weight)
Hay-based dietHay and flaxseed dietSilage-based dietSilage and flaxseed diet
Alfalfa grass hay (chopped)47.547.500
Barley silage0047.547.5
Steamed rolled barley grain47.532.547.532.5
Ground flaxseeds015015
Other5555
Open in a separate window

TABLE 2.

Oligonucleotide probes and their target populations used in this study for FISH analyses
Probe nameaTarget rRNADesigned target(s)% FAbReference
EUB338 (00159)16SDomain Bacteria0-5016
EUB338II (00160)16SPhylum Planctomycetes0-5016
EUB338III (00161)16SPhylum Verrucomicrobia0-5016
NONEUB (00243)16SControl probe complementary to EUB3380-5016
ALF968 (00021)16SClass Alphaproteobacteria, phylum Proteobacteria2016
BET42a (00034)23SClass Betaproteobacteria, phylum Proteobacteria3516
GAM42a (00174)23SClass Gammaproteobacteria, phylum Proteobacteria3516
SRB385 (00300)16SClass Deltaproteobacteria, phylum Proteobacteria3516
SRB385Db (00301)16SClass Deltaproteobacteria, phylum Proteobacteria3516
HGC69a (00182)23SPhylum Actinobacteria2516
GNSB941 (00718)16SPhylum Chloroflexi3516
CFX1223 (00719)16SPhylum Chloroflexi3516
SPIRO1400 (01004)16SSubgroup of family Spirochaetaceae2016
TM7-905 (00600)16SCandidate phylum TM72016
LGC354A (00195)16SPhylum Firmicutes3516
LGC354B (00196)16SPhylum Firmicutes3516
LGC354C (00197)16SPhylum Firmicutes3516
RUM83116SRumen clones in family Ruminococcaceae, phylum Firmicutes35This study
RAL143616SRuminococcus albus-related clones, phylum Firmicutes20This study
RFL15516SRuminococcus flavefaciens-related clones, phylum Firmicutes45This study
LAC43516SClones in family Lachnospiraceae, phylum Firmicutes35This study
BFI82616SButyrivibrio fibrisolvens-related clones, phylum Firmicutes35This study
BAC108016SClones in order Bacteroidales, phylum Bacteroidetes20This study
Fibr225 (00005)16SFibrobacter succinogenes-related clones, phylum Fibrobacteres20c16
ARCH915 (00027)16SDomain Archaea2016
Open in a separate windowaThe numbers in parentheses after the probe names represent the probe accession numbers in probeBase (16).bFA, formamide concentration used in the FISH buffer.cThe optimum formamide concentration for the probe was determined in this study.The optimal formamide concentrations (OFC) of the new probes used in FISH were assessed in different ways. Probes RUM831 and BAC1080 were assessed by using pure cultures of Ruminococcus and Prevotella strains with zero and one mismatch (Fig. (Fig.1)1) to the probes. The OFC of probes LAC435 and BFI826 were assessed using Clone-FISH (21) with zero and one mismatch 16S rRNA clone (Fig. (Fig.1)1) by following the procedure described previously (9, 10). The highest formamide concentration (tested in 5% stepwise increases) at which a clear fluorescent signal was observed with the reference bacterium or competent cells with zero mismatches after FISH probing, but not with bacteria or competent cells with one mismatch, was selected. The OFC of probes FIB225 (designed by Stahl et al. [23]), RFL155, and RAL1436 were assessed using only pure cultures of F. succinogenes, R. flavefaciens, and R. albus, respectively, all having perfect matches to each probe (Fig. (Fig.1).1). The highest formamide concentration (tested in 5% stepwise increases) at which a clear fluorescent signal was observed with the reference bacterium after FISH probing was selected. These probes were employed with other available probes (Table (Table2)2) chosen from probeBase (16) based on the alignment and classification of the 16S rRNA gene sequences retrieved from rumen communities.Open in a separate windowFIG. 1.Alignments of the probe sequences and their target sites and sequences of corresponding sites in reference bacteria or clones. The probe names in parentheses after the abbreviated names are according to Oligonucleotide Probe Database nomenclature (2). Only the nucleotides that are different from target sequences are shown. E, empty space; R., Ruminococcus; P., Prevotella; F., Fibrobacter.The digest samples from the top, bottom, and middle of the rumen were collected through a cannula, thoroughly mixed, and fractioned as liquid fraction (LiqF) and solid fraction (SolF). On-site, about 100 ml was transferred to a heavy-wall 250-ml beaker and squeezed using a Bodum coffee maker plunger (Bodum Inc., Triengen, Switzerland). The extruded liquid samples (containing the planktonic cells) were fixed in ethanol and paraformaldehyde (PFA) for FISH probing (3). The remaining liquid was discarded, and the squeezed particulate samples (used to collect particulate-attached cells) were washed with 100 ml phosphate buffer (5.23 g/liter K2HPO4, 2.27 g/liter KH2PO4, 3.00 g/liter NaHCO3, and 20 ml/liter 2.5% cysteine HCl) by stirring gently with a spatula, followed by squeezing again and decanting. Washed particulate samples (5 g) were then fixed for FISH as described above.After fixation, the particulate samples plus the fixation solution were transferred into a stomacher bag and “stomached” (Stomacher 400 Circulator, Seaward England) at 230 rpm for 6 min. Treated samples were then transferred into a clean 250-ml beaker and squeezed again. Microscopic examination of the squeezed residues after DAPI (4′,6-diamidino-2-phenylindole) staining (100 μl [0.003 mg/ml] for 10 min) showed only a few bacterial cells attached on the plant fibers, indicating that most bacterial cells had been “stomached” into the liquid (data not shown). To recover cells, filtrates were centrifuged (5,000 × g), and the cell pellet was washed three times with phosphate buffer before being used for FISH probing. On the day of sampling, each cow was sampled twice, at 1100 h and 1600 h. The liquid FISH samples obtained from the 3 cows fed with the same diet (at two different sampling times) were mixed, as were the particulate FISH samples, and used in qFISH analysis. FISH was carried out according to Amann (3). FISH was carried out on glass coverslips (24 by 60 mm) coated with gelatin (9). DAPI staining of biomass samples was carried out after FISH probing. FISH and DAPI images were captured with a Zeiss epifluorescence microscope (Zeiss PM III) equipped with a Canon 5D Mark II camera. Raw images captured randomly were transferred into gray TIF images and sharpened in Adobe Photoshop CS3. Cells stained with DAPI and hybridized to the probes were enumerated using the function provided in ImageJ (1). The percent compositions of these probe-defined groups (against all DAPI-stained cells in the same microscopic field) in the different fractions of rumen contents from cows fed different diets are presented in Table Table33.

TABLE 3.

Distribution and composition of FISH probe-defined groups in rumen microbial communities in cows fed with different diets
Probe-defined microbial groupComposition (mean value [%] ± SD)a
Hay-based diet
Hay and flaxseed diet
Silage-based diet
Silage and flaxseed diet
LiqFSolFLiqFSolFLiqFSolFLiqFSolF
BAC10809.6 ± 1.330.1 ± 0.0219.2 ± 3.714.2 ± 0.7214.2 ± 3.1118.8 ± 3.8814.4 ± 2.8916.7 ± 4.33
ALF9680.2 ± 0.020.2 ± 0.020.2 ± 0.030.2 ± 0.040.7 ± 0.141.5 ± 0.410.1 ± 0.010.1 ± 0.01
BET42a000.6 ± 0.011.2 ± 0.270.1 ± 0.01<0.10.4 ± 0.060.2 ± 0.04
GAM42a3.2 ± 0.534.4 ± 0.574.2 ± 0.764.5 ± 0.672.0 ± 0.321.2 ± 0.238.2 ± 1.235.3 ± 0.95
SRBmix5.8 ± 0.8811.6 ± 2.439.0 ± 1.5210.1 ± 2.5628.3 ± 4.4323.3 ± 4.547.7 ± 0.7813.2 ± 2.22
CHLmix1.7 ± 0.2700.5 ± 0.010 ± 00.2 ± 0.020.4 ± 0.070.1 ± 0.010.1 ± 0.02
SPIRO14000.5 ± 0.091.9 ± 0.321.7 ± 0.332.0 ± 0.211.4 ± 0.311.9 ± 0.330.4 ± 0.030.4 ± 0.07
TM7-9050.6 ± 0.080.8 ± 0.070.5 ± 0.010.1 ± 0.031.5 ± 0.230.2 ± 0.020.6 ± 0.020.3 ± 0.08
HGC69a1.3 ± 0.282.1 ± 0.310.3 ± 0.060.3 ± 0.050.4 ± 0.030.1 ± 0.020.5 ± 0.090.2 ± 0.02
RUM8315.5 ± 0.135.7 ± 0.895.8 ± 0.738.9 ± 1.3218.0 ± 4.1323.8 ± 3.115.6 ± 1.147.4 ± 1.32
RAL14360.4 ± 0.060.3 ± 0.030.2 ± 0.060.2 ± 0.030.3 ± 0.050.6 ± 0.090.7 ± 0.130.6 ± 0.12
RFL1550.7 ± 0.110.2 ± 0.030.3 ± 0.070.7 ± 0.190.1 ± 0.010.8 ± 0.110.5 ± 0.061.2 ± 0.34
LAC43525.5 ± 3.9810.0 ± 1.519.6 ± 1.3111.7 ± 1.6712.6 ± 2.5620.2 ± 3.239.3 ± 1.5116.1 ± 3.31
BFI8260.3 ± 0.060.4 ± 0.050.4 ± 0.060.7 ± 0.120.5 ± 0.050.3 ± 0.082.4 ± 0.370.2 ± 0.02
Fibr225000.2 ± 0.040.1 ± 0.020.8 ± 0.140.7 ± 0.140.4 ± 0.110.1 ± 0.04
ARCH9150.3 ± 0.080.2 ± 0.070.6 ± 0.010.3 ± 0.070.6 ± 0.090.1 ± 0.020.4 ± 0.050.4 ± 0.06
Total hybridizedb54.13752.443.780.991.64860.7
Otherc45.96347.656.319.18.45239.3
Open in a separate windowaThe two numbers represent the mean value (%) and the standard deviation of individual probe-defined microbial groups in a specified rumen digest fraction, which were calculated based on 3 mean values, each consisting of 20 enumerations.bThe numbers represent the sum of percentages of all individual probe-defined microbial groups in a specified rumen digest fraction. The percentages obtained with FISH probes RAL1436, RFL155, and BFI826 were not counted in the sum because the bacterial cells hybridizing with the former two probes also hybridized with RUM831, and the bacterial cells hybridizing with the last probe also hybridized with probe LAC435.cThe numbers represent the percentages of microorganisms which were not identified by FISH in a specified rumen digest fraction.We provided quantitative data by using qFISH to show that Bacteroidetes, Firmicutes, and Proteobacteria were abundant in both the LiqF and the SolF, constituting 31.8 to 87.3% of the total cell numbers. These FISH data add weight to the view that Firmicutes and Bacteroidetes might be dominant in rumens, as suggested previously from their high ratios retrieved from 16S rRNA clone libraries (e.g., see references 12, 26, and 27). However, information emerging from 16S rRNA gene clone library data cannot be used to reach conclusions on the quantitative composition of the rumen bacterial community. Bacteria may have 1 to 14 copies of rRNA genes, and several biases are known to be associated with their PCR amplification (8).These 3 dominant bacterial groups have been identified at a high-resolution level. They belong mainly to the order Bacteroidales (0.1 to 19.2%), hybridizing with probe BAC1080 (Fig. (Fig.22 A); the families Lachnospiraceae (9.3 to 25.5%) and Ruminococcaceae (5.5 to 23.8%), hybridizing with LAC435 (Fig. (Fig.2E)2E) and RUM831 (Fig. (Fig.2D),2D), respectively; and the classes Deltaproteobacteria (5.8 to 28.3%) and Gammaproteobacteria (1.2 to 8.2%), hybridizing with SRBmix (equal moles of SRB385 and SRB385Db) (Fig. (Fig.2C)2C) and GAM42a (Fig. (Fig.2B),2B), respectively. All were more abundant in the microbial communities in the rumens of cows fed diets containing silage (75.2 to 87.3%) than in those in the rumens of cows fed diets containing hay (31.8 to 49.5%). These results show how diets containing different forages (hay or silage) may influence the distribution of the microbial populations, which is in line with data by Tajima et al. (25). We also found in this study that the addition of flaxseed (to inhibit methane emission) reduced their abundance in the rumens of cows fed silage-based diets (to 45.2 to 58.7%) but did not change markedly their abundance in the rumens of cows fed hay-based diets (31.8 to 49.5%), suggesting that adding flaxseed to these diets also affected rumen microbial community composition, although the extent of its influence reflected the forage used, being more profound with a silage-based diet than when hay was used.Open in a separate windowFIG. 2.Images of digest samples from the rumens of cows fed hay- or silage-based diets with and without flaxseed after color combination. Images from probes are labeled in red, and those from DAPI staining are in green. The yellow (combination of red and green), including those partly colored cells in panels A to F, hybridized with probes BAC1080, GAM42a, SRBmix, RUM831, LAC435, and ARCH915, respectively. A few cells (arrows) hybridizing with SRBmix (C) were not stained by DAPI. Bars, 10 μm.We also present evidence here to suggest that Proteobacteria are common members of the microbial community, with sulfur-reducing bacteria (SRB) belonging to Deltaproteobacteria in particular being readily detected (up to 28% of the total cells) in both the LiqF and the SolF of rumen contents from cows fed the four different diets examined here. SRB have seldom been retrieved in clone libraries obtained from rumen samples. Lin et al. (15) have estimated SRB abundance in the rumen using DNA hybridization and concluded that they were of minor importance (0.7 to 0.8% of the total rRNA). Our estimates are much higher than those for every diet regime examined, possibly reflecting the coverage of the probes used in the two different studies. The probe mixture SRBmix used here targets most members of the Deltaproteobacteria, while those of Lin et al. (15) covered mainly members of the Desulfobacteraceae, Desulfovibrionaceae, and Desulfobulbaceae. We also recognized that the probe mixture SRBmix perfectly matched with the 16S rRNA genes of some bacteria other than SRB in Deltaproteobacteria. The possibility of overestimation of SRB cannot be ruled out. Interestingly, our data suggest that Gammaproteobacteria were abundant in some of the rumen communities we examined by FISH, comprising 1.2 to 8.2% of total cells.The other unexpected finding was that the fibrolytic bacteria and archaeal methanogens accounted for only a minor fraction of the communities. Of the three characterized fibrolytic bacterial species, F. succinogenes was not detected in the rumen digesta from cattle fed the hay-based diet but was present in the remainder of the diets. In contrast, R. albus and R. flavefaciens were present in both the LiqF and the SolF of the rumen digesta from cows fed all four diets. Although the importance of these bacteria within the rumen microbial community cannot be denied, these three populations accounted for only 0.7 to 2.1% of the total microbial cells. This numerical range compares well with that determined previously for F. succinogenes (0.1 to 6.9% of total rRNA) (4, 23) and Ruminococcus spp. (1.5 to 2.9% of total rRNA) (11), considering that different animals and diets were used in those studies and that different specificities of the probes and different detection methods were used. However, this is much lower than the 9% (of total rRNA) detected by Michalet-Doreau et al. (19) in their work. The abundance of fibrolytic B. fibrisolvens-related species was also low, being present at <1% in all fractions, except in the LiqF in cows fed the mixture of silage and flaxseed, where they contributed 2.4% of total cells.Methanogens hybridized to ARCH915 (Fig. (Fig.2F)2F) were present (0.1 to 0.6%) in all rumen samples examined by FISH, which is close to or within the range (0.3 to 3.3%) estimated in other studies (15, 22). Interestingly, no marked difference in abundance of the methanogens could be seen between the samples from the rumens of cows fed diets with flaxseed and those from the rumens of cows fed diets without flaxseed, although it has been reported (7) that the addition of fatty acids could decrease methane production in the rumen. This may be due to the presence of methanogens with different activities in different rumen samples or the inability of probe ARCH915 to hybridize to all methanogens in the rumen samples examined here.Bacteria belonging to Chloroflexi, TM7, Spirochetes, and Actinobacteria hybridizing with CHLmix, TM7-905, SPRO1400, and HGC69a, respectively, accounted for only a minor fraction of the total cell numbers observed. In most cases, their abundances in each fraction did not change markedly with diet, always being present in small numbers (0 to 1%), suggesting that they have a minor role there. This conclusion, however, has to be confirmed since many (8.4 to 63.0%, depending on diet) of the bacteria could not be identified in the rumens of cows fed with all diets except the silage-based diet (Table (Table33).FISH with the probes designed in this study failed to identify all of the bacterial cells. This is because the probes do not target all rumen 16S rRNA gene sequences and/or the true extent of rumen biodiversity has not been revealed from cloning analyses. This indicates that our current understanding of the quantitative composition of the rumen microbial community is far from complete. Moreover, no physiological data were generated in this study to suggest what the role(s) of most of the dominant populations (except the SRB hybridized with probe SRBmix) identified by FISH might be, meaning that it is still not possible to link their abundance to their in situ function. Furthermore, each FISH-probed population probably includes bacteria with different phenotypes. Clearly, much needs to be done before the structure and function of the rumen microbial community are fully understood.FISH is a useful tool in the investigation of microbial composition in complex ecosystems (3). However, FISH probes targeting rumen bacterial populations are limited. By comparison with other culture-independent methods, e.g., quantitative PCR, FISH has several advantages (8). In particular, in combination with histochemical staining methods (20) and microautoradiography (MAR-FISH) (13), the in situ ecophysiology of a targeted population can be determined under specified electron acceptor conditions. These techniques may provide important clues as to the functional role of microbial populations within complex communities, like that of the rumen. The possession of the FISH probes described in this paper could allow such studies to be undertaken in herbivore rumens.  相似文献   

5.
We used a mixture of surrogates (Acinetobacter baumannii, Mycobacterium terrae, hepatitis A virus, and spores of Geobacillus stearothermophilus) for bioagents in a standardized approach to test environmental surface disinfectants. Each carrier containing 10 μl of mixture received 50 μl of a test chemical or saline at 22 ± 2°C. Disinfectant efficacy criteria were ≥6 log10 reduction for the bacteria and the spores and ≥3 log10 reduction for the virus. Peracetic acid (1,000 ppm) was effective in 5 min against the two bacteria and the spores but not against the virus. Chlorine dioxide (CD; 500 and 1,000 ppm) and domestic bleach (DB; 2,500, 3,500, and 5,000 ppm) were effective in 5 min, except for sporicidal activity, which needed 20 min of contact with either 1,000 ppm of CD or the two higher concentrations of DB.Disinfectant testing with a single type of organism does not represent field conditions, where bioagents or other pathogens may be mixed with other contaminants. Such an approach also cannot predict the true spectrum of microbicidal activity of a given chemical, while the identity of the target pathogen(s) is often unknown. We used a mixture of Acinetobacter baumannii, Mycobacterium terrae (15), hepatitis A virus (HAV) (4), and the spores of Geobacillus stearothermophilus as surrogates for infectious bioagents, with an added soil load on disks (1 cm in diameter; 0.75 mm thick) of brushed stainless steel (AISI no. 430; Muzeen & Blythe, Winnipeg, MB, Canada), to better simulate environmental surface disinfection (1, 11). Table Table11 gives details on the microbial strains, media used for their culture and recovery, and methods for preparing working stocks. The quantitative carrier test (QCT) method, ASTM standard E-2197 (1), was used to test the organisms singly and in a mixture. Each 200 μl of the inoculum contained 34 μl each of the four organisms, 40 μl of bovine mucin, 14 μl of yeast extract, and 10 μl of bovine serum albumin stocks.

TABLE 1.

Organisms in the mixture and their growth/recovery media and titers
Organism (ATCC no.)Growth/recovery medium or host cell lineProcedure for culture and prepn of stockViability titer in stock
Mycobacterium terrae pBEN genetically modified in-house (ATCC 15755)Middlebrook 7H11 agar, OADC,a and kanamycin (10 μg/ml); incubation 20 days at 36 ± 1°C7H9 broth with ADCb and glycerol; cells washed and resuspended in deionized water (8 ml) in a Bijoux bottle (Wheaton, Millville, NJ) with glass beads (Sigma-Aldrich; 3 mm in diam; catalog no. Z143928) and stored at 4°C3.7 × 109 CFU/ml
Geobacillus stearothermophilus (ATCC 12980)Trypticase soy agar plates incubated at 56°C for 48 hSpores heat shocked at 100°C for 45 min, washed in deionized H2O, and stored at 4°C1.5 × 108 CFU/ml
Acinetobacter baumannii (ATCC 19606)Trypticase soy agar plates incubated at 36 ± 1°C for 24 hInoculated into Trypticase soy broth and incubated for 24 h at 36 ± 1°C, broth centrifuged, and pellet resuspended in deionized H2O and stored at 4°C1.2 × 109 CFU/ml
Hepatitis A virus (ATCC VR-1402)FRhK-4 cells (CRL-1688) infected and incubated for 6 daysCells grown in MEMc with 7% (vol/vol) fetal bovine serum (Fisher; M33-500) and 1% nonessential amino acids (Gibco; 11140) at 36 ± 1°C, monolayers infected and incubated at 36 ± 1°C for 7 days in medium with no antibiotics, flasks frozen and thawed (thrice), cell lysate centrifuged, and supernatant aliquoted for storage at −80°C8 × 108 PFU/ml
Open in a separate windowaOADC, oleic acid-albumin-dextrose-catalase.bADC, albumin dextrose-catalase.cMEM, minimal essential medium.Disinfectants tested were peracetic acid (PAA; 500 and 1,000 ppm), chlorine dioxide (CD; 500 and 1,000 ppm), and domestic bleach (DB; 2,500, 3,300, and 5,000 ppm). Buffered saline (pH 7.2) was the control fluid, eluent, and diluent. Hard water (400 ppm CaCO3) was the diluent for disinfectants (1).Each disk received 10 μl of the inoculum, dried and covered with 50 μl of test substance, or saline at 22 ± 2°C. At the end of the contact time, each disk was eluted in a neutralizer and the eluates were assayed (1, 9, 11, 12). The neutralizer consisted of 1% dextrose (Difco), 0.7% lecithin (Alfa Aesar), 0.25% sodium bisulfite (J. T. Baker), 0.1% sodium thioglycolate (Sigma), 0.6% sodium thiosulfate (Analar), 0.2% l-cysteine (Sigma), 0.5% tryptone (Oxoid), and 0.1% Tween 80 (Bioshop) in buffered saline (pH 7.2). In each experiment, three control and three test carriers were used, and all experiments were repeated thrice. The performance criteria for the tested substances were ≥3.0 log10 reduction in PFU of the virus and ≥6.0 log10 reductions in the CFU for the other three organisms. When the mixture of test organisms was used, the components were separated by first passing the mixture through a membrane filter (0.22-μm pore diameter) to retain all the organisms except the virus. The filtrate was subjected to plaque assays for HAV in FRhK-4 cells. For the three bacteria, separate filters were placed on appropriate agar plates (Table (Table1)1) and incubated.The data for 5-min contact are given in Table Table2.2. All levels of the disinfectants tested met the criterion for M. terrae and A. baumannii when tested individually or in mixture. Only 1,000 ppm of PAA was effective against the spores. Both levels of PAA were ineffective against HAV, while the other disinfectants could reduce its titer between 3.5 and 4 log10. Only 1,000 ppm of PAA could consistently meet the criterion for sporicidal activity after 10 min (data not shown). Extending the contact time to 20 min allowed both levels of PAA and DB to meet the criterion for sporicidal activity, while 500 ppm of CD failed to do so; CD at 1,000 ppm barely met the criterion when tested alone against the spores but could not do so in the mixture (Fig. (Fig.11).Open in a separate windowFIG. 1.Reductions of G. stearothermophilus spores by the test formulations after 20 min of contact, individually and in a mixture at 22 ± 2°C.

TABLE 2.

Reductions by the test formulations in 5 min at 22 ± 2°C when tested against each organism individually and in a mixture
Disinfectant (concn [ppm])Mean log10 reduction ± SD of:
M. terrae
A. baumannii
G. stearothermophilus
Hepatitis A virus
IndividualMixtureIndividualMixtureIndividualMixtureIndividualMixture
Peracetic acid (500)8.18 ± 0.197.33 ± 0.167.19 ± 0.036.33 ± 0.034.03 ± 0.084.45 ± 0.98Not tested0.30 ± 0.01
Peracetic acid (1,000)8.18 ± 0.197.33 ± 0.167.19 ± 0.036.33 ± 0.038.03 ± 0.287.21 ± 0.590.58 ± 0.220.68 ± 0.09
Chlorine dioxide (500)8.18 ± 0.197.72 ± 0.217.22 ± 0.036.37 ± 0.131.47 ± 0.450.69 ± 0.054.30 ± 0.183.97 ± 0.19
Chlorine dioxide (1,000)8.18 ± 0.197.72 ± 0.217.22 ± 0.036.37 ± 0.133.07 ± 0.091.27 ± 0.054.30 ± 0.183.97 ± 0.19
Domestic bleach (2,500)8.18 ± 0.197.72 ± 0.217.22 ± 0.036.37 ± 0.130.27 ± 0.030.25 ± 0.024.41 ± 0.233.97 ± 0.29
Domestic bleach (3,500)8.18 ± 0.197.72 ± 0.217.22 ± 0.036.37 ± 0.130.27 ± 0.030.25 ± 0.024.41 ± 0.233.45 ± 0.09
Domestic bleach (5,000)8.18 ± 0.197.72 ± 0.217.22 ± 0.036.37 ± 0.130.28 ± 0.010.25 ± 0.024.41 ± 0.233.97 ± 0.29
Open in a separate windowThe study showed the feasibility of testing liquid chemicals against a mixture of suitable surrogates for infectious bioagents. This approach allowed standardized and simultaneous assessment of the spectrum of microbicidal activities of the test formulations under identical conditions that better simulate field conditions and that can be readily adapted to test foams and gaseous chemicals on other carrier materials. The surrogates selected covered the spectrum of microbicide resistances of all currently known classes of infectious bioagents.A. baumannii is among the more environmentally stable and microbicide-resistant vegetative bacteria known (7, 13). M. terrae represented pathogens with generally higher resistance to microbicides (3) and possibly drug-resistant Mycobacterium tuberculosis and category C agents (6). HAV, a small, nonenveloped virus known for its stability and microbicide resistance (9), represented select agents (CBW, biological weapons classification, 2001 [http://www.selectagents.gov/Select%20Agents%20and%20Toxins%20List.html]) and also food- and waterborne pathogens listed as biothreats (2, 10). The spores of G. stearothermophilus may be more resistant to oxidizing chemicals than the spores of Bacillus anthracis (8); their thermophilic nature made them safer to handle and easy to separate from the mixtures.The disinfectants were selected for their commercial availability and broad-spectrum and relatively rapid action (5, 14). The last criterion excluded all but oxidizers because other common active agents are limited as microbicides and/or require hours of contact for sporicidal action.For PAA tests, the recovery of infectious HAV in the absence of any viable spores is somewhat anomalous but not surprising. While we do not believe HAV to be more resistant than bacterial spores, the small size of the virus in the dried inocula likely afforded it significant protection. Compared to HAV, the mycobacterium proved more susceptible to all the disinfectants tested. This highlights a serious weakness in the traditional rankings of disinfectant susceptibility, where mycobacteria are often considered more resistant than nonenveloped viruses (5, 14).In the initial trials with the mixtures, the titer of A. baumannii dropped sharply; using virus pools without antibiotics resolved the issue. The ability of A. baumannii to grow on 7H11 agar and thus interfere with the recovery of M. terrae was addressed by replacing the standard strain of M. terrae with one containing a kanamycin resistance gene (15). Incorporation of enough kanamycin in 7H11 suppressed the growth of A. baumannii while allowing the mycobacterium to grow.Using a mixture of surrogates in QCT not only proved feasible but also highlighted the need to review certain long-held concepts about the relative sensitivities of classes of pathogens to disinfectants. The details reported should allow extension of the work to CL-3 and possibly CL-4 agents to confirm that the results obtained with the carefully chosen surrogates are indeed applicable to various classes of infectious bioagents.  相似文献   

6.
7.
8.
Presented here is the first report describing the detection of potentially diarrheal Vibrio parahaemolyticus strains isolated from cultured bivalves on the Mediterranean coast, providing data on the presence of both tdh- and trh-positive isolates. Potentially diarrheal V. parahaemolyticus strains were isolated from four species of bivalves collected from both bays of the Ebro delta, Spain.Gastroenteritis caused by Vibrio parahaemolyticus has been reported worldwide, though only sporadic cases have been reported in Europe (7, 14). The bacterium can be naturally present in seafood, but pathogenic isolates capable of inducing gastroenteritis in humans are rare in environmental samples (2 to 3%) (15) and are often not detected (10, 19, 20).The virulence of V. parahaemolyticus is based on the presence of a thermostable direct hemolysin (tdh) and/or the thermostable direct hemolysin-related gene (trh) (1, 5). Both are associated with gastrointestinal illnesses (2, 9).Spain is not only the second-largest producer in the world of live bivalve molluscs but also one of the largest consumers of bivalve molluscs, and Catalonia is the second-most important bivalve producer of the Spanish Autonomous Regions. Currently, the cultivation of bivalves in this area is concentrated in the delta region of the Ebro River. The risk of potentially pathogenic Vibrio spp. in products placed on the market is not assessed by existing legislative indices of food safety in the European Union, which emphasizes the need for a better knowledge of the prevalence of diarrheal vibrios in seafood products. The aim of this study was to investigate the distribution and pathogenic potential of V. parahaemolyticus in bivalve species exploited in the bays of the Ebro delta.Thirty animals of each species of Mytilus galloprovincialis, Crassostrea gigas, Ruditapes decussatus, and Ruditapes philippinarum were collected. They were sampled from six sites of the culture area, three in each bay of the Ebro River delta, at the beginning (40°37′112"N, 0°37′092"E [Alfacs]; 40°46′723"N, 0°43′943"E [Fangar]), middle (40°37′125"N, 0°38′570"E [Alfacs]; 40°46′666"N, 0°45′855"E [Fangar]), and end (40°37′309"N, 0°39′934"E [Alfacs]; 40°46′338"N, 0°44′941"E [Fangar]) of the culture polygon. Clams were sampled from only one site per bay as follows: in the Alfacs Bay from a natural bed of R. decussatus (40°37′44"N, 0°38′0"E) and in the Fangar Bay from an aquaculture bed of R. philippinarum (40°47′3"N, 0°43′8"E). In total, 367 samples were analyzed in 2006 (180 oysters, 127 mussels, 30 carpet shell clams, and 30 Manila clams) and 417 samples were analyzed in 2008 (178 oysters, 179 mussels, 30 carpet shell clams, and 30 Manila clams).All animals were individually processed and homogenized, and 1 ml of the homogenate was inoculated into 9 ml of alkaline peptone water (Scharlau, Spain). Following a 6-h incubation at 37°C, one loopful of the contents of each tube of alkaline peptone water was streaked onto CHROMagar vibrio plates (CHROMagar, France) and incubated for 18 h at 37°C. Mauve-purple colonies were purified, and each purified isolate was cryopreserved at −80°C (135 isolates in 2006 and 96 in 2008). From the initial homogenate portion, 100 μl was inoculated onto marine agar (Scharlau, Spain) and onto thiosulfate citrate-bile salts-sucrose agar (Scharlau, Spain) for total heterotrophic marine bacteria counts and total vibrio counts, respectively (Table (Table11).

TABLE 1.

Vibrio parahaemolyticus isolates, serotypes, and origins and total number of vibrios/heterotrophic bacteria contained in the bivalvea
IsolateDate of collectionOrganism and site of originTemp (°C)Salinity (‰)Gene(s)SerotypeBacterial count using indicated medium (CFU ml−1)
TCBS agarMarine agar
I7458 August 2006Mg-F24.537tdhND1.5 × 1041.2 × 104
I79314 August 2006Cg-A2535tdhND9.2 × 1028.5 × 103
I80514 August 2006Cg-A2535tdhO2:KUT7.2 × 1029 × 103
I80614 August 2006Cg-A2535tdh and trhO3:K331.9 × 1034.6 × 103
I80914 August 2006Cg-A2535tdhO2:K288 × 1047.3 × 102
I6784 July 2006Rd-A28.636tdhO2:K283.1 × 1052.5 × 105
I6284 July 2006Rd-A28.636tdhO4:KUT2.9 × 1048.4 × 104
I7758 August 2006Cg-A24.537tdhND4.21 × 1031.1 × 104
I6914 July 2006Rd-A28.636trhO1:K322.2 × 1052.6 × 105
I71227 July 2006Mg-A29.435.5trhO1:KUT8.6 × 1038.4 × 103
I7658 August 2006Cg-F24.537trhO4:K341 × 104Uncountable
I98022 July 2008Cg-A26.733.5tdhO1:K322.7 × 1041.3 × 104
I98122 July 2008Cg-A26.733.5trhO1:KUT1 × 1042.2 × 104
I99322 July 2008Cg-A26.733.5tdhO5:K173 × 1031.1 × 104
I99429 July 2008Mg-A27.737trhO3:KUT3.4 × 1037 × 103
I10315 August 2008Cg-F27.737tdhO5:KUT5.5 × 1043.3 × 104
I10345 August 2008Cg-F27.737tdhO3:KUT8.7 × 1044 × 104
I10405 August 2008Cg-F27.737tdhO3:KUT1.6 × 1043.2 × 104
I10425 August 2008Cg-F27.737tdh and trhND2.8 ×1043 × 104
I10505 August 2008Cg-F27.737tdhO1:KUT4.7 × 1047.3 × 104
I106320 August 2008Mg-F25.936tdhO3:KUT7.9 ×1041.4 × 104
I106520 August 2008Mg-F25.936tdhO2:KUT2.2 × 1031.2 × 104
I106820 August 2008Mg-F25.936tdhO5:KUT2.6 × 1045.2 × 104
I106920 August 2008Mg-F25.936tdhO3:KUT2.4 × 1035.3 × 104
I107320 August 2008Mg-F25.936tdhO5:KUT2.3 × 1037.5 × 103
I107420 August 2008Mg-F25.936tdhO3:KUT7.6 × 1046.9 × 104
I107720 August 2008Mg-F25.936tdhO4:KUT1.7 × 1031.6 × 103
I107920 August 2008Mg-F25.936trhO3:KUT2.5 × 1031.1 × 104
I109220 August 2008Mg-F25.936tdhND1.7 × 1031.6 × 103
I113025 August 2008Rd-A26.435tdhND1.7 × 1043.8 × 104
I114325 August 2008Rd-A26.435tdhND1.1 × 1041.9 × 104
I116525 August 2008Rd-A26.435trhO2:KUT4.4 × 1046.8 × 104
I113325 August 2008Rp-F25.536.5tdhND3.4 × 1044 × 104
I113425 August 2008Rp-F25.536.5tdhND3.9 × 1045.8 × 104
I115825 August 2008Rp-F25.536.5trhO4:KUT6.6 × 1044.7 × 104
I116125 August 2008Rp-F25.536.5trhO3:KUT2.2 × 1046.6 × 104
Open in a separate windowaMg, Mytilus galloprovincialis; Cg, Crassostrea gigas; Rd, Ruditapes decussatus; Rp, R. phillipinarum; A, Alfacs; F, Fangar; ND, not determined; TCBS, thiosulfate citrate-bile salts-sucrose.Total DNA was extracted from each purified isolate using the Wizard genomic DNA purification kit (Promega), following the instructions of the manufacturer. A one-step PCR analysis was performed to identify/confirm which isolates were tl positive (species marker for V. parahaemolyticus). Further detection of the tdh or trh gene was carried out on all positive tl strains. All PCR analyses were carried out using the primers described by Bej et al. (2) with the following amplification conditions on the thermocycler (Eppendorf Mastercycler Personal): an initial denaturation at 95°C for 8 min, followed by 40 cycles of a 1-min denaturation at 94°C, annealing at 55°C for 1 min, elongation at 72° for 1 min, and a final extension of 10 min at 72°C. Positive and negative controls were included in all reaction mixtures: two positive controls, tl and tdh CAIM 1400 and trh CAIM 1772 (Collection of Aquatic Important Microorganisms [http://www.ciad.mx/caim/CAIM.html]), and negative control DNA-free molecular grade water (Sigma-Aldrich, Spain). Expected amplicons were visualized in 2% agarose gels stained with ethidium bromide.Fifty-eight isolates contained the gene tl in 2006 and 96 in 2008, which confirmed their identity as V. parahaemolyticus. In 2006, the distribution of the 58 isolates was as follows: 7 from 127 mussels, 34 from 180 oysters, and 17 from 30 R. decussatus clams. No tl-positive isolates were found in R. philippinarum. PCR analysis of the tl-positive isolates for the presence of the tdh or trh gene indicated that eight isolates contained the tdh gene and four contained the trh gene. In 2008, the source of the confirmed V. parahaemolyticus isolates was as follows: 31 from 88 oysters, 44 from 89 mussels, 9 from 30 R. decussatus clams, and 12 from 30 R. philippinarum clams. Of these, 17 were found to contain the tdh gene and 7 contained the trh gene. Two isolates (I806 and I1042) contained both toxigenic genes, tdh and trh.Putative tdh- and trh-positive PCR products were purified using the QIAquick PCR purification kit (Qiagen) following the manufacturer''s instructions and were sequenced bidirectionally by Macrogen Inc. Sequences were aligned using BioEdit (8) and analyzed using BLAST (National Center for Biotechnology Information). None of the toxigenic isolates was found positive by PCR analysis for the presence of open reading frame 8 of the phage 237 (16), a marker for the pandemic strain O3:K6.The isolates were fingerprinted by repetitive extragenic palindromic PCR (rep-PCR) as described previously (3), and the resulting electrophoretic band patterns were analyzed with the GelCompar II software (v4.5; Applied Maths). The similarity matrix was calculated with the Jaccard coefficient with a band position tolerance of 0.8%, and the dendrogram was constructed with the Ward algorithm. A high level of genomic diversity was found among the 32 toxigenic isolates characterized by rep-PCR. Three clonal groups were identified (those having identical rep-PCR band patterns) (Fig. 1a to c).Open in a separate windowFIG. 1.rep-PCR dendrogram of toxigenic isolates of V. parahaemolyticus isolated in the Ebro delta. Letters denote clonal groups of isolates.In vitro antibiotic susceptibility tests were performed using the diffusion disc test following a previously described protocol (18). The antibiotics used were gentamicin (10 μg), oxolinic acid (10 μg), amoxicillin (25 μg), polymyxin B (300 UI), vancomycin (30 μg), trimethoprim sulfamethoxazole (1.25/23.75 μg), nitrofurantoin (300 μg), doxycyclin (30 μg), ceftazidime (30 μg), streptomycin (10 μg), neomycin (30 UI), penicillin (6 μg), flumequine (30 μg), tetracycline (30 μg), ampicillin (10 μg), kanamycin (30 μg), ciprofloxacin (5 μg), and sulfonamide (300 μg). All tests were performed in duplicate. A Student t test for two samples with unequal variance was performed to compare the sensitivity of all 2006 isolates against the sensitivity of 2008 isolates for each antibiotic (Microsoft Office Excel 97-2003). Antibiogram results revealed a lower susceptibility in 2008 than in 2006, indicating a possible shift in overall susceptibility. Results from the t test indicated that significantly lower susceptibility in 2008 was detected (P ≤ 0.05; n = 36) for the following antibiotics: vancomycin, polymyxin B, ampicillin, amoxicillin, gentamicin, neomycin, trimethoprim sulfamethoxazole, nitrofurantoin, doxycyclin, ceftazidime, tetracycline, flumequine, and ciprofloxacin.The serological types for 27 strains were determined by the agglutination method using commercially available V. parahaemolyticus antisera (Denka Seiken Ltd.; Cosmos Biomedical Ltd, United Kingdom) following the manufacturer''s instructions. Potentially toxigenic V. parahaemolyticus isolates collected in 2006 were serologically heterogeneous (8 out of the 11 isolates) (Table (Table1).1). In isolates collected in 2008, results were more homogenous, with seven serotypes found among 19 isolates analyzed. The O3:K6 serotype was not detected in any of the strains analyzed, in agreement with the open reading frame 8 PCR results.The present study is the first to report the detection of potentially diarrheal V. parahaemolyticus strains isolated from cultured bivalves on Spanish Mediterranean coasts, providing data on the presence of both tdh- and trh-positive isolates. V. parahaemolyticus has previously been detected in several European countries (4, 13, 21, 22). A recent study carried out in Spain detected tdh-positive V. parahaemolyticus strains from patients who had consumed fresh oysters in a market in Galicia on the Atlantic coast of Spain (12) and potentially pathogenic V. parahaemolyticus strains have also been reported in France (17). These studies indicate that the risk of infections caused by V. parahaemolyticus in Europe is low compared to that in America or Asia (15). However, this risk could have been underestimated, since V. parahaemolyticus is not included in the current European surveillance programs, such as the European Network for Epidemiological Surveillance and Control of Communicable Diseases.Toxigenic V. parahaemolyticus strains detected in this study were genomically and serologically heterogeneous. The pandemic serotype O3:K6 was not detected, and although attempts to isolate O3:K6 from the environment and from seafood have not always been successful in previous studies reviewed by Nair and coauthors (15), this finding seems to be in agreement with the fact that no outbreak of diarrhea was observed in the area. Interestingly, isolates I806 and I1042 have been found positive for both tdh and trh in PCR tests. The coexistence of tdh and trh genes has already been reported in isolates from Japan, the United States, and Mexico (3, 6, 11, 19, 23). To our knowledge, no occurrence of an environmental isolate positive for both tdh and trh had previously been reported in Europe. All isolates tested were slightly different in their antibiotic resistance profiles. Typically, a high level of resistance could be determined. The detection of tdh- and/or trh-positive V. parahaemolyticus strains for the first time on the Mediterranean coast emphasizes the need to monitor for the presence of potentially diarrheal vibrios and bacterial gastroenteritis, and these data should be taken into consideration to revise the European legislation on the requirements for shellfish harvested for consumption in order to include the surveillance of these pathogens in Europe.  相似文献   

9.
10.
Twelve cluster groups of Escherichia coli O26 isolates found in three cattle farms were monitored in space and time. Cluster analysis suggests that only some O26:H11 strains had the potential for long-term persistence in hosts and farms. As judged by their virulence markers, bovine enterohemorrhagic O26:H11 isolates may represent a considerable risk for human infection.Shiga toxin (Stx)-producing Escherichia coli (STEC) strains comprise a group of zoonotic enteric pathogens (42). In humans, infections with some STEC serotypes result in hemorrhagic or nonhemorrhagic diarrhea, which can be complicated by hemolytic-uremic syndrome (HUS) (49). These STEC strains are also designated “enterohemorrhagic E. coli” (EHEC). Consequently, EHEC strains represent a subgroup of STEC with a high pathogenic potential for humans. Strains of the E. coli serogroup O26 were originally classified as enteropathogenic E. coli due to their association with outbreaks of infantile diarrhea in the 1940s. In 1977, Konowalchuk et al. (37) recognized that these bacteria produced Stx, and 10 years later, the Stx-producing E. coli O26:H11/H− strains were classified as EHEC. EHEC O26 strains constitute the most common non-O157 EHEC group associated with diarrhea and HUS in Europe (12, 21, 23, 24, 26, 27, 55, 60). Reports on an association between EHEC O26 and HUS or diarrhea from North America including the United States (15, 30, 33), South America (51, 57), Australia (22), and Asia (31, 32) provide further evidence for the worldwide spread of these organisms. Studies in Germany and Austria (26, 27) on sporadic HUS cases between 1996 and 2003 found that EHEC O26 accounted for 14% of all EHEC strains and for ∼40% of non-O157 EHEC strains obtained from these patients. A proportion of 11% EHEC O26 strains was detected in a case-control study in Germany (59) between 2001 and 2003. In the age group <3 years, the number of EHEC O26 cases was nearly equal to that of EHEC O157 cases, although the incidence of EHEC O26-associated disease is probably underestimated because of diagnostic limitations in comparison to the diagnosis of O157:H7/H− (18, 34). Moreover, EHEC O26 has spread globally (35). Beutin (6) described EHEC O26:H11/H−, among O103:H2, O111:H, O145:H28/H−, and O157:H7/H−, as the well-known pathogenic “gang of five,” and Bettelheim (5) warned that we ignore the non-O157 STEC strains at our peril.EHEC O26 strains produce Stx1, Stx2, or both (15, 63). Moreover, these strains contain the intimin-encoding eae gene (11, 63), a characteristic feature of EHEC (44). In addition, EHEC strains possess other markers associated with virulence, such as a large plasmid that carries further potential virulence genes, e.g., genes coding for EHEC hemolysin (EHEC-hlyA), a catalase-peroxidase (katP), and an extracellular serine protease (espP) (17, 52). The efa1 (E. coli factor for adherence 1) gene was identified as an intestinal colonization factor in EHEC (43). EHEC O26 represents a highly dynamic group of organisms that rapidly generate new pathogenic clones (7, 8, 63).Ruminants, especially cattle, are considered the primary reservoir for human infections with EHEC. Therefore, the aim of this study was the molecular characterization of bovine E. coli field isolates of serogroup O26 using a panel of typical virulence markers. The epidemiological situation in the beef herds from which the isolates were obtained and the spatial and temporal behavior of the clonal distribution of E. coli serogroup O26 were analyzed during the observation period. The potential risk of the isolates inducing disease in humans was assessed.In our study, 56 bovine E. coli O26:H11 isolates and one bovine O26:H32 isolate were analyzed for EHEC virulence-associated factors. The isolates had been obtained from three different beef farms during a long-term study. They were detected in eight different cattle in farm A over a period of 15 months (detected on 10 sampling days), in 3 different animals in farm C over a period of 8 months (detected on 3 sampling days), and in one cow on one sampling day in farm D (Table (Table1)1) (28).

TABLE 1.

Typing of E. coli O26 isolates
Sampling day, source, and isolateSerotypeVirulence profile by:
fliC PCR-RFLPstx1 genestx2 geneStx1 (toxin)Stx2 (toxin)Subtype(s)
efa1 genebEHEC-hlyA genekatP geneespP genePlasmid size(s) in kbCluster
stx1/stx2eaetirespAespB
Day 15
    Animal 6 (farm A)
        WH-01/06/002-1O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/06/002-2O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/06/002-3O26:H11H11++stx1ββββ+/++++110, 127
    Animal 8 (farm A)
        WH-01/08/002-2O26:H11H11++stx1ββββ+/++++110, 127
    Animal 26 (farm A)
        WH-01/26/001-2O26:H11H11++stx1ββββ+/++++130, 127
        WH-01/26/001-5O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/26/001-6O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/26/001-7O26:H11H11++stx1ββββ+/−+++110, 127
Day 29
    Animal 2 (farm A)
        WH-01/02/003-1O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-2O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-5O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-6O26:H11H11++stx1ββββ+/+++110, 126
        WH-01/02/003-7O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-8O26:H11H11++stx1ββββ−/++++110, 126
        WH-01/02/003-9O26:H11H11++stx1ββββ+/++++1106
        WH-01/02/003-10O26:H11H11++stx1ββββ+/++++1106
    Animal 26 (farm A)
        WH-01/26/002-2O26:H11H11++stx1ββββ+/++++130, 125
        WH-01/26/002-5O26:H11H11++stx1ββββ+/++++130, 125
        WH-01/26/002-8O26:H11H11++stx1ββββ+/++++130, 125
        WH-01/26/002-9O26:H11H11++stx1ββββ+/++110, 125
        WH-01/26/002-10O26:H11H11++stx1ββββ+/++++130, 125
Day 64
    Animal 20 (farm A)
        WH-01/20/005-3O26:H11H11++stx1ββββ+/+130, 2.52
Day 78
    Animal 29 (farm A)
        WH-01/29/002-1O26:H11H11++stx1ββββ+/−+130, 12, 2.54
        WH-01/29/002-2O26:H11H11++stx1ββββ+/++++130, 12, 2.54
        WH-01/29/002-3O26:H11H11++stx1ββββ+/++++130, 12, 2.54
        WH-01/29/002-4O26:H11H11++stx1ββββ+/++++130, 12, 2.54
        WH-01/29/002-5O26:H11H11++stx1ββββ+/++130, 12, 2.54
Day 106
    Animal 27 (farm A)
        WH-01/27/005-2O26:H11H11++stx1ββββ+/−+++145, 110, 123
        WH-01/27/005-5O26:H11H11++stx1ββββ+/++++130, 12, 2.55
        WH-01/27/005-6O26:H11H11++stx1ββββ+/+130, 12, 2.55
Day 113
    Animal 7 (farm C)
        WH-04/07/001-2O26:H11H11++++stx1/stx2ββββ+/+++55, 35, 2.511
        WH-04/07/001-4O26:H11H11++++stx1/stx2ββββ+/++++5512
        WH-04/07/001-6O26:H11H11++++stx1/stx2ββββ+/++++5512
Day 170
    Animal 22 (farm C)
        WH-04/22/001-1O26:H11H11++stx1ββββ+/++++110, 12, 6.312
        WH-04/22/001-4O26:H11H11++stx1ββββ+/++++110, 12, 6.312
        WH-04/22/001-5O26:H11H11++stx1ββββ+/++++110, 12, 6.312
Day 176
    Animal 14 (farm D)
        WH-03/14/004-8O26:H11H11++stx1ββββ+/+++11010
Day 218
    Animal 27 (farm A)
        WH-01/27/009-1O26:H11H11++++stx1/stx2ββββ+/++++110, 129
        WH-01/27/009-2O26:H11H11++++stx1/stx2ββββ+/++++110, 129
        WH-01/27/009-3O26:H11H11++++stx1/stx2ββββ+/++++110, 128
        WH-01/27/009-8O26:H11H11++++stx1/stx2ββββ+/++110, 128
        WH-01/27/009-9O26:H11H11++++stx1/stx2ββββ+/++++110, 129
Day 309
    Animal 29 (farm A)
        WH-01/29/010-1O26:H11H11++stx1ββββ+/++++110, 35, 124
        WH-01/29/010-2O26:H11H11++stx1ββββ+/++130, 55, 358
        WH-01/29/010-3O26:H11H11++stx1ββββ+/++++130, 35, 128
Day 365
    Animal 8 (farm C)
        WH-04/08/008-6O26:H11H11++stx1ββββ+/++++110, 5512
Day 379
    Animal 9 (farm A)
        WH-01/09/016-2O26:H32H32++stx1/stx2−/−145, 130, 1.81
    Animal 27 (farm A)
        WH-01/27/014-3O26:H11H11++stx1ββββ+/++++110, 129
        WH-01/27/014-4O26:H11H11++stx1ββββ+/++++110, 129
        WH-01/27/014-5O26:H11H11++stx1ββββ+/++++110, 128
Day 407
    Animal 29 (farm A)
        WH-01/29/013-4O26:H11H11++stx1ββββ+/++++110, 12, 2.58
        WH-01/29/013-7O26:H11H11++stx1ββββ+/++++110, 12, 2.58
Day 478
    Animal 27 (farm A)
        WH-01/27/017-1O26:H11H11++++stx1/stx2ββββ+/++++110, 128
        WH-01/27/017-5O26:H11H11++++stx1/stx2ββββ+/++++110, 128
        WH-01/27/017-6O26:H11H11++++stx1/stx2ββββ+/++++1108
        WH-01/27/017-7O26:H11H11++++stx1/stx2ββββ+/++++1108
        WH-01/27/017-10O26:H11H11+++stx1ββββ+/++++130, 12, 2.58
Open in a separate windowastx1/stx2, gene stx1 or stx2.befa1 was detected by two hybridizations (with lifA1-lifA2 and lifA3-lifA4 probes). +/+, complete gene; +/− or −/+, incomplete gene; −/−, efa1 negative.The serotyping of the O26 isolates was confirmed by the results of the fliC PCR-restriction fragment length polymorphism (RFLP) analysis performed according to Fields et al. (25), with slight modifications described by Zhang et al. (62). All O26:H11 isolates showed the H11 pattern described by Zhang et al. (62). In contrast, the O26:H32 isolate demonstrated a different fliC RFLP pattern that was identical to the H32 pattern described by the same authors. It has been demonstrated that EHEC O26:H11 strains belong to at least four different sequence types (STs) in the common clone complex 29 (39). In the multilocus sequence typing analysis for E. coli (61), the tested five EHEC O26:H11 isolates (WH-01/02/003-1, WH-01/20/005-3, WH-01/27/009-9, WH-03/14/004-8, and WH-04/22/001-1) of different farms and clusters were characterized as two sequence types (ST 21 and ST 396). The isolates from farms A and C belong to ST 21, the most frequent ST of EHEC O26:H11 isolates found in humans and animals (39), but the single isolate from farm D was characterized as ST 396.Typing and subtyping of genes (stx1 and/or stx2, eae, tir, espA, espB, EHEC-hlyA, katP, and espP) associated with EHEC were performed with LightCycler fluorescence PCR (48) and different block-cycler PCRs. To identify the subtypes of the stx2 genes and of the locus of enterocyte effacement-encoding genes eae, tir, espA, and espB, the PCR products were digested by different restriction endonucleases (19, 26, 46). The complete pattern of virulence markers was detected in most bovine isolates examined in our study. An stx1 gene was present in all O26 isolates. In addition, an stx2 gene was found in nine O26:H11 isolates in farm A and in three isolates of the same type in farm C, as well as in the O26:H32 isolate. Both Stx1 and Stx2 were closely related to families of Stx1 and Stx2 variants or alleles. EHEC isolates with stx2 genes are significantly more often associated with HUS and other severe disease manifestations than isolates with an stx1 gene, which are more frequently associated with uncomplicated diarrhea and healthy individuals (13). In contrast to STEC strains harboring stx2 gene variants, however, STEC strains of the stx2 genotype were statistically significantly associated with HUS (26). The stx2 genotype was found in all O26 isolates with an stx2 gene, while the GK3/GK4 amplification products after digestion with HaeIII and FokI restriction enzymes showed the typical pattern for this genotype described by Friedrich et al. (26). The nucleotide sequences of the A and B subunits of the stx2 gene of the selected bovine O26:H11 isolate WH-01/27/017-1 (GenBank accession no. EU700491) were identical to the stx2 genes of different sorbitol-fermenting EHEC O157:H− strains associated with human HUS cases and other EHEC infections in Germany (10) and 99.3% identical in their DNA sequences to the stx2 gene of the EHEC type strain EDL933, a typical O157:H7 isolate from an HUS patient. A characteristic stx1 genotype was present in all O26 isolates. The nucleotide sequences of the A and B subunits of the stx1 gene of the tested bovine O26:H11 isolate WH-01/27/017-1 (GenBank accession no. EU700490) were nearly identical to those of the stx1 genes of the EHEC O26:H11 reference type strains H19 and DEC10B, which had been associated with human disease outbreaks in Canada and Australia. Nucleotide exchanges typical for stx1c and stx1d subtypes as described by Kuczius et al. (38) were not found. All bovine O26:H11 strains produced an Stx1 with high cytotoxicity for Vero cells tested by Stx enzyme-linked immunosorbent assay and Vero cell neutralization assay (53). The Stx2 cytotoxicity for Vero cells was also very high in the O26:H11 isolates.Not only factors influencing the basic and inducible Stx production are important in STEC pathogenesis. It has been suggested that the eae and EHEC-hlyA genes are likely contributors to STEC pathogenicity (2, 3, 13, 50). Ritchie et al. (50) found both genes in all analyzed HUS-associated STEC isolates. In all O26:H11 isolates we obtained, stx genes were present in combination with eae genes. Only the O26:H32 isolate lacked an eae gene. To date, 10 distinct variants of eae have been described (1, 19, 36, 45, 47). Some serotypes were closely associated with a particular intimin variant: the O157 serogroup was linked to γ-eae, the O26 serogroup to β-eae, and the O103 serogroup to ɛ-eae (4, 19, 20, 58). Our study confirms these associations. All bovine O26:H11 isolates were also typed as members of the β-eae subgroup. A translocated intimin receptor gene (tir gene) and the type III secreted proteins encoded by the espA and espB genes were found in all 56 O26:H11 isolates but not in the O26:H32 isolate. These other tested locus of enterocyte effacement-associated genes belonged to the β-subgroups. These results are in accord with the results of China et al. (19), who detected the pathotypes β-eae, β-tir, β-espA, and β-espB in all investigated human O26 strains. Like the eae gene, the EHEC-hlyA gene was found in association with severe clinical disease in humans (52). Aldick et al. (2) showed that EHEC hemolysin is toxic (cytolytic) to human microvascular endothelial cells and may thus contribute to the pathogenesis of HUS. In our study, the EHEC-hlyA gene was detected in 50 of the 56 bovine E. coli O26:H11 isolates which harbored virulence-associated plasmids of different sizes (Table (Table1).1). The presence of virulence-associated plasmids corresponded to the occurrence of additional virulence markers such as the espP and katP genes (17). The katP gene and the espP gene were detected in 49 and 50 of the 56 O26:H11 isolates, respectively. The espP gene was missing in six of the seven bovine O26:H11 isolates in which the katP genes were also absent. Both genes were not found in the O26:H32 isolate (Table (Table1).1). Although we found large plasmids of the same size in O26:H11 isolates, they lacked one or more of the plasmid-associated virulence factors (Table (Table1).1). Two DNA probes were used to detect the efa1 genes by colony hybridization. (DNA probes were labeled with digoxigenin [DIG] with lifA1-lifA2 and lifA3-lifA4 primers [14] using the PCR DIG probe synthesis kit [Roche Diagnostics, Mannheim, Germany]; DIG Easy Hyb solution [Roche] was used for prehybridization and hybridization.) Positive results with both DNA probes were obtained for 52 of 56 E. coli O26:H11 isolates. A positive signal was only found in three isolates with the lifA1-lifA2 DNA probe and in one isolate with the lifA3-lifA4 probe. An efa1 gene was not detected in the O26:H32 isolate (Table (Table11).We also analyzed the spatial and temporal behavior of the O26:H11/H32 isolates in the beef herds by cluster analysis (conducted in PAUP* for Windows version 4.0, 2008 [http://paup.csit.fsu.edu/about.html]). This was performed with distance matrices using the neighbor-joining algorithm, an agglomerative cluster method which generates a phylogenetic tree. The distance matrices were calculated by pairwise comparisons of the fragmentation patterns produced by genomic typing through pulsed-field gel electrophoresis analysis with four restriction endonucleases (XbaI, NotI, BlnI, and SpeI) and the presence or absence of potential virulence markers (Fig. (Fig.11 and Table Table1).1). To this end, the total character difference was used, which counts the pairwise differences between two given patterns. During a monitoring program of 3 years in four cattle farms (29), different O26:H11 cluster groups and one O26:H32 isolate were detected in three different farms. The genetic distance of the O26:H32 isolate was very high relative to the O26:H11 isolates. Therefore, the O26:H32 isolate was outgrouped. The O26:H11 isolates of each farm represented independent cluster groups. The single isolate from farm D fitted better to the isolates from farm C than to those from farm A. This finding is in accord with the geographical distance between the farms. The fact that the farms were located in neighboring villages may suggest that direct or indirect connections between the farms were possible (e.g., by person contacts or animal trade). However, the isolates from farm C and farm D belonged to different sequence types (ST 21 and ST 396), which may argue against a direct connection. Interestingly, O26:H11 isolates with and without stx2 genes were detected in the same clusters. This phenomenon was observed in both farm A and farm C. In farm A, the isolates with additional stx2 genes were found in animal 27 and were grouped in clusters 8 and 9 (day 218). An stx2 gene was repeatedly found (four isolates) in the same animal (animal 27). The isolates grouped in cluster 8 on a later day of sampling (day 478). All other O26:H11 isolates grouped in the same clusters and obtained from the same animals (27 and 29) on different sampling days lacked an stx2 gene. Also, the isolates obtained from animal 27 on previous sampling days, which grouped in clusters 3 and 5, exhibited no stx2 genes. In farm C, the three isolates with additional stx2 genes obtained from animal 7 grouped in clusters 11 and 12. An stx2 gene was absent from all other O26:H11 isolates grouped in the same cluster 12 on later sampling days, and no other isolates of cluster 11 were found later on. However, we detected members of many clusters over relatively long periods (clusters 5, 8, and 9 in farm A and cluster 12 in farm C), but members of other clusters were only found on single occasions. This patchy temporal pattern is apparently not a unique property of O26:H11, as we found similar results for cluster groups of other EHEC serotypes of bovine origin (28). The isolates grouped in the dominant cluster 8 were found on 5 of 9 sampling days over a period of 10 months. In contrast, we found the members of clusters 4, 5, 9, and 12 only on two nonconsecutive sampling days. The period during which isolates of these groups were not detected was particularly long for cluster 4 (231 days). We also observed the coexistence of different clusters over long periods in the same farm and in the same cattle (clusters 8 and 9), while one of the clusters dominated. Transmission of clusters between cattle was also observed. These results suggest that some of the EHEC O26:H11 strains had the potential for a longer persistence in the host population, while others had not. The reasons for this difference are not yet clear. Perhaps the incomplete efa1 gene found in isolates of clusters which were only detected once might explain why some strains disappeared rapidly. Efa1 has been discussed as a potential E. coli colonization factor for the bovine intestine used by non-O157 STEC, including O26 (54, 56). The O165:H25 cluster detected during a longer period in farm B may have disappeared after it had lost its efa1 gene (28). The precise biological activity of Efa1 in EHEC O26 is not yet known, but it has been demonstrated that the molecule is a non-Stx virulence determinant which can increase the virulence of EHEC O26 in humans (8).Open in a separate windowFIG. 1.Neighbor-joining tree of bovine E. coli O26:H11/H32 strains based on the restriction pattern obtained after digestion with XbaI, NotI, BlnI, and SpeI.We distinguished 12 different clusters, but complete genetic identity was only found in two isolates. The variations in the O26:H11 clusters may be due to increasing competition between the bacterial populations of the various subtypes in the bovine intestine or to potential interactions between EHEC O26:H11 and the host.The ephemeral occurrence of additional stx2 genes in different clusters and farms may be the result of recombination events due to horizontal gene transfer (16). The loss of stx genes may occur rapidly in the course of an infection, but the reincorporation by induction of an stx-carrying bacteriophage into the O26:H11 strains is possible at any time (9, 40). Nevertheless, an additional stx2 gene may increase the dangerousness of the respective EHEC O26:H11 strains. While all patients involved in an outbreak caused by an EHEC O26:H11 strain harboring the gene encoding Stx2 developed HUS (41), the persons affected by another outbreak caused by an EHEC O26:H11 strain that produced exclusively Stx1 had only uncomplicated diarrhea (60).In conclusion, our results showed that bovine O26:H11 isolates can carry virulence factors of EHEC that are strongly associated with EHEC-related disease in humans, particularly with severe clinical manifestations such as hemorrhagic colitis and HUS. Therefore, strains of bovine origin may represent a considerable risk for human infection. Moreover, some clusters of EHEC O26:H11 persisted in cattle and farms over longer periods, which may increase the risk of transmission to other animals and humans even further.  相似文献   

11.
Melioidosis has been considered an emerging disease in Brazil since the first cases were reported to occur in the northeast region. This study investigated two municipalities in Ceará state where melioidosis cases have been confirmed to occur. Burkholderia pseudomallei was isolated in 26 (4.3%) of 600 samples in the dry and rainy seasons.Melioidosis is an endemic disease in Southeast Asia and northern Australia (2, 4) and also occurs sporadically in other parts of the world (3, 7). Human melioidosis was reported to occur in Brazil only in 2003, when a family outbreak afflicted four sisters in the rural part of the municipality of Tejuçuoca, Ceará state (14). After this episode, there was one reported case of melioidosis in 2004 in the rural area of Banabuiú, Ceará (14). And in 2005, a case of melioidosis associated with near drowning after a car accident was confirmed to occur in Aracoiaba, Ceará (11).The goal of this study was to investigate the Tejuçuoca and Banabuiú municipalities, where human cases of melioidosis have been confirmed to occur, and to gain a better understanding of the ecology of Burkholderia pseudomallei in this region.We chose as central points of the study the residences and surrounding areas of the melioidosis patients in the rural areas of Banabuiú (5°18′35″S, 38°55′14″W) and Tejuçuoca (03°59′20″S, 39°34′50′W) (Fig. (Fig.1).1). There are two well-defined seasons in each of these locations: one rainy (running from January to May) and one dry (from June to December). A total of 600 samples were collected at five sites in Tejuçuoca (T1, T2, T3, T4, and T5) and five in Banabuiú (B1, B2, B3, B4, and B5), distributed as follows (Fig. (Fig.2):2): backyards (B1 and T1), places shaded by trees (B2 and T2), water courses (B3 and T3), wet places (B4 and T4), and stock breeding areas (B5 and T5).Open in a separate windowFIG. 1.Municipalities of Banabuiú (5°18′35″S, 38°55′14″W) and Tejuçuoca (03°59′20″S, 39°34′50″W).Open in a separate windowFIG. 2.Soil sampling sites in Banabuiú and Tejuçuoca.Once a month for 12 months (a complete dry/rainy cycle), five samples were gathered at five different depths: at the surface and at 10, 20, 30 and 40 cm (Table (Table1).1). The samples were gathered according to the method used by Inglis et al. (9). Additionally, the sample processing and B. pseudomallei identification were carried out as previously reported (1, 8, 9).

TABLE 1.

Distribution of samples with isolates by site and soil depth
Sitesa and depth (cm)No. of B. pseudomallei isolates in samples from:
Banabuiú (n = 300)Tejuçuoca (n = 300)Total (n = 600)
B1/T13
    Surface2
    10
    201
    30
    40
B2/T21
    Surface1
    10
    20
    30
    40
B3/T315
    Surface2
    102
    204
    303
    404
B4/T45
    Surface
    101
    201
    3011
    401
B5/T52
    Surface
    10
    20
    302
    40
Total62026
Open in a separate windowaSites designated with B are in Banabuiú, and sites designated with T are in Tejuçuoca. See the text for details.The data on weather and soil composition were obtained from specialized government institutions, such as FUNCEME, IPECE, and EMBRAPA. The average annual temperature in both municipalities is between 26 and 28°C. In 2007, the annual rainfall in Tejuçuoca was 496.8 mm, and that in Banabuiú was 766.8 mm. There are a range of soil types in both Tejuçuoca and Banabuiú: noncalcic brown, sodic planossolic, red-yellow podzolic, and litholic. In Banabuiú, there are also alluvial and cambisol soils. The characteristic vegetation in both municipalities is caatinga (scrublands).There were isolates of B. pseudomallei in 26 (4.3%) of the 600 samples collected. The bacterium was isolated at a rate (3%) similar to that previously reported (9). The bacterium isolation occurred in both the dry (53.8%) and the rainy (46.2%) seasons. Tejuçuoca represented 76.9% (20/26) of the strains isolated. Four sites in Tejuçuoca (T1, T3, T4, and T5) and three in Banabuiú (B1, B2, and B4) presented isolates of the bacterium (Table (Table1).1). The isolation of the B. pseudomallei strains varied from the surface down to 40 cm. However, 17 of the 26 positive samples (65.3%) were found at depths between 20 and 40 cm (Table (Table1).1). Only two isolates were found at the surface during the dry season.A study in Vietnam (13) and one in Australia (9) reported the presence of B. pseudomallei near the houses of melioidosis patients. In our study, the same thing happened. Site T3 (15/26; 57.6%) was located 290 m from the patient''s house, as reported by the Rolim group (14).B. pseudomallei was isolated from a sheep paddock in Australia, where animals sought shelter below mango and fig trees (17). In our study, the bacterium was isolated at site T5, a goat corral alongside the house where the outbreak occurred in Tejuçuoca. Four sites in places shaded by trees yielded positive samples (30.7%) in both Tejuçuoca (palm trees) and Banabuiú (mango trees). Additionally, B. pseudomallei was isolated on three occasions from a cornfield (site 4B) located alongside the house of the melioidosis patient in Banabuiú.In the main areas of endemicity, the disease is more prevalent in the rainy season (4, 5, 16). The outbreak in Tejuçuoca was related to rainfall (14). Besides the association of cases of the disease with rainfall itself, the isolation of B. pseudomallei in soil and water was also demonstrated during the dry season (12, 15). An Australian study isolated strains from soil and water during the dry and rainy seasons (17). A Thai study also reported B. pseudomallei in the dry season (18). In our study, the isolation of B. pseudomallei took place either at the end of the wet season or in the dry months. Fourteen of the positive samples (53.8%) were collected during the dry season, albeit near a river or reservoir (sites T3 and B4).Physical, biological, and chemical soil features appear to influence the survival of B. pseudomallei (6, 10). In the present study, the soil was classified as litholic with sandy or clayey textures. It is susceptible to erosion, and when there is a lack of water, it is subject to salinization. During the dry season, the clay layer becomes dried, cracked, and very hard. During the rainy season, it becomes soggy and sticky. The isolation of B. pseudomallei in the dry season is possibly related to the capacity for adaptation of this soil, since the extreme conditions of lithosols do not prevent the bacterial growth and survival.It has been shown that B. pseudomallei is more often isolated at depths between 25 and 45 cm (17). In our study, 65.3% of the positive samples were taken at depths between 20 and 40 cm. Moreover, of these 17 samples, 10 (58.8%) were collected during the dry months. Also, unlike in other regions, two positive samples were taken from the surface in the period without rainfall.The rainfall in Tejuçuoca and Banabuiú is generally low, and temperatures do not vary significantly during the year. Therefore, the isolation of B. pseudomallei in these places occurs outside the rainfall, temperature, and moisture conditions observed in other regions of endemicity. Our data thus suggest that peculiar environmental features, such as soil composition, might favor the multiplication of B. pseudomallei in northeast Brazil.  相似文献   

12.
13.
Short-chain alcohol dehydrogenase, encoded by the gene Tsib_0319 from the hyperthermophilic archaeon Thermococcus sibiricus, was expressed in Escherichia coli, purified and characterized as an NADPH-dependent enantioselective oxidoreductase with broad substrate specificity. The enzyme exhibits extremely high thermophilicity, thermostability, and tolerance to organic solvents and salts.Alcohol dehydrogenases (ADHs; EC 1.1.1.1.) catalyze the interconversion of alcohols to their corresponding aldehydes or ketones by using different redox-mediating cofactors. NAD(P)-dependent ADHs, due to their broad substrate specificity and enantioselectivity, have attracted particular attention as catalysts in industrial processes (5). However, mesophilic ADHs are unstable at high temperatures, sensitive to organic solvents, and often lose activity during immobilization. In this relation, there is a considerable interest in ADHs from extremophilic microorganisms; among them, Archaea are of great interest. The representatives of all groups of NAD(P)-dependent ADHs have been detected in genomes of Archaea (11, 12); however, only a few enzymes have been characterized, and the great majority of them belong to medium-chain (3, 4, 14, 16, 19) or long-chain iron-activated ADHs (1, 8, 9). Up to now, a single short-chain archaeal ADH from Pyrococcus furiosus (10, 18) and only one archaeal aldo-keto reductase also from P. furiosus (11) have been characterized.Thermococcus sibiricus is a hyperthermophilic anaerobic archaeon isolated from a high-temperature oil reservoir capable of growth on complex organic substrates (15). The complete genome sequence of T. sibiricus has been recently determined and annotated (13). Several ADHs are encoded by the T. sibiricus genome, including three short-chain ADHs (Tsib_0319, Tsib_0703, and Tsib_1998) (13). In this report, we describe the cloning and expression of the Tsib_0319 gene from T. sibiricus and the purification and the biochemical characterization of its product, the thermostable short-chain ADH (TsAdh319).The Tsib_0319 gene encodes a protein with a size of 234 amino acids and the calculated molecular mass of 26.2 kDa. TsAdh319 has an 85% degree of sequence identity with short-chain ADH from P. furiosus (AdhA; PF_0074) (18). Besides AdhA, close homologs of TsAdh319 were found among different bacterial ADHs, but not archaeal ADHs. The gene flanked by the XhoI and BamHI sites was PCR amplified using two primers (sense primer, 5′-GTTCTCGAGATGAAGGTTGCTGTGATAACAGGG-3′, and antisense primer, 5′-GCTGGATCCTCAGTATTCTGGTCTCTGGTAGACGG-3′) and cloned into the pET-15b vector. TsAdh319 was overexpressed, with an N-terminal His6 tag in Escherichia coli Rosetta-gami (DE3) and purified to homogeneity by metallochelating chromatography (Hi-Trap chelating HP column; GE Healthcare) followed by gel filtration on Superdex 200 10/300 GL column (GE Healthcare) equilibrated in 50 mM Tris-HCl (pH 7.5) with 200 mM NaCl. The homogeneity and the correspondence to the calculated molecular mass of 28.7 kDa were verified by SDS-PAGE (7). The molecular mass of native TsAdh319 was 56 to 60 kDa, which confirmed the dimeric structure in solution.The standard ADH activity measurement was made spectrophotometrically at the optimal pH by following either the reduction of NADP (in 50 mM Gly-NaOH buffer; pH 10.5) or the oxidation of NADPH (in 0.1 M sodium phosphate buffer; pH 7.5) at 340 nm at 60°C. The enzyme exhibited a strong preference for NADP(H) and broad substrate specificity (Table (Table1).1). The highest oxidation rates were found with pentoses d-arabinose (2.0 U mg−1) and d-xylose (2.46 U mg−1), and the highest reduction rates were found with dimethylglyoxal (5.9 U mg−1) and pyruvaldehyde (2.2 U mg−1). The enzyme did not reduce sugars which were good substrates for the oxidation reaction. The kinetic parameters of TsAdh319 determined for the preferred substrates are shown in Table Table2.2. The enantioselectivity of the enzyme was estimated by measuring the conversion rates of 2-butanol enantiomers. TsAdh319 showed an evident preference, >2-fold, for (S)-2-butanol over (RS)-2-butanol. The enzyme stereoselectivity is confirmed by the preferred oxidation of d-arabinose over l-arabinose (Table (Table1).1). The fact that TsAdh319 is metal independent was supported by the absence of a significant effect of TsAdh319 preincubation with 10 mM Me2+ for 30 min before measuring the activity in the presence of 1 mM Me2+ or EDTA (Table (Table3).3). TsAdh319 also exhibited a halophilic property, so the enzyme activity increased in the presence of NaCl and KCl and the activation was maintained even at concentration of 4 M and 3 M, respectively (Table (Table33).

TABLE 1.

Substrate specificity of TsAdh319
SubstrateaRelative activity (%)
Oxidation reactionb
    Methanol0
    2-Methoxyethanol0
    Ethanol36
    1-Butanol80
    2-Propanol100
    (RS)-(±)-2-Butanol86
    (S)-(+)-2-Butanol196
    2-Pentanol67
    1-Phenylmethanol180
    1.3-Butanediol91
    Ethyleneglycol0
    Glycerol16
    d-Arabinose*200
    l-Arabinose*17
    d-Xylose*246
    d-Ribose*35
    d-Glucose*146
    d-Mannose*48
    d-Galactose*0
    Cellobiose*71
Reduction reactionc
    Pyruvaldehyde100
    Dimethylglyoxal270
    Glyoxylic acid36
    Acetone0
    Cyclopentanone0
    Cyclohexanone4
    3-Methyl-2-pentanone*13
    d-Arabinose*0
    d-Xylose*0
    d-Glucose*0
    Cellobiose*0
Open in a separate windowaSubstrates were present in 250 mM or 50 mM (*) concentrations.bRelative rates, measured under standard conditions, were calculated by defining the activity for 2-propanol as 100%, which corresponds to 1.0 U mg−1. Data are averages from triplicate experiments.cRelative rates, measured under standard conditions, were calculated by defining the activity for pyruvaldehyde as 100%, which corresponds to 2.2 U mg−1. Data are averages from triplicate experiments.

TABLE 2.

Apparent Km and Vmax values for TsAdh319
Coenzyme or substrateApparent Km (mM)Vmax (U mg−1)kcat (s−1)
NADPa0.022 ± 0.0020.94 ± 0.020.45 ± 0.01
NADPHb0.020 ± 0.0033.16 ± 0.111.51 ± 0.05
2-Propanol168 ± 291.10 ± 0.090.53 ± 0.04
d-Xylose54.4 ± 7.41.47 ± 0.090.70 ± 0.04
Pyruvaldehyde17.75 ± 3.384.26 ± 0.402.04 ± 0.19
Open in a separate windowaActivity was measured under standard conditions with 2-propanol. Data are averages from triplicate experiments.bActivity was measured under standard conditions with pyruvaldehyde. Data are averages from triplicate experiments.

TABLE 3.

Effect of various ions and EDTA on TsAdh319a
CompoundConcn (mM)Relative activity (%)
None0100
NaCl400206
600227
4,000230
KCl600147
2,000200
3,000194
MgCl21078
CoCl210105
NiSO410100
ZnSO41079
FeSO41074
EDTA1100
580
Open in a separate windowaThe activity was measured under standard conditions with 2-propanol; relative rates were calculated by defining the activity without salts as 100%, which corresponds to 0.9 U mg−1. Data are averages from duplicate experiments.The most essential distinctions of TsAdh319 are the thermophilicity and high thermostability of the enzyme. The optimum temperature for the 2-propanol oxidation catalyzed by TsAdh319 was not achieved. The initial reaction rate of oxidation increased up to 100°C (Fig. (Fig.1).1). The Arrhenius plot is a straight line, typical of a single rate-limited thermally activated process, but there is no obvious transition point due to the temperature-dependent conformational changes of the protein molecule. The activation energy for the oxidation of 2-propanol was estimated at 84.0 ± 5.8 kJ·mol−1. The thermostability of TsAdh319 was calculated from residual TsAdh319 activity after preincubation of 0.4 mg/ml enzyme solution in 50 mM Tris-HCl buffer (pH 7.5) containing 200 mM NaCl at 70, 80, 90, or 100°C. The preincubation at 70°C or 80°C for 1.5 h did not cause a decrease in the TsAdh319 activity, but provoked slight activation. The residual TsAdh319 activities began to decrease after 2 h of preincubation at 70°C or 80°C and were 10% and 15% down from the control, respectively. The determined half-life values of TsAdh319 were 2 h at 90°C and 1 h at 100°C.Open in a separate windowFIG. 1.Temperature dependence of the initial rate of the 2-propanol reduction by TsAdh319. The reaction was initiated by enzyme addition to a prewarmed 2-propanol-NADP mixture. The inset shows the Arrhenius plot of the same data.Protein thermostability often correlates with such important biotechnological properties as increased solvent tolerance (2). We tested the influence of organic solvents at a high concentration (50% [vol/vol]) on TsAdh319 by using either preincubation of the enzyme at a concentration of 0.2 mg/ml with solvents for 4 h at 55°C or solvent addition into the reaction mixture to distinguish the effect of solvent on the protein stability and on the enzyme activity. TsAdh319 showed significant solvent tolerance in both cases (Table (Table4),4), and the effects of solvents could be modulated by salts, acting apparently as molecular lyoprotectants (17). Furthermore, TsAdh319 maintained 57% of its activity in 25% (vol/vol) 2-propanol, which could be used as the cosubstrate in cofactor regeneration (6).

TABLE 4.

Influence of various solvents on TsAdh319 activitya
SolventRelative activity (%)bRelative activity (%)c
Buffer without NaClBuffer with 600 mM NaCl
None100100100
DMSOd98040
DMFAe1011341
Methanol98259
Acetonitrile9500
Ethyl acetate470*33*
Chloroform10579*81*
n-Hexane10560*118*
n-Decane3691*107*
Open in a separate windowaThe activity measured at the standard condition with 2-propanol as a substrate. Data are averages from triplicate experiments.bPreincubation for 4 h at 55°C in the presence of 50% (vol/vol) of solvent prior the activity assay.cWithout preincubation, solvent addition to the reaction mixture up to 50% (vol/vol) or using the buffer saturated by a solvent (*).dDMSO, dimethyl sulfoxide.eDMFA, dimethylformamide.From all the aforesaid we may suppose TsAdh319 or its improved variant to be interesting both for the investigation of structural features of protein tolerance and for biotechnological applications.  相似文献   

14.
A molecular diagnostic system using single nucleotide polymorphisms (SNPs) was developed to identify four Sclerotinia species: S. sclerotiorum (Lib.) de Bary, S. minor Jagger, S. trifoliorum Erikss., and the undescribed species Sclerotinia species 1. DNAs of samples are hybridized with each of five 15-bp oligonucleotide probes containing an SNP site midsequence unique to each species. For additional verification, hybridizations were performed using diagnostic single nucleotide substitutions at a 17-bp sequence of the calmodulin locus. The accuracy of these procedures was compared to that of a restriction fragment length polymorphism (RFLP) method based on Southern hybridizations of EcoRI-digested genomic DNA probed with the ribosomal DNA-containing plasmid probe pMF2, previously shown to differentiate S. sclerotiorum, S. minor, and S. trifoliorum. The efficiency of the SNP-based assay as a diagnostic test was evaluated in a blind screening of 48 Sclerotinia isolates from agricultural and wild hosts. One isolate of Botrytis cinerea was used as a negative control. The SNP-based assay accurately identified 96% of Sclerotinia isolates and could be performed faster than RFLP profiling using pMF2. This method shows promise for accurate, high-throughput species identification.Sclerotinia is distinguished morphologically from other genera in the Sclerotiniaceae (Ascomycota, Pezizomycotina, Leotiomycetes) by the production of tuberoid sclerotia that do not incorporate host tissue, by the production of microconidia that function as spermatia but not as a disseminative asexual state, and by the development of a layer of textura globulosa composing the outer tissue of apothecia (8). Two hundred forty-six species of Sclerotinia have been reported, most distinguished morphotaxonomically (Index Fungorum [www.indexfungorum.org]). These include the four species of agricultural importance now recognized plus many that are imperfectly known, seldom collected, or apparently endemic to relatively small geographic areas (2, 5, 6, 7, 8, 9, 17).The main species of phythopathological interest in the genus Sclerotinia are S. sclerotiorum (Lib.) de Bary, S. minor Jagger, S. trifoliorum Erikss., and the undescribed species Sclerotinia species 1. Sclerotinia species 1 is an important cause of disease in vegetables in Alaska (16) and has been found in association with wild Taraxacum sp., Caltha palustris, and Aconitum septentrionalis in Norway (7). It is morphologically indistinguishable from S. sclerotiorum, but it was shown to be a distinct species based on distinctive polymorphisms in sequences from internal transcribed spacer 2 (ITS2) of the nuclear ribosomal repeat (7). The other three species have been delimited using morphological, cytological, biochemical, and molecular characters (3, 8, 9, 10, 12, 15). Interestingly, given that the ITS is sufficiently polymorphic in many fungal genera to resolve species, in Sclerotinia, only species 1 and S. trifoliorum are distinguished by characteristic ITS sequence polymorphisms; S. sclerotiorum and S. minor cannot be distinguished based on ITS sequence (2, 7).Sclerotinia sclerotiorum is a necrotrophic pathogen with a broad host range (1). S. minor has a more restricted host range but causes disease in a variety of important crops such as lettuce, peanut, and sunflower crops (11). S. trifoliorum has a much narrower host range, limited to the Fabaceae (3, 8, 9). Sclerotial and ascospore characteristics also serve to differentiate among the three species. Sclerotinia minor has small sclerotia that develop throughout the colony in vitro and aggregate to form crusts on the host, while the sclerotia of S. sclerotiorum and S. trifoliorum are large and form at the colony periphery in vitro, remaining separate on the host (8, 9). The failure of an isolate to produce sclerotia or apothecia in vitro is not unusual, especially after serial cultivation (8). The presence of dimorphic, tetranucleate ascospores characterizes S. trifoliorum, while S. sclerotiorum and S. minor both have uniformly sized ascospores that are binucleate and tetranucleate, respectively (9, 14).With the apparent exception of Sclerotinia species 1, morphological characteristics are sufficient to delimit Sclerotinia species given that workers have all manifestations of the life cycle in hand. In cultures freshly isolated from infected plants, investigators usually have mycelia and sclerotia but not apothecia. Restriction fragment length polymorphisms (RFLPs) in ribosomal DNA (rDNA) are diagnostic for Sclerotinia species (3, 10), but the assay requires cloned probes (usually accessed from other laboratories) hybridized to Southern blots from vertical gels, an impractical procedure for large samples. We have analyzed sequence data from previous phylogenetic studies (2) and have identified diagnostic variation for the rapid identification of the four Sclerotinia species. The single nucleotide polymorphism (SNP) assay that we report here is amenable to a high throughput of samples and requires only PCR amplification with a standard set of primers and oligonucleotide hybridizations to Southern blots in a dot format.The SNP assay was performed using two independent sets of species-specific oligonucleotide probes, all with SNP sites shown to differentiate the four Sclerotinia species (Fig. (Fig.1).1). A panel of 49 anonymously coded isolates (Table (Table1)1) was screened using these species-specific SNP probes, as outlined in Fig. Fig.1.1. The assay was validated by comparison to Southern hybridizations of EcoRI-digested genomic DNA hybridized with pMF2, a plasmid probe containing the portion of the rDNA repeat with the 18S, 5.8S, and 26S rRNA cistrons of Neurospora crassa (4, 10).Open in a separate windowFIG. 1.Protocol for the SNP-based identification of Sclerotinia species, with diagnostic SNP sites underlined and in boldface type for each hybridization probe.

TABLE 1.

Isolates and hybridization results for all SNP-based oligonucleotide probesf
Collector''s isolateAnonymous codePrescreened presumed species identityOriginHostSpecies-specific SNP
IGS50CAL448 S.trifolCAL124CAL448 S.minorRAS148CAL446 S.sp1CAL19ACAL19BCAL448 S.sclero
LMK1849Botrytis cinereaOntario, CanadaAllium cepa
FA2-13Sclerotinia minorNorth CarolinaArachis hypogaea++
W15Sclerotinia minorNorth CarolinaCyperus esculentus++
W1030Sclerotinia minorNorth CarolinaOenothra laciniata++
PF1-138Sclerotinia minorNorth CarolinaArachis hypogaea++
PF18-49714Sclerotinia minorOklahomaArachis hypogaea++
PF17-48246Sclerotinia minorOklahomaArachis hypogaea++
PF19-51948Sclerotinia minorOklahomaArachis hypogaea++
LF-2720Sclerotinia minorUnited StatesLactuca sativa++
AR12811Sclerotinia sclerotiorumArgentinaArachis hypogaea++
AR128216Sclerotinia sclerotiorumArgentinaArachis hypogaea++
LMK2116Sclerotinia sclerotiorumCanadaBrassica napus++
LMK5725Sclerotinia sclerotiorumNorwayRanunculus ficaria++
LMK75415Sclerotinia sclerotiorumNorwayRanunculus ficaria++
UR1939Sclerotinia sclerotiorumUruguayLactuca sativa++
UR4789Sclerotinia sclerotiorumUruguayLactuca sativa++
CA90132Sclerotinia sclerotiorumCaliforniaLactuca sativa++
CA99540Sclerotinia sclerotiorumCaliforniaLactuca sativa++
CA104441Sclerotinia sclerotiorumCaliforniaLactuca sativa++
1980a34Sclerotinia sclerotiorumNebraskaPhaseolus vulgaris++
Ss00113Sclerotinia sclerotiorumNew YorkbGlycine max++
Ssp00531Sclerotinia sclerotiorumNew YorkGlycine max++
H02-V2833Sclerotinia species 1AlaskacUnknown vegetable crop++
H01-V1426Sclerotinia species 1AlaskaUnknown vegetable crop++
LMK74521Sclerotinia species 1NorwayTaraxacum sp.++
02-2611Sclerotinia trifoliorumFinlanddTrifolium pratense+
06-1429Sclerotinia trifoliorumFinlandTrifolium pratense++
2022Sclerotinia trifoliorumFinlandTrifolium pratense++
2-L945Sclerotinia trifoliorumFinlandTrifolium pratense++
3-A524Sclerotinia trifoliorumFinlandTrifolium pratense
5-L912Sclerotinia trifoliorumFinlandTrifolium pratense++
K14Sclerotinia trifoliorumFinlandTrifolium pratense++
K237Sclerotinia trifoliorumFinlandTrifolium pratense++
L-11223Sclerotinia trifoliorumFinlandTrifolium pratense++
L-11944Sclerotinia trifoliorumFinlandTrifolium pratense++
LMK3619Sclerotinia trifoliorumTasmaniaTrifolium repens++
Ssp00118Sclerotinia trifoliorumNew YorkLotus corniculatus++
Ssp00210Sclerotinia trifoliorumNew YorkLotus corniculatus++
Ssp00328Sclerotinia trifoliorumNew YorkLotus corniculatus++
Ssp00436Sclerotinia trifoliorumNew YorkLotus corniculatus++
LMK4743Sclerotinia trifoliorumVirginiaMedicago sativa++
MBRS-127UnknownAustraliaeBrassica spp.++
MBRS-27UnknownAustraliaBrassica spp.++
MBRS-342UnknownAustraliaBrassica spp.++
MBRS-522UnknownAustraliaBrassica spp.++
WW-135UnknownAustraliaBrassica spp.++
WW-28UnknownAustraliaBrassica spp.++
WW-317UnknownAustraliaBrassica spp.++
WW-447UnknownAustraliaBrassica spp.++
Open in a separate windowaThe annotated genome for S. sclerotiorum strain 1980 (ATCC 18683) is publicly available through the Broad Institute, Cambridge, MA (http://www.broad.mit.edu/annotation/genome/sclerotinia_sclerotiorum/Home.html).bAll isolates from New York were provided by Gary C. Bergstrom, Cornell University, Ithaca, NY. Isolates Ss001 and Ssp005 were submitted as S. sclerotiorum, and Ssp001 through Ssp004 were submitted as S. trifoliorum.cAll isolates from Alaska, submitted as Sclerotinia species 1, were provided by Lori Winton, USDA-ARS Subarctic Agricultural Research Unit, University of Alaska, Fairbanks.dAll isolates from Finland, submitted as S. trifoliorum, were provided by Tapani Yli-Mattila, University of Turku, Turku, Finland.eAll isolates from Australia, presumed to be S. sclerotiorum but requiring species confirmation, were provided by Martin Barbetti, DAF Plant Protection Branch, South Perth, Australia.fThe probes that are diagnostic for S. minor, S. sclerotiorum, S. trifoliorum, and Sclerotinia species 1 are listed, with a “+” indicating a positive hybridization for the probe and a “−” indicating no hybridization of the probe.  相似文献   

15.
Arthrobacter sp. strain JBH1 was isolated from nitroglycerin-contaminated soil by selective enrichment. Detection of transient intermediates and simultaneous adaptation studies with potential intermediates indicated that the degradation pathway involves the conversion of nitroglycerin to glycerol via 1,2-dinitroglycerin and 1-mononitroglycerin, with concomitant release of nitrite. Glycerol then serves as the source of carbon and energy.Nitroglycerin (NG) is manufactured widely for use as an explosive and a pharmaceutical vasodilator. It has been found as a contaminant in soil and groundwater (7, 9). Due to NG''s health effects as well as its highly explosive nature, NG contamination in soils and groundwater poses a concern that requires remedial action (3). Natural attenuation and in situ bioremediation have been used for remediation in soils contaminated with certain other explosives (16), but the mineralization of NG in soil and groundwater has not been reported.To date, no pure cultures able to grow on NG as the sole carbon, energy, and nitrogen source have been isolated. Accashian et al. (1) observed growth associated with the degradation of NG under aerobic conditions by a mixed culture originating from activated sludge. The use of NG as a source of nitrogen has been studied in mixed and pure cultures during growth on alternative sources of carbon and energy (3, 9, 11, 20). Under such conditions, NG undergoes a sequential denitration pathway in which NG is transformed to 1,2-dinitroglycerin (1,2DNG) or 1,3DNG followed by 1-mononitroglycerin (1MNG) or 2MNG and then glycerol, under both aerobic and anaerobic conditions (3, 6, 9, 11, 20), and the enzymes involved in denitration have been characterized in some detail (4, 8, 15, 21). Pure cultures capable of completely denitrating NG as a source of nitrogen when provided additional sources of carbon include Bacillus thuringiensis/cereus and Enterobacter agglomerans (11) and a Rhodococcus species (8, 9). Cultures capable of incomplete denitration to MNG in the presence of additional carbon sources were identified as Pseudomonas putida, Pseudomonas fluorescens (4), an Arthobacter species, a Klebsiella species (8, 9), and Agrobacterium radiobacter (20).Here we describe the isolation of bacteria able to degrade NG as the sole source of carbon, nitrogen, and energy. The inoculum for selective enrichment was soil historically contaminated with NG obtained at a facility that formerly manufactured explosives located in the northeastern United States. The enrichment medium consisted of minimal medium prepared as previously described (17) supplemented with NG (0.26 mM), which was synthesized as previously described (18). During enrichment, samples of the inoculum (optical density at 600 nm [OD600] ∼ 0.03) were diluted 1/16 in fresh enrichment medium every 2 to 3 weeks. Isolates were obtained by dilution to extinction in NG-supplemented minimal medium. Cultures were grown under aerobic conditions in minimal medium at pH 7.2 and 23°C or in tryptic soy agar (TSA; 1/4 strength).Early stages of enrichment cultures required extended incubation with lag phases of over 200 h and exhibited slow degradation of NG (less than 1 μmol substrate/mg protein/h). After a number of transfers over 8 months, the degradation rates increased substantially (2.2 μmol substrate/mg protein/h). A pure culture capable of growth on NG was identified based on 16S rRNA gene analysis (504 bp) as an Arthrobacter species with 99.5% similarity to Arthrobacter pascens (GenBank accession no. GU246730). Purity of the cultures was confirmed microscopically and by formation of a single colony type on TSA plates. 16S gene sequencing and identification were done by MIDI Labs (Newark, DE) and SeqWright DNA Technology Services (Houston, TX). The Arthrobacter cells stained primarily as Gram-negative rods with a small number of Gram-positive cocci (data not shown); Gram variability is also a characteristic of the closely related Arthrobacter globiformis (2, 19). The optimum growth temperature is 30°C, and the optimum pH is 7.2. Higher pH values were not investigated because NG begins to undergo hydrolysis above pH 7.5 (data not shown). The isolated culture can grow on glycerol, acetate, succinate, citrate, and lactate, with nitrite as the nitrogen source. Previous authors described an Arthrobacter species able to use NG as a nitrogen source in the presence of additional sources of carbon. However, only dinitroesters were formed, and complete mineralization was not achieved (9).To determine the degradation pathway, cultures of the isolated strain (5 ml of inoculum grown on NG to an OD600 of 0.3) were grown in minimal medium (100 ml) supplemented with NG at a final concentration of 0.27 mM. Inoculated bottles and abiotic controls were continuously mixed, and NG, 1,2DNG, 1,3DNG, 1MNG, 2MNG, nitrite, nitrate, CO2, total protein, and optical density were measured at appropriate intervals. Nitroesters were analyzed with an Agilent high-performance liquid chromatograph (HPLC) equipped with an LC-18 column (250 by 4.6 mm, 5 μm; Supelco) and a UV detector at a wavelength of 214 nm (13). Methanol-water (50%, vol/vol) was used as the mobile phase at a flow rate of 1 ml/min. Nitrite and nitrate were analyzed with an ion chromatograph (IC) equipped with an IonPac AS14A anion-exchange column (Dionex, CA) at a flow rate of 1 ml/min. Carbon dioxide production was measured with a Micro Oxymax respirometer (Columbus Instruments, OH), and total protein was quantified using the Micro BCA protein assay kit (Pierce Biotechnology, IL) according to manufacturer''s instructions. During the degradation of NG the 1,2DNG concentration was relatively high at 46 and 72 h (Fig. (Fig.1).1). 1,3DNG, detected only at time zero, resulted from trace impurities in the NG stock solution. Trace amounts of 1MNG appeared transiently, and trace amounts of 2MNG accumulated and did not disappear. Traces of nitrite at time zero were from the inoculum. The concentration of NG in the abiotic control did not change during the experiment (data not shown).Open in a separate windowFIG. 1.Growth of strain JBH1 on NG. ×, NG; ▵, 1,2DNG; ⋄, 1MNG; □, 2MNG; ○, protein.Results from the experiment described above were used to calculate nitrogen and carbon mass balances (Tables (Tables11 and and2).2). Nitrogen content in protein was approximated using the formula C5H7O2N (14). Because all nitrogen was accounted for throughout, we conclude that the only nitrogen-containing intermediate compounds are 1,2DNG and 1MNG, which is consistent with previous studies (6, 9, 20). The fact that most of the nitrogen was released as nitrite is consistent with previous reports of denitration catalyzed by reductase enzymes (4, 8, 21). The minor amounts of nitrate observed could be from abiotic hydrolysis (5, 12) or from oxidation of nitrite. Cultures supplemented with glycerol or other carbon sources assimilated all of the nitrite (data not shown).

TABLE 1.

Nitrogen mass balance
Time (h)% of total initial nitrogen by mass recovered ina:
Total recovery (%)
1MNG2MNG1,2DNG1,3DNGNGProteinNitriteNitrate
0NDbND0.9 ± 0.70.8 ± 0.682 ± 5.20.8 ± 0.214 ± 0.70.8 ± 0.3100 ± 5.3
460.1 ± 0.00.8 ± 0.27.9 ± 0.4ND35 ± 3.62.0 ± 0.549 ± 1.11.7 ± 0.096 ± 4.2
720.1 ± 0.00.9 ± 0.24.3 ± 4.2ND5.0 ± 0.43.3 ± 0.281 ± 4.23.9 ± 1.998 ± 6.8
94ND0.6 ± 0.4NDND0.6 ± 0.43.2 ± 0.095 ± 102.6 ± 1.6102 ± 10
Open in a separate windowaData represent averages of four replicates ± standard deviations.bND, not detected.

TABLE 2.

Carbon mass balance
Time (h)% of total initial carbon by mass recovered in:
Total recovery (%)
1MNGa2MNGa1,2DNGa1,3DNGaNGaProteinaCO2b
0NDcND1.6 ± 1.21.9 ± 0.492 ± 5.84.4 ± 0.9100 ± 8.4
460.5 ± 0.22.6 ± 0.613 ± 0.7ND39 ± 3.913 ± 3.028 ± 5.796 ± 14.1
720.4 ± 0.02.9 ± 0.77.3 ± 7.0ND5.6 ± 0.422 ± 1.259 ± 8.397 ± 17.6
94ND2.8 ± 0.3NDND0.8 ± 0.518 ± 0.371 ± 4.593 ± 5.6
Open in a separate windowaData represent averages of four replicates ± standard deviations.bData represent averages of duplicates ± standard deviations.cND, not detected.In a separate experiment cells grown on NG were added to minimal media containing 1,3DNG, 1,2DNG, 1MNG, or 2MNG and degradation over time was measured. 1,2DNG, 1,3DNG, and 1MNG were degraded at rates of 6.5, 3.8, and 8 μmol substrate/mg protein/hour. No degradation of 2MNG was detected (after 250 h), which indicates that 2MNG is not an intermediate in a productive degradation pathway. Because 1,3DNG was not observed at any point during the degradation of NG and its degradation rate is approximately one-half the degradation rate of 1,2DNG, it also seems not to be part of the main NG degradation pathway used by Arthrobacter sp. strain JBH1. The above observations indicate that the degradation pathway involves a sequential denitration of NG to 1,2DNG, 1MNG, and then glycerol, which serves as the source of carbon and energy (Fig. (Fig.2).2). The productive degradation pathway differs from that observed by previous authors using both mixed (1, 3, 6) and pure cultures (4, 9, 11, 20), in which both 1,3- and 1,2DNG were intermediates during NG transformation. Additionally, in previous studies both MNG isomers were produced regardless of the ratio of 1,2DNG to 1,3DNG (3, 4, 6, 9, 20). Our results indicate that the enzymes involved in denitration of NG in strain JBH1 are highly specific and catalyze sequential denitrations that do not involve 1,3DNG or 2MNG. Determination of how the specificity avoids misrouting of intermediates will require purification and characterization of the enzyme(s) involved.Open in a separate windowFIG. 2.Proposed NG degradation pathway.Mass balances of carbon and nitrogen were used to determine the following stoichiometric equation that describes NG mineralization by Arthrobacter sp. strain JBH1: 0.26C3H5(ONO2)3 + 0.33O2 → 0.03C5H7O2N + 0.63CO2 + 0.75NO2 + 0.75H+ + 0.17H2O. The result indicates that most of the NG molecule is being used for energy. The biomass yield is relatively low (0.057 mg protein/mg NG), with an fs (fraction of reducing equivalents of electron donor used for protein synthesis) of 0.36 (10), which is low compared to the aerobic degradation of other compounds by pure cultures, for which fs ranges between 0.4 and 0.6 (10, 14). The results are consistent with the requirement for relatively large amounts of energy during the initiation of the degradation mechanism (each denitration probably requires 1 mole of NADH or NADPH [21]).Although NG degradation rates were optimal at pH 7.2, they were still substantial at values as low as 5.1. The results suggest that NG degradation is possible even at low pH values typical of the subsurface at sites where explosives were formerly manufactured or sites where nitrite production lowers the pH.NG concentrations above 0.5 mM are inhibitory, but degradation was still observed at 1.2 mM (data not shown). The finding that NG can be inhibitory to bacteria at concentrations that are well below the solubility of the compound is consistent with those of Accashian et al. (1) for a mixed culture.The ability of Arthrobacter sp. strain JBH1 to grow on NG as the carbon and nitrogen source provides the basis for a shift in potential strategies for natural attenuation and bioremediation of NG at contaminated sites. The apparent specificity of the denitration steps raises interesting questions about the evolution of the pathway.  相似文献   

16.
17.
Eight VanA-type enterococcal strains were isolated from 8 of 171 domestic poultry products by using enrichment by incubation in buffered peptone water at 35°C and 42°C. The pulsed-field gel electrophoresis patterns of all six VanA-type Enterococcus faecalis isolates were nearly indistinguishable, indicating the presence of a specific clone in Japan.The use of avoparcin as a growth promoter and prophylactic agent in feed has been associated with the occurrence of vancomycin-resistant enterococci (VRE) in farm animals in the European Union (EU) (2, 3, 4, 11). In Japan, avoparcin was also used as a food additive on animal farms for approximately 7 years until it was banned in March 1997 (10, 22). However, there has been no documentation in the literature on the isolation of VanA-type VRE from domestic poultry products in Japan (8, 10). Several traditional methods for detecting enterococci, including VRE, in poultry products include an enrichment step using buffered peptone water (BPW) (7, 12, 14, 21). Although BPW is typically incubated at 35 to 37°C, based on the reported optimal growth temperature for enterococci of 35°C (19, 20), the growth of VRE at this temperature can be inhibited by antagonistic activities of background microflora in samples contaminated with low levels of VRE, as 35 to 37°C is also the optimal growth temperature for most mesophilic microorganisms. Several investigators have demonstrated that incubation at an elevated temperature using selective enrichment medium was useful in the isolation of enterococcal strains from retail meat (9, 17, 18). To our knowledge, however, no study has evaluated cultivation temperature during the enrichment process in the isolation of VanA-type VRE from poultry products to examine the influence of background microorganisms on VanA-type VRE growth in BPW. In this study, we carried out isolation of VanA-type VRE from retail Japanese domestic chicken products by using two different enrichment temperatures (35°C and 42°C) and characterized VanA-type VRE isolates by antimicrobial susceptibility testing and pulsed-field gel electrophoresis (PFGE). We also assessed the effects of incubation temperature on the growth of VanA-type VRE under pure culture conditions, on the rates of recovery of inoculated VanA-type VRE strains from artificially contaminated poultry samples, and on the inhibition of suspected enterococcal strains by background microorganisms in poultry samples.A total of 171 retail domestic poultry products obtained in Osaka, Japan, between September 2008 and April 2009 were examined for the presence of VanA-type VRE. Twenty-five grams of each sample was removed aseptically and placed in a sterile plastic bag (30 by 19 cm), to which 225 ml of BPW (Eiken Chemical Co., Ltd., Tokyo, Japan) was added. The sample was then incubated for 24 h at either 35°C or 42°C. Ten microliters of the culture supernatants was spread plated on Enterococcosel agar (Becton, Dickinson and Company, Sparks, MD) plates containing 4 μg/ml vancomycin (Wako Pure Chemical Industries, Ltd., Hyogo, Japan) and incubated at 36°C for 48 h. Screening for the vanA gene was done using the colony-sweep PCR method with phosphate-buffered saline (PBS) washed-cell suspension, Z Taq polymerase (Takara Biochemicals, Shiga, Japan), and previously described primers (1, 16). PCR-positive sweeps were streaked for isolated VanA-type VRE colonies on an Enterococcosel agar plate containing 32 μg/ml vancomycin. The plates were incubated at 36°C for 48 h, and suspected VRE colonies were confirmed as enterococci by conventional biochemical methods (8). In addition, identification of Enterococcus faecalis and Enterococcus faecium harboring the vanA gene was performed by a multiple PCR assay previously described by Depardieu et al. (6). For the five VanA-positive poultry product samples obtained in October 2008 and April 2009, the number of VanA-type VRE was determined by plating 1-ml volumes of samples diluted 10-fold in sterile peptone saline onto Enterococcosel agar plates containing 32 μg/ml vancomycin. Colonies were counted after incubation at 36°C for 48 h. MICs of vanA-genotype VRE isolates to vancomycin and teicoplanin were determined using the Etest method (AB Biodisk, Solna, Sweden) in accordance with the criteria of the Clinical and Laboratory Standards Institute (5). Subtyping of VanA-type VRE isolates was performed by PFGE with SmaI-digested chromosomal DNA (8, 15). PFGE was performed on a 1.0% SeaKem Gold agarose gel (Cambrex Bio Science, Rockland, ME) and a CHEF-DRIII apparatus (Bio-Rad Laboratories, Hercules, CA). The PFGE standard strain Salmonella enterica serovar Braenderup H9812 was obtained from the PulseNet program, Enteric Diseases Laboratory Branch, Centers for Disease Control and Prevention, through the National Institute of Infectious Diseases (Japan).The two representative VanA-type VRE strains were precultured overnight in Trypticase soy broth (Becton, Dickinson and Company) at 36°C. Each culture medium was diluted in BPW to obtain 2 log CFU/ml, and 100 μl of the culture dilution was inoculated into 40 ml of brain heart infusion (BHI) broth (Becton, Dickinson and Company) in a 200-ml Erlenmeyer flask. The flask was incubated at 35°C or 42°C with constant shaking (160 rpm) for 24 h, and the optical density at 600 nm (OD600) of the cultures was monitored every 30 min using an OD-Monitor A & S (Taitec Co., Ltd., Saitama, Japan). Each strain was examined in triplicate. All VanA-type isolates in this study were inoculated onto the poultry samples to compare the effect of BPW incubation at 42°C on the isolation of VanA-type VRE with the effect of incubation at 35°C. The strains were precultured at 36°C for 24 h under static conditions and then serially diluted in BPW to obtain a cell concentration of approximately 3 log CFU/ml. One hundred microliters of each bacterial cell suspension was individually inoculated onto two 25-g portions of each poultry sample in sterile plastic bags. Each inoculated poultry sample was added to 225 ml of BPW and homogenized for 1 min. One sample was incubated at 35°C for 24 h, and the second was incubated at 42°C for 24 h. Colony-sweep PCR analyses with Enterococcosel agar containing 4 μg/ml vancomycin for the presence of the vanA gene were then performed as described above. The artificially contaminated poultry sample experiment was performed in triplicate. To evaluate the success of cultivation using the different enrichment temperatures, data regarding the identification of the vanA gene from the colony-sweep PCR were analyzed using Fisher''s exact test. To examine the effect of enrichment temperature on inhibition of suspected enterococcal strains by background microflora, growth kinetics were compared among these microorganisms in three different poultry samples. For each sample, two 25-g portions were placed into sterile plastic bags to which 225 ml of BPW was added. After homogenization, the samples were incubated at 35°C and 42°C. Each culture was sampled every 3 h for the initial 15 h and after incubation for 24 h. The number of background microorganisms was determined by plating 1-ml volumes of samples serially diluted in PBS onto standard agar (Eiken) plates. Colonies were counted after incubation at 36°C for 24 h. The growth of enterococcal strains was monitored by plating 100-μl volumes of appropriate dilutions onto EF agar plates (Nissui Pharmaceutical Co., Ltd., Tokyo, Japan), and characteristic enterococcal colonies were counted after incubation at 36°C for 48 h.Colony-sweep PCR analyses revealed that 8 of 171 domestic poultry samples were positive for the vanA gene. Isolation of VRE strains from the PCR-positive sweeps resulted in the identification of six vanA-genotype E. faecalis strains and two vanA-genotype E. faecium strains. Of the eight vanA-genotype VRE-positive samples, five were identified from only the 42°C BPW enrichment culture, while only a single isolate was uniquely identified from incubation at 35°C (Table (Table1).1). The number of VanA-type VRE was beneath the detection limit (i.e., less than 10 CFU/g) in each of the five VRE-positive poultry samples obtained in October 2008 and April 2009, suggesting that VanA-type VRE levels might be markedly low in domestic chickens in Japan. All vanA-genotype E. faecalis and E. faecium strains had vancomycin and teicoplanin MICs of >256 μg/ml and ≥32 μg/ml, respectively, confirming that these strains corresponded to the VanA phenotype (Table (Table1).1). Although the PFGE patterns of two VanA-type E. faecium strains were distinct (Fig. (Fig.1,1, lanes 7 and 8), the patterns of all E. faecalis isolates were almost indistinguishable (lanes 1 to 6). Given that the six VanA-type E. faecalis strains were isolated from distinct poultry samples that were obtained from 5 different retail shops, these results indicated that a specific E. faecalis clone harboring the vanA gene may be widely disseminated in Japan. The dominant species of VanA-type VRE isolated from poultry sources has been shown to differ depending on region. In New Zealand, greater than 80% of VRE isolates from broiler fecal samples were identified as vanA-genotype E. faecalis strains with very similar PFGE patterns, indicating that once established, a single, dominant clonal strain is capable of widespread contamination, and supporting the idea of clonal VRE dissemination rather than horizontal transfer of vancomycin resistance elements (13). Our findings also suggest that the epidemiological features of VanA-type VRE strains associated with poultry production environments in Japan may be similar to those observed in New Zealand. To our knowledge, this is the first report of the isolation of VanA-type VRE from retail domestic poultry products in Japan.Open in a separate windowFIG. 1.DNA fingerprinting of VanA-type VRE (E. faecalis and E. faecium strains) isolated from retail domestic poultry samples. Isolated genomic DNA of each isolate was digested with SmaI and subjected to PFGE. Lane 1, E. faecalis Ha8; lane 2, E. faecalis Ha16; lane 3, E. faecalis Ha36; lane 4, E. faecalis Ha55; lane 5, E. faecalis Ha58; lane 6, E. faecalis Ha61; lane 7, E. faecium Ha20; lane 8, E. faecium Ha26; lanes M, XbaI-digested PFGE patterns of DNA size standard Salmonella enterica serovar Braenderup H9812. Numbers to the left of the gel image indicate the molecular size in kilobase pairs.

TABLE 1.

Characteristics of vancomycin-resistant enterococcal strains isolated from domestic poultry samples in this study
Strain no.Enterococcus speciesVancomycin resistance geneMIC (μg/ml)
Enrichment tempa (°C)Isolation date
VancomycinTeicoplanin
Ha8E. faecalisvanA>256>25642October 2008
Ha16E. faecalisvanA>256>25642October 2008
Ha36E. faecalisvanA>256>25642March 2009
Ha55E. faecalisvanA>25612842April 2009
Ha58E. faecalisvanA>2563235April 2009
Ha61E. faecalisvanA>25625635/42April 2009
Ha20E. faeciumvanA>2563242November 2008
Ha26E. faeciumvanA>2563235/42December 2008
Open in a separate windowaAbbreviations: 35, a strain isolated only from BPW enrichment culture incubated at 35°C; 42, a strain isolated only from BPW enrichment culture incubated at 42°C; 35/42, a strain isolated from BPW enrichment culture incubated at 35°C and 42°C.The growth kinetics of E. faecalis Ha8 and E. faecium Ha20 cultured under pure culture conditions in BHI broth incubated at 35°C or 42°C are shown in Fig. Fig.2.2. For both isolates, initiation of the exponential growth phase was shortened by approximately 2 to 3 h at the 42°C incubation temperature. In poultry samples artificially contaminated with VanA-type VRE isolates, a significant difference in the detection of the vanA gene using colony-sweep PCR with BPW was observed between incubation at 35°C and incubation at 42°C, with detection rates of 11/24 and 19/24, respectively (P < 0.05) (Table (Table2),2), indicating that the use of the 42°C enrichment step increased the recovery rate of the VanA-type VRE strains, which was significantly higher than that at 35°C. Figure Figure33 illustrates the growth kinetics of background microflora and the suspected enterococcal strains isolated from three independent retail poultry products in BPW enrichment cultures incubated at 35°C and 42°C. The initial levels of background microflora and suspected enterococcal strains in samples 1, 2, and 3 were 4.3 and 1.0 log CFU/ml, 3.2 and 0.7 log CFU/ml, and 5.1 and 2.9 log CFU/ml, respectively. After the BPW enrichment step, the maximum densities of background microflora in cultures incubated at 35°C were similar to those at 42°C. In contrast, the maximum levels of suspected enterococcal strains in BPW enrichment cultures incubated at 35°C and 42°C in samples 1, 2, and 3 were 6.0 and 7.5 log CFU/ml, 5.6 and 7.9 log CFU/ml, and 7.3 and 8.0 log CFU/ml, respectively (Fig. (Fig.3).3). These results suggested that the impact of elevated incubation temperature on the enrichment of VRE may be greater in poultry with low levels of enterococcal contamination, such as the VanA-positive samples obtained in this study. We conclude that BPW incubation at 42°C is superior to incubation at 35°C for the enrichment and detection of VanA-type VRE in poultry samples.Open in a separate windowFIG. 2.Growth curves of E. faecalis Ha8 (A) and E. faecium Ha20 (B) cultured in BHI broth incubated at either 35°C (▪) or 42°C (○). Results obtained from three independent experiments are presented as means ± standard deviations.Open in a separate windowFIG. 3.Growth curves of background microflora and suspected enterococcal strains in three different poultry samples cultured in BPW incubated at either 35°C or 42°C. ▴, background microflora; □, suspected enterococcal strains. Solid lines show the data for incubation at 35°C, and dotted lines show those for incubation at 42°C.

TABLE 2.

Comparison of vanA gene detection by colony-sweep PCR after enrichment incubation with BPW at 35°C and 42°C
Inoculum strainInoculum dose (CFU/25 g)No. of positive samples/no. of samples tested with incubation temp:
35°C42°C
E. faecalis Ha8300-3102/33/3
E. faecalis Ha16130-6002/33/3
E. faecalis Ha36100-2901/32/3
E. faecalis Ha55630-1,4002/33/3
E. faecalis Ha58260-6602/33/3
E. faecalis Ha61780-1,7002/33/3
E. faecium Ha20260-4500/32/3
E. faecium Ha26350-5100/30/3
Total11/2419/24
Open in a separate window  相似文献   

18.
19.
A real-time quantitative PCR-based detection assay targeting the dnaJ gene (encoding heat shock protein 40) of the coral pathogen Vibrio coralliilyticus was developed. The assay is sensitive, detecting as little as 1 CFU per ml in seawater and 104 CFU per cm2 of coral tissue. Moreover, inhibition by DNA and cells derived from bacteria other than V. coralliilyticus was minimal. This assay represents a novel approach to coral disease diagnosis that will advance the field of coral disease research.Vibrio coralliilyticus has recently emerged as a coral pathogen of concern on reefs throughout the Indo-Pacific. It was first implicated as the etiological agent responsible for bleaching and tissue lysis of the coral Pocillopora damicornis on Zanzibar reefs (2). More recently, V. coralliilyticus has been identified as the causative agent of white syndrome (WS) outbreaks on several Pacific reefs (14). WS is a collective term describing coral diseases characterized by a spreading band of tissue loss exposing white skeleton on Indo-Pacific scleractinian corals (16). V. coralliilyticus is an emerging model pathogen for understanding the mechanisms linking bacterial infection and coral disease (13) and therefore provides an ideal model for the development of diagnostic assays to detect coral disease. Current coral disease diagnostic methods, which are based primarily upon field-based observations of macroscopic disease signs, often detect disease only at the latest stages of infection, when control measures are least effective. The development of diagnostic tools targeting pathogens underlying coral disease pathologies may provide early indications of infection, aid the identification of disease vectors and reservoirs, and assist managers in developing strategies to prevent the spread of coral disease outbreaks. In this paper, we describe the development and validation of a TaqMan-based real-time quantitative PCR (qPCR) assay that targets a segment of the V. coralliilyticus heat shock protein 40-encoding gene (dnaJ).Nucleotide sequences of the dnaJ gene were retrieved from relevant Vibrio species, including V. coralliilyticus (LMG 20984), using the National Center for Biotechnology Information''s (NCBI) Entrez Nucleotide Database search tool (http://www.ncbi.nlm.nih.gov/). Gene sequences of strains not available in public databases (V. coralliilyticus strains LMG 21348, LMG 21349, LMG 21350, LMG 10953, LMG 20538, LMG 23696, LMG 23691, LMG 23693, LMG 23692, and LMG 23694) were obtained through extraction of total DNA using a Promega Wizard Prep DNA Purification Kit (Promega, Sydney, Australia), PCR amplification, and sequencing using primers and thermal cycling parameters described by Nhung et al. (8). A 128-bp region (nucleotides 363 to 490) containing high concentrations of single nucleotide polymorphisms (SNPs), which were conserved within V. coralliilyticus strains but differed from non-V. coralliilyticus strains, was identified, and oligonucleotide primers Vc_dnaJ_F1 (5′-CGG TTC GYG GTG TTT CAA AA-3′) and Vc_dnaJ_R1 (5′-AAC CTG ACC ATG ACC GTG ACA-3′) and a TaqMan probe, Vc_dnaJ_TMP (5′-6-FAM-CAG TGG CGC GAA G-MGBNFQ-3′; 6-FAM is 6-carboxyfluorescein and MGBNFQ is molecular groove binding nonfluorescent quencher), were designed to target this region. The qPCR assay was optimized and validated using DNA extracted from V. coralliilyticus isolates, nontarget Vibrio species, and other bacterial species grown in marine broth (MB) (Table (Table1),1), under the following optimal conditions: 1× TaqMan buffer A, 0.5 U of AmpliTaq Gold DNA polymerase, 200 μM deoxynucleotide triphosphates (with 400 μM dUTP replacing deoxythymidine triphosphate), 0.2 U of AmpErase uracil N-glycosylase (UNG), 3 mM MgCl2, 0.6 μM each primer, 0.2 μM fluorophore-labeled TaqMan, 1 μl of template, and sterile MilliQ water for a total reaction volume to 20 μl. All assays were conducted on a RotoGene 300 (Corbett Research, Sydney, Australia) real-time analyzer with the following cycling parameters: 50°C for 120 s (UNG activation) and 95°C for 10 min (AmpliTaq Gold DNA polymerase activation), followed by 40 cycles of 95°C for 15 s (denaturation) and 60°C for 60 s (annealing/extension). During the annealing/extension phase of each thermal cycle, fluorescence was measured in the FAM channel (470-nm excitation and 510-nm detection).

TABLE 1.

Species, strain, and threshold cycle for all bacterial strains testeda
SpeciesStrainbOriginHost organismCT ± SEMcdnaJ gene sequence accession no.Reference
Vibrio coralliilyticusLMG 23696Nelly Bay, Magnetic Island, AustraliaMontipora aequituberculata12.43 ± 0.20HM21557014
LMG 23691Majuro Atoll, Republic of Marshall IslandsAcropora cytherea14.07 ± 1.33HM21557114
LMG 23693Nikko Bay, PalauPachyseris speciosa10.83 ± 2.76HM21557214
LMG 23692Nikko Bay, PalauPachyseris speciosa9.40 ± 0.36HM21557314
LMG 23694Nikko Bay, PalauPachyseris speciosad12.54 ± 0.24HM21557414
LMG 20984TIndian Ocean, Zanzibar, TanzaniaPocillopora damicornis12.80 ± 0.71HM2155752
LMG 21348Red Sea, Eilat, IsraelPocillopora damicornis13.81 ± 0.49HM2155763
LMG 21349Red Sea, Eilat,Pocillopora damicornis12.98 ± 0.94HM2155773
LMG 21350Red Sea, Eilat,Pocillopora damicornis11.49 ± 0.19HM2155783
LMG 10953Kent, United KingdomCrassostrea gigas (oyster) larvae10.53 ± 0.40HM2155793
LMG 20538Atlantic Ocean, Florianópolis, BrazilNodipecten nodosus (bivalve) larvae12.13 ± 0.50HM2155803
C1Caribbean Sea, La Parguera, Puerto RicoPseudopterogorgia americana14.53 ± 0.28HM21556815
C2Caribbean Sea, La Parguera, Puerto RicoPseudopterogorgia americanaNAHM21556915
Vibrio alginolyticusATCC 1774933.74 ± 0.33
Vibio brasiliensisDSM 1718437.84†
Vibrio calviensisDSM 1434727.06 ± 0.52
Vibrio campbelliiATCC 25920T39.10†
Enterovibrio campbelliiLMG 2136337.33 ± 2.41
Alliivibrio fischeriDSM 50731.36 ± 1.42
Vibrio fortisDSM 19133NA
Vibrio furnissiiDSM 19622NA
Vibrio harveyiDSM 19623NA
Vibrio natriegensATCC 1404828.56 ± 0.60
Vibrio neptuniusLMG 20536NA
Vibrio ordaliiATCC 3350925.56 ± 0.41
Vibrio parahaemolyticusATCC 17802NA
Vibrio proteolyticusATCC 1533830.00 ± 0.89††
Vibrio rotiferianusLMG 21460NA
Vibrio splendidusATCC 3312532.31 ± 0.82
Vibrio tubiashiiATCC 19109NA
Vibrio xuiiLMG 21346NA
Escherichia coliATCC 25922NA
Psychrobacter sp.AIMS 1618NA
Shewanella sp.AIMS C04125.34 ± 0.45
Open in a separate windowaOrigin, host organism, and dnaJ gene sequence accession numbers are shown for V. coralliilyticus strains.bStrain designations beginning with LMG were derived from the Belgian Coordinated Collections of Microorganisms, ATCC strains are from the American Type Culture Collection, DSM strains are from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH culture collection, AIMS strains are from the Australian Institute of Marine Science culture collection, and C1 and C2 were provided by Pamela Morris.c†, amplification in one of three reactions; ††, amplification in two of three reactions; NA, no amplification.dIsolated from seawater above coral.The qPCR assay specifically detected 12 out of 13 isolated V. coralliilyticus strains tested in this study (Table (Table1).1). The exception was one Caribbean strain (C2), which failed to give specific amplification despite repeated attempts. Positive detection of the target gene segment was determined by the increase in fluorescent signal beyond the fluorescence threshold value (normalized fluorescence, 0.010) at a specific cycle, referred to as the threshold cycle (CT). Specific detection was further confirmed by gel electrophoresis, which revealed a PCR product of the correct theoretical size (128 bp) (data not shown), and DNA sequencing, which confirmed the target amplified product to be a segment of the dnaJ gene. No amplification with the assay was detected for 13 other closely related Vibrio strains, including the closely related Vibrio neptunius and two non-Vibrio species (Table (Table1).1). A total of five other Vibrio strains and one non-Vibrio strain (Shewanella sp.) exhibited CT values less than the cutoff of 32 cycles. However, CT values for these strains (mean ± standard error of the mean [SEM], 27.96 ± 2.40) were all much higher than those for V. coralliilyticus strains (12.30 ± 1.52), and no amplicons were evident in post-qPCR gel electrophoresis (data not shown).The detection limit for purified V. coralliilyticus genomic DNA was 0.1 pg of DNA, determined by performing 10-fold serial dilutions (100 ng to 0.01 pg per reaction), followed by qPCR amplification. Similarly, qPCR assays of serial dilutions of V. coralliilyticus (LMG 23696) cells cultured overnight in MB (108 CFU ml−1 to extinction) were able to detect as few as 104 CFU (Fig. (Fig.1).1). Standard curves revealed a strong linear negative correlation between CT values and both DNA and cell concentrations of V. coralliilyticus over several orders of magnitude, with r2 values of 0.998 and 0.953 for DNA and cells, respectively (Fig. (Fig.11).Open in a separate windowFIG. 1.Standard curves delineating threshold (CT) values of fluorescence for indicators of pathogen presence: (A) concentration of V. coralliilyticus DNA and (B) number of V. coralliilyticus cells in pure culture. Error bars indicate standard error of the mean for three replicate qPCRs.Little interference of the qPCR assay was observed when purified V. coralliilyticus (LMG 23696) DNA (10 ng) was combined with 10-fold serial dilutions (0.01 to 100 ng per reaction) of non-V. coralliilyticus DNA (i.e., Vibrio campbellii [ATCC 25920T]). Over the entire range of nontarget DNA concentrations tested, the resulting CT values (mean ± SEM, 17.76 ± 0.53) were not significantly different from those of a control treatment containing 10 ng of V. coralliilyticus DNA and no nonspecific DNA (16.75 ± 0.18; analysis of variance [ANOVA], P = 0.51) (Table (Table2).2). Detection of V. coralliilyticus (LMG 23696) bacterial cells (104, 105, 106, 107, or 108 CFU per ml) in a background of non-V. coralliilyticus cells (i.e., V. campbellii [ATCC 25920T] at 0, 10, 104, or 107 CFU per ml) showed little reduction in assay sensitivity (see Fig. S1 in the supplemental material). For example, when V. coralliilyticus was seeded at 107 cells with similarly high concentrations of nontarget cells, little inhibition of the assay was observed.

TABLE 2.

Effect of nontarget bacterial DNA on the detection of 10 ng of purified V. coralliilyticus DNA
Amt of nontarget DNA (ng)CT (mean ± SEM)
10016.97 ± 0.33
1016.9 ± 0.08
116.74 ± 0.10
0.117 ± 0.09
0.0116.37 ± 0.43
0a16.75 ± 0.18
NTCb35.04 ± 0.02
Open in a separate windowaV. coralliilyticus (LMG 23696) DNA (10 ng) free of nontarget DNA and cells served as positive controls.bA qPCR mixture containing no bacterial DNA served as a no-template, or negative, control (NTC).The assay''s detection limit in seawater was tested by inoculating 10-fold serial dilutions of V. coralliilyticus (LMG 23696) cultures (grown overnight in MB medium, pelleted at 14,000 rpm for 10 min, and washed twice with sterile phosphate-buffered saline [PBS]) into 1 liter of seawater (equivalent final concentrations were 106 to 1 CFU ml−1). The entire volume of V. coralliilyticus-seeded seawater was filtered through a Sterivex-GP filter (Millipore), and DNA was extracted using the method described by Schauer et al. (11). The lowest detection limit for V. coralliilyticus cells seeded into seawater was 1 CFU ml−1 (Fig. (Fig.2),2), with no detection in a 1-liter volume of an unseeded seawater negative control. Standard curves revealed a strong correlation between CT values and the concentrations of V. coralliilyticus bacteria seeded into the seawater over several orders of magnitude (r2 of 0.968) (Fig. (Fig.22).Open in a separate windowFIG. 2.Standard curves showing CT values of the fluorescent signal versus the number of V. coralliilyticus cells per ml seawater (▿), and cells per cm2 of M. aequituberculata tissue, with (○) or without (·) enrichment. Each dot represents an independent experiment. Error bars indicate standard error of the mean for three replicate qPCR runs.The detection limit in seeded coral tissue homogenate was determined by seeding 10-fold dilutions (1010 to 103 CFU ml−1) of pelleted, PBS-washed and resuspended (in 10 ml of sterile PBS) V. coralliilyticus cells onto healthy fragments (∼10 cm2) of the coral Montipora aequituberculata collected from Nelly Bay (Magnetic Island, Australia). Corals were collected in March 2009 and maintained in holding tanks supplied with flowthrough ambient seawater. Resuspended cells were inoculated onto M. aequituberculata fragments, each contained in an individual 3.8-liter plastic bag, allowed to sit at room temperature for 30 min, and then air brushed with compressed air until only white skeleton remained. One-milliliter aliquots of the resulting slurry (PBS, bacteria, and coral tissue) was vortexed for 10 min at 14,000 rpm, and DNA was extracted using a PowerPlant DNA Isolation Kit (Mo Bio, Carlsbad, CA). The lowest detection limits for V. coralliilyticus cells seeded onto coral fragments was 104 CFU per cm2 of coral tissue (Fig. (Fig.2).2). Again, standard curves revealed a strong correlation between CT values and the concentrations of seeded bacteria over several orders of magnitude (r2 of 0.981) (Fig. (Fig.2).2). When a 1-ml aliquot of the slurry was also inoculated into 25 ml of MB and enriched for 6 h at 28°C (with shaking at 170 rpm), the detection limit increased by 1 order of magnitude, to 103 CFU of V. coralliilyticus per cm2 of coral tissue (Fig. (Fig.2).2). The slope of the standard curve reveals some inhibition, particularly at the highest V. coralliilyticus concentrations, which could result from lower replication rates in the cultures with the highest bacterial densities (i.e., 109 CFU). However, since this effect is most pronounced only at the highest bacterial concentrations, the detection limit is still valid. In all trials, unseeded coral fragments and enrichment cultures derived from uninoculated coral fragments served as negative controls.The current study describes the first assay developed to detect and quantify a coral pathogen using a real-time quantitative PCR (qPCR) approach. While previous studies have utilized antibodies or fluorescent in situ hybridization (FISH) to detect coral pathogens (1, 6), the combination of high sensitivity and specificity, low contamination risk, and ease and speed of performance (5) make qPCR technology an ideal choice for rapid pathogen detection in complex hosts, such as corals. The assay developed is highly sensitive for V. coralliilyticus, detecting as few as 1 CFU ml−1 of seawater and 104 CFU cm−2 of coral tissue (103 CFU cm−2 of coral tissue with a 6-h enrichment). These detection limits are likely to be within biologically relevant pathogen concentrations. For example, antibodies for specific detection of the coral bleaching pathogen Vibrio shiloi showed that bacterial densities reached 8.4 × 108 cells cm−3 1 month prior to maximum visual bleaching signs on the coral Oculina patagonica (6). Each seeded seawater and coral (enriched and nonenriched) dilution assay was performed in triplicate. The linearity of the resulting standard curves indicates consistent extraction efficiencies over V. coralliilyticus concentrations spanning 6 orders of magnitude (Fig. (Fig.2)2) and provides strong support for the robustness of the assay. In addition, the presence of competing, non-V. coralliilyticus bacterial cells and DNA had a minimal impact on the detection of V. coralliilyticus. This is an important consideration for accurate detection within the complex coral holobiont, where the target organism is present within a matrix of other microbial and host cells.V. coralliilyticus, like V. shiloi (10), is becoming a model pathogen for the study of coral disease. Recent research efforts have characterized the organism''s genome (W. R. Johnson et al., submitted for publication), proteome (N. E. Kimes et al., submitted for publication), resistome (15), and metabolome (4) and enhanced our understanding of the genetic (7, 9) and physiological (7, 13) basis of its virulence. Before effective management response plans can be formulated, however, continuing research on the genetic and cellular aspects of V. coralliilyticus must be complemented with knowledge of the epidemiology of this pathogen, including information on its distribution, incidence of infection, and rates of transmission throughout populations. The V. coralliilyticus-specific qPCR assay developed in this study will provide important insights into the dynamics of pathogen invasion and spread within populations (6) while also aiding in the identification of disease vectors and reservoirs (12). These capabilities will play an important role in advancing the field of coral disease research and effective management of coral reefs worldwide.   相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号