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 78 毫秒
1.
Mycobacterium avium subsp. paratuberculosis, the causative agent of Johne''s disease in cattle, was identified in settled-dust samples of Dutch commercial dairy farms, both in the dairy barn and in the young stock housing. Bioaerosols may play a role in within-farm M. avium subsp. paratuberculosis transmission.Paratuberculosis is an infectious enteric disease caused by Mycobacterium avium subsp. paratuberculosis leading to economic losses in dairy cattle globally (2, 10). The main transmission route is the fecal/oral route from infectious adult cattle to susceptible calves (12).Preventive calf management was a key point in model studies (7), but 20-year implementation did not lead to farm-level eradication, suggesting uncontrolled routes of transmission (1, 7).Environmental samples were used to classify commercial dairy herds (3, 9, 11), based on long-term survival of M. avium subsp. paratuberculosis in the environment (16). Recently, bioaerosols containing viable M. avium subsp. paratuberculosis were identified in an experimental setting with 100% M. avium subsp. paratuberculosis prevalence (6) and may thus be a mode of transmission. Dust containing M. avium subsp. paratuberculosis might be ingested or inhaled by calves (4). Experimental M. avium subsp. paratuberculosis challenge studies in sheep successfully used inhalation (8). These transmission routes could hamper current control programs. Our objective was to study whether M. avium subsp. paratuberculosis could be detected in bioaerosols on commercial Dutch dairy farms.Dairy herds in three Dutch veterinary practices were sampled in 2009. All farms participated in a Dutch M. avium subsp. paratuberculosis monitoring program in 2008, either the Dutch Paratuberculosis Program (PPN; n = 2) or the Dutch Bulk Milk Quality Assurance Program (BMQAP; n = 22) (15). Both PPN herds were certified M. avium subsp. paratuberculosis-free. Herds corresponding to the BMQAP had at least one positive animal identified by enzyme-linked immunosorbent assay (ELISA) (Pourquier ELISA; Institut Pourquier, France). Farms were grouped into three M. avium subsp. paratuberculosis test prevalence levels (control, zero positive animals; group A, one positive animal; group B, two or more positive animals; Table Table11).

TABLE 1.

Overview of the results of the questionnaire about relevant M. avium subsp. paratuberculosis management practicesa
ParameterValue for groupb
Control (n = 2)A (n = 8)B (n = 14)
Mean herd size (SD)69 (15)67 (19)102 (26)
Median no. of ELISA-positive cows (maximum)0 (0)1 (1)3 (10)
No. of farms with:
    Cow brush in barn2513
    Cow barn cleaned in summer with high-pressure cleaner064
    Dry cows in young stock housing033
    Young stock housed separately178
    Young stock housing empty in summer000
    Young stock housing cleaned with high-pressure cleaner061
Open in a separate windowaResults of the questionnaire about relevant M. avium subsp. paratuberculosis management practices in 24 Dutch farms enrolled in this study with 0 (control), 1 (group A), or ≥2 (group B) ELISA-positive animals.bn, number of farms.Farms were visited twice during the housing period. Sampling locations were above the animal level inside the barn. At the first visit (sampling 1 [S1]), settled dust was collected with wipes and a short management questionnaire was taken. At the same time, five to seven electrostatic dust collectors (EDC; Zeeman, Alphen a/d Rhijn, Netherlands) were installed and collected after 4 weeks (sampling 2 [S2]) (6). Settled-dust samples were processed according to a previously described method (6). Results are presented as proportions of positive locations. McNemar''s χ2 test was performed to investigate whether S1 differed from S2.No M. avium subsp. paratuberculosis was detected by real-time PCR in any of the settled-dust samples at control farms (Fig. (Fig.1).1). M. avium subsp. paratuberculosis DNA was detected in dust samples at S1 and S2 in more than 50% of the group A and B farms, with seven farms consistently positive. M. avium subsp. paratuberculosis DNA was detected in the young stock area in 3/6 (S1) and 2/6 (S2) farms of group B with single-barn housing. M. avium subsp. paratuberculosis DNA was also detected in settled-dust samples from separate young stock housings in three farms, of which two cohoused dry cows.Open in a separate windowFIG. 1.Proportions of farms with M. avium subsp. paratuberculosis DNA detected in settled-dust samples collected at samplings 1 and 2. Black bar, control (n = 2); checked bar, group A (n = 8); white bar, group B (n = 14).At control farms, no viable M. avium subsp. paratuberculosis was detected in any of the collected dust samples (Fig. (Fig.2).2). Viable M. avium subsp. paratuberculosis was detected in 6 B farms at S1. At S2, viable bacteria were present in 3 A farms and in the majority of B farms (Table (Table2).2). On five farms in group B, viable M. avium subsp. paratuberculosis was detected at both samplings.Open in a separate windowFIG. 2.Proportions of farms with viable M. avium subsp. paratuberculosis detected in settled-dust samples collected at samplings 1 and 2. Black bar, control (n = 2); checked bar, group A (n = 8); white bar, group B (n = 14).

TABLE 2.

Detection of M. avium subsp. paratuberculosis DNA or viable M. avium subsp. paratuberculosis in 5 to 7 settled-dust samples collected at sampling 1 or 2
No. of positive dust samplesNo. of farms with:
M. avium subsp. paratuberculosis DNA
Viable M. avium subsp. paratuberculosis
Control (n = 2)
Group A (n = 8)
Group B (n = 14)
Control (n = 2)
Group A (n = 8)
Group B (n = 14)
S1S2S1S2S1S2S1S2S1S2S1S2
0224345228586
13446124
243112
31111112
412
Open in a separate windowViable M. avium subsp. paratuberculosis was detected in the young stock housing in 4 and 3 farms of group B with single-barn housing at S1 and S2, respectively. No viable M. avium subsp. paratuberculosis was detected in separate young stock housings.To our knowledge, this study is the first to confirm the presence of M. avium subsp. paratuberculosis DNA as well as viable M. avium subsp. paratuberculosis in settled-dust samples of commercial dairy farms. M. avium subsp. paratuberculosis dispersion by bioaerosols under experimental conditions was already described (6). These findings support the concept of dust-based environmental dispersion of M. avium subsp. paratuberculosis within farms.The relatively small number of farms and the convenience sampling are limitations of this study that could have introduced bias. However, this study is a proof of principle that viable M. avium subsp. paratuberculosis can be detected in settled-dust samples on farms with a low M. avium subsp. paratuberculosis prevalence. The environmental method also seems specific for M. avium subsp. paratuberculosis, since no M. avium subsp. paratuberculosis could be detected in any samples of known M. avium subsp. paratuberculosis-free herds.Paratuberculosis control measures aim to prevent fecal-oral contact between infectious shedding adults and susceptible calves as the main transmission route of M. avium subsp. paratuberculosis. Several studies showed that “calf hygiene improvement” decreased prevalence but did not eliminate the disease (1, 7, 14), suggesting the existence of other transmission routes. In utero transmission, transmission via milk, and calf-to-calf transmission have been described previously (1, 12, 13). Additionally, infection via ingestion and/or inhalation of bioaerosols may be possible (4, 8).Twenty-three of 24 herds were housed in free stalls with one tie-stall herd. Most farmers (n = 15) separated young stock from adult cattle as standard procedure. However, six of these farmers cohoused dry cows in the young stock housing occasionally, indicating the difficulties of consequently implementing management advice. Three farmers did not raise young stock on their farms. In almost all barns, cow brushes were present, as they were recommended to enhance cow well-being in group housings (5), but at the same time they contribute to aerosolization of dust. Animal movement on slatted floors also contributes to dust formation, especially during the winter housing period.Most farmers from group A farms, compared to only a few from group B farms, intended to clean their barns yearly, but only 50% met this aim. Young stock housings were never totally empty, but high-pressure cleaning was occasionally performed at 6/8 farms of group A and at 1 of group B. The numbers of farms in this study precluded statistical testing, but the difference in cleaning attitude seemed remarkable.Comparison of the two methods of dust collection showed no statistical difference. No M. avium subsp. paratuberculosis, neither DNA nor viable M. avium subsp. paratuberculosis, could be detected on known negative farms, whereas on farms of groups A and B, M. avium subsp. paratuberculosis DNA was present in comparable numbers of locations. Viable M. avium subsp. paratuberculosis was present only in group B farms at S1 and in both group A and B farms at S2. It seems that M. avium subsp. paratuberculosis can survive in dust for some time. Besides having a possible role in M. avium subsp. paratuberculosis transmission, dust might also be a useful predictor of M. avium subsp. paratuberculosis presence or M. avium subsp. paratuberculosis introduction on dairy farms, even on farms with low M. avium subsp. paratuberculosis prevalence.In conclusion, this study showed that dust on farms with a low M. avium subsp. paratuberculosis seroprevalence contained viable M. avium subsp. paratuberculosis, which indicated a role in M. avium subsp. paratuberculosis transmission. Further research is needed to study if and how infection with M. avium subsp. paratuberculosis-contaminated dust is possible. Additionally, dust sampling may be an alternative tool to monitor M. avium subsp. paratuberculosis status in control programs.  相似文献   

2.
A microtiter alamarBlue assay was adapted and optimized for Mycobacterium avium subsp. paratuberculosis. Using cell concentrations ranging from 104 to 108 CFU/ml, a minimum incubation time to indicate viability was obtained after 24 h. Rifampin (rifampicin) was used to demonstrate that this method has applications for high-throughput screening against M. avium subsp. paratuberculosis.Mycobacterium avium subsp. paratuberculosis is a chronic enteric pathogen which is widely distributed throughout the food chain (4, 16). Its association with Johne''s disease in cattle is economically significant, with the United States alone suffering losses of $1.5 billion a year (6). Furthermore, its potential to cause human disease is disconcerting and controversial (7, 10, 11, 13). Therefore, the identification and development of novel anti-M. avium subsp. paratuberculosis agents are urgently required. In order to facilitate this, it is important to have available rapid anti-M. avium subsp. paratuberculosis assays permitting high-throughput analysis. A microtiter alamarBlue assay (currently untested for M. avium subsp. paratuberculosis) is a reliable means of determining cellular viability in bacteria (9). This rapid and inexpensive assay lends itself to a high-throughput screening format and has been shown to be applicable to some species of mycobacteria (3, 15). Furthermore, its correlation with other more expensive methods for determining mycobacterium viability is high, between 93 and 100% (1, 5, 15). These include the Mycobacteria Growth Indicator Tube, the Bactec radiometric method, and luciferase reporter systems. This study set out to establish and optimize a microtiter alamarBlue assay for a broad range of M. avium subsp. paratuberculosis titers and to evaluate applications for this assay, including high-throughput screening of novel anti-M. avium subsp. paratuberculosis compounds, and antibiotic resistance profiling of M. avium subsp. paratuberculosis.M. avium subsp. paratuberculosis (CIT03) was isolated from the feces of an infected cow, as described by Ristow et al. (12), and cultivated on Herrold''s egg yolk medium agar supplemented with mycobactin J (2 μl/ml), amphotericin B (50 μg/ml), vancomycin (50 μg/ml), and nalidixic acid (50 μg/ml) for 16 weeks at 37°C. The identity of M. avium subsp. paratuberculosis was confirmed using acid-fast staining, mycobactin dependency, and PCR analysis as previously described (2, 8, 14). To generate sufficient biomass, M. avium subsp. paratuberculosis was subsequently grown in Middlebrook 7H9 broth (MB broth), supplemented with oleic acid, albumin, dextrose, catalase (10%; Becton Dickinson), glycerol (0.2%), and mycobactin J (0.2%). This generally took 16 weeks.Prior to the assay, 10 ml of the 16-week culture was centrifuged at 15,000 rpm for 20 min. The pellet was washed in fresh MB broth and resuspended in 10 ml of fresh supplemented MB broth containing 0.2% mycobactin J. The turbidity was adjusted to match McFarland standard no. 1 (3 × 108 CFU/ml). From this suspension, a series of 1:5 dilutions ranging from 3 × 108 to 9.6 × 104 CFU/ml was set up in MB broth (5-ml volumes), using sterile Falcon tubes. The microtiter plate was organized into rows B, C, D, E, F, and G. Two-hundred-microliter aliquots of 3 × 108 CFU/ml M. avium subsp. paratuberculosis were added to 10 wells in row B. Two-hundred-microliter aliquots of 6 × 107 CFU/ml were added to row C, 1.2 × 107 CFU/ml to row D, 2.4 × 106 CFU/ml to row E, 4.8 × 105 CFU/ml to row F, and 9.6 × 104 CFU/ml to row G. This assay was allowed to progress over a period of 11 days. Twenty microliters (10% of the volume in the well) of a fresh alamarBlue reagent (AbD Serotec) was added, with mixing, to each column on each sampling day. Plates were covered and resealed with Parafilm and incubated at 37°C after the addition of the dye. Absorbance readings at 570 and 600 nm were then taken at 6 h, 24 h, and 48 h for each column. The assay was performed in triplicate, and percent reduction values of alamarBlue were determined using the appropriate formula (www.biokom.com.pl/files/alamarblue.pdf).In terms of optimization, this assay examined the influence of cell numbers, the cellular incubation time, and the optimal incubation time with alamarBlue. All three had a significant impact on color development and percent reduction of the dye. For the highest concentration of cells (3 × 108 CFU/ml) (Fig. (Fig.1a),1a), a strong reduction of the dye was observed after 1 day of cellular incubation. Further incubation of the cells or incubation with the dye did not result in an appreciable increase in dye reduction values. Indeed, a decrease in the percent reduction was noted, most likely due to buffering agents reaching their maximum buffering efficiencies in the reagent mix (www.abdserotec.com/about/alamarblue). At the mid-range cellular levels (2.4 × 106 CFU/ml) (Fig. (Fig.1b),1b), detectable dye reduction occurred after 2 days. The reduction was substantial after day 4. At the lowest concentration of cells (9.6 × 104 CFU/ml) (Fig. (Fig.1c),1c), a noticeable change in dye reduction was observed after 9 days. While longer incubation with alamarBlue led to greater dye reduction with a given cell titer, as shown with each suspension at 6, 24, and 48 h, the 24-h reading was considered sufficient to give a clear indication of viability.Open in a separate windowFIG. 1.Optimization of alamarBlue conditions using 3 × 108 CFU/ml (a), 2.4 × 106 CFU/ml (b), and 9.6 × 104 CFU/ml (c) over 11 days. Following the addition of alamarBlue, readings were taken at 6, 24, and 48 h.Percent reduction of the dye was standardized to 10, 20, 40, and 60% for each concentration of cells (Table (Table1).1). These values serve as definitive indicators of metabolic activity and may be used for multiple applications, such as comparing the relative viabilities of strains, or the influence of media composition or environmental stress on M. avium subsp. paratuberculosis. In particular, we feel that this assay is suited to comparing the relative efficacies of multiple anti-M. avium subsp. paratuberculosis compounds and/or antibiotic resistance profiling in a high-throughput screening format. The success of this assay requires strict adherence to specific cell numbers, growth phases, and their equivalent incubation times.

TABLE 1.

Required time taken for M. avium subsp. paratuberculosis to reduce alamarBlue
M. avium subsp. paratuberculosis cell concn (CFU/ml)Time (h) required to reach indicated % reductiona
10
20
40
60
CellularalamarBlueCellularalamarBlueCellularalamarBlueCellularalamarBlue
3 × 10824624624242424
6 × 107246242424489624
1.2 × 10724249624962414448
2.4 × 106966964812048N/AN/A
4.8 × 105966144482164826448
9.6 × 10496621648>264N/AN/AN/A
Open in a separate windowaN/A, not applicable.Traditionally, detection of viability with the alamarBlue assay has been achieved on the basis of the pink-blue color change (1, 5, 9, 15). However, this method for M. avium subsp. paratuberculosis is also quantitative, as it determines the threshold percent reduction values required for visual color change of the medium (<12% blue, 12 to 18% purple, and >20% pink). Visually, all wells with calculated values below 12% after the addition of the dye were consistently blue, most probably due to insufficient metabolism needed to indicate viability. All wells which were purple were recorded as having percent reduction values of 12 to 18%. Wells with values over 20% were consistently pink, which is indicative of cellular metabolism. The time taken to indicate definitive viability varied significantly for each dilution of cells, ranging from 1 day plus 6 h with the dye (3 × 108 CFU/ml) to 9 days plus 48 h with the dye (9.6 × 104 CFU/ml), as seen in Table Table11.To demonstrate the application of the assay, it was used to assess the susceptibility of M. avium subsp. paratuberculosis to rifampin (rifampicin). Rifampin was prepared in dimethyl sulfoxide at double the maximum concentration required. The assay was carried out using 6 × 107 CFU/ml over a period of 4 days, plus a 24-h incubation with alamarBlue in supplemented MB broth containing 0.2% mycobactin J. The final antibiotic concentration ranged from 125 to 0.48 μg/ml. The MIC90 of rifampin for M. avium subsp. paratuberculosis was determined as 1.92 μg/ml, which correlates well with other studies (17) (Fig. (Fig.2).2). This result highlights the assay''s potential as a high-throughput screening platform for antibiotic resistance profiling and the identification of novel anti-M. avium subsp. paratuberculosis agents.Open in a separate windowFIG. 2.Determination of the MIC90 of rifampin against M. avium subsp. paratuberculosisGiven the association of M. avium subsp. paratuberculosis with both animal and human diseases, such rapid, quantitative, and inexpensive (approximately 10 euro cents per sample) viability assays will be very useful for rapid screening of anti-M. avium subsp. paratuberculosis compounds and antibiotic profiling.  相似文献   

3.
4.
Predator-prey relationships among prokaryotes have received little attention but are likely to be important determinants of the composition, structure, and dynamics of microbial communities. Many species of the soil-dwelling myxobacteria are predators of other microbes, but their predation range is poorly characterized. To better understand the predatory capabilities of myxobacteria in nature, we analyzed the predation performance of numerous Myxococcus isolates across 12 diverse species of bacteria. All predator isolates could utilize most potential prey species to effectively fuel colony expansion, although one species hindered predator swarming relative to a control treatment with no growth substrate. Predator strains varied significantly in their relative performance across prey types, but most variation in predatory performance was determined by prey type, with Gram-negative prey species supporting more Myxococcus growth than Gram-positive species. There was evidence for specialized predator performance in some predator-prey combinations. Such specialization may reduce resource competition among sympatric strains in natural habitats. The broad prey range of the Myxococcus genus coupled with its ubiquity in the soil suggests that myxobacteria are likely to have very important ecological and evolutionary effects on many species of soil prokaryotes.Predation plays a major role in shaping both the ecology and evolution of biological communities. The population and evolutionary dynamics of predators and their prey are often tightly coupled and can greatly influence the dynamics of other organisms as well (1). Predation has been invoked as a major cause of diversity in ecosystems (11, 12). For example, predators may mediate coexistence between superior and inferior competitors (2, 13), and differential trajectories of predator-prey coevolution can lead to divergence between separate populations (70).Predation has been investigated extensively in higher organisms but relatively little among prokaryotes. Predation between prokaryotes is one of the most ancient forms of predation (27), and it has been proposed that this process may have been the origin of eukaryotic cells (16). Prokaryotes are key players in primary biomass production (44) and global nutrient cycling (22), and predation of some prokaryotes by others is likely to significantly affect these processes. Most studies of predatory prokaryotes have focused on Bdellovibrionaceae species (e.g., see references 51, 55, and 67). These small deltaproteobacteria prey on other Gram-negative cells, using flagella to swim rapidly until they collide with a prey cell. After collision, the predator cells then enter the periplasmic space of the prey cell, consume the host cell from within, elongate, and divide into new cells that are released upon host cell lysis (41). Although often described as predatory, the Bdellovibrionaceae may also be considered to be parasitic, as they typically depend (apart from host-independent strains that have been observed [60]) on the infection and death of their host for their reproduction (47).In this study, we examined predation among the myxobacteria, which are also deltaproteobacteria but constitute a monophyletic clade divergent from the Bdellovibrionaceae (17). Myxobacteria are found in most terrestrial soils and in many aquatic environments as well (17, 53, 74). Many myxobacteria, including the model species Myxococcus xanthus, exhibit several complex social traits, including fruiting body formation and spore formation (14, 18, 34, 62, 71), cooperative swarming with two motility systems (64, 87), and group (or “wolf pack”) predation on both bacteria and fungi (4, 5, 8, 9, 15, 50). Using representatives of the genus Myxococcus, we tested for both intra- and interspecific variation in myxobacterial predatory performance across a broad range of prey types. Moreover, we examined whether prey vary substantially in the degree to which they support predatory growth by the myxobacteria and whether patterns of variation in predator performance are constant or variable across prey environments. The latter outcome may reflect adaptive specialization and help to maintain diversity in natural populations (57, 59).Although closely related to the Bdellovibrionaceae (both are deltaproteobacteria), myxobacteria employ a highly divergent mode of predation. Myxobacteria use gliding motility (64) to search the soil matrix for prey and produce a wide range of antibiotics and lytic compounds that kill and decompose prey cells and break down complex polymers, thereby releasing substrates for growth (66). Myxobacterial predation is cooperative both in its “searching” component (6, 31, 82; for details on cooperative swarming, see reference 64) and in its “handling” component (10, 29, 31, 32), in which secreted enzymes turn prey cells into consumable growth substrates (56, 83). There is evidence that M. xanthus employs chemotaxis-like genes in its attack on prey cells (5) and that predation is stimulated by close contact with prey cells (48).Recent studies have revealed great genetic and phenotypic diversity within natural populations of M. xanthus, on both global (79) and local (down to centimeter) scales (78). Phenotypic diversity includes variation in social compatibility (24, 81), the density and nutrient thresholds triggering development (33, 38), developmental timing (38), motility rates and patterns (80), and secondary metabolite production (40). Although natural populations are spatially structured and both genetic diversity and population differentiation decrease with spatial scale (79), substantial genetic diversity is present even among centimeter-scale isolates (78). No study has yet systematically investigated quantitative natural variation in myxobacterial predation phenotypes across a large number of predator genotypes.Given the previous discovery of large variation in all examined phenotypes, even among genetically extremely similar strains, we anticipated extensive predatory variation as well. Using a phylogenetically broad range of prey, we compared and contrasted the predatory performance of 16 natural M. xanthus isolates, sampled from global to local scales, as well as the commonly studied laboratory reference strain DK1622 and representatives of three additional Myxococcus species: M. flavescens (86), M. macrosporus (42), and M. virescens (63) (Table (Table1).1). In particular, we measured myxobacterial swarm expansion rates on prey lawns spread on buffered agar (31, 50) and on control plates with no nutrients or with prehydrolyzed growth substrate.

TABLE 1.

List of myxobacteria used, with geographical origin
Organism abbreviation used in textSpeciesStrainGeographic originReference(s)
A9Myxococcus xanthusA9Tübingen, Germany78
A23Myxococcus xanthusA23Tübingen, Germany78
A30Myxococcus xanthusA30Tübingen, Germany78
A41Myxococcus xanthusA41Tübingen, Germany78
A46Myxococcus xanthusA46Tübingen, Germany78
A47Myxococcus xanthusA47Tübingen, Germany78
A75Myxococcus xanthusA75Tübingen, Germany78
A85Myxococcus xanthusA85Tübingen, Germany78
TVMyxococcus xanthusTvärminneTvärminne, Finland79
PAKMyxococcus xanthusPaklenicaPaklenica, Croatia79
MADMyxococcus xanthusMadeira 1Madeira, Portugal79
WARMyxococcus xanthusWarwick 1Warwick, UK79
TORMyxococcus xanthusToronto 1Toronto, Ontario, Canada79
SUL2Myxococcus xanthusSulawesi 2Sulawesi, Indonesia79
KALMyxococcus xanthusKalalauKalalau, HI79
DAVMyxococcus xanthusDavis 1ADavis, CA79
GJV1Myxococcus xanthusGJV 1Unknown35, 72
MXFL1Myxococcus flavescensMx fl1Unknown65
MXV2Myxococcus virescensMx v2Unknown65
CCM8Myxococcus macrosporusCc m8Unknown65
Open in a separate window  相似文献   

5.
6.
7.
8.
9.
A 30-probe assay was developed for simultaneous classification of Listeria monocytogenes isolates by lineage (I to IV), major serogroup (4b, 1/2b, 1/2a, and 1/2c), and epidemic clone (EC) type (ECI, ECIa, ECII, and ECIII). The assay was designed to facilitate rapid strain characterization and the integration of subtype data into risk-based inspection programs.Listeria monocytogenes is a facultative intracellular pathogen that can cause serious invasive illness (listeriosis) in humans and other animals. L. monocytogenes is responsible for over 25% of food-borne-disease-related deaths attributable to known pathogens and is a leading cause of food recalls due to microbial adulteration (12, 21). However, not all L. monocytogenes subtypes contribute equally to human illness, and substantial differences in the ecologies and virulence attributes of different L. monocytogenes subtypes have been identified (9, 13, 14, 23, 24, 33, 35, 36). Among the four major evolutionary lineages of L. monocytogenes, only lineages I and II are commonly isolated from contaminated food and human listeriosis patients (19, 27, 29, 33). Lineage I strains are overrepresented among human listeriosis isolates, particularly those associated with epidemic outbreaks, whereas lineage II strains are overrepresented in foods and the environment (13, 14, 24). Lineage III strains account for approximately 1% of human listeriosis cases but are common among animal listeriosis isolates and appear to be a host-adapted group that is poorly adapted to food-processing environments (6, 34-36). The ecological and virulence attributes of lineage IV are poorly understood, as this lineage is rare and was only recently described based on a small number of strains (19, 26, 29, 33).L. monocytogenes is differentiated into 13 serotypes; however, four major serogroups (4b, 1/2b, 1/2a, and 1/2c) from within lineages I and II account for more than 98% of human and food isolates (16, 31). Serogroups refer to evolutionary complexes typified by a predominant serotype but which include very rare serotypes that represent minor evolutionary variants (7, 9, 33). Phylogenetic analyses have indicated that rare serotypes may have evolved recently, or even multiple times, from one of the major serotypes (9), and numerous molecular methods fail to discriminate minor serotypes as independent groups (1, 4, 7, 9, 18, 22, 33, 38, 39). Serotyping is one of the most common methods for L. monocytogenes subtyping, and serogroup classifications are a useful component of strain characterization because ecotype divisions appear largely congruent with serogroup distinctions (16, 34). Serogroup 4b strains are of particular public health concern because contamination with these strains appears to increase the probability that a ready-to-eat (RTE) food will be implicated in listeriosis (16, 28). Serogroup 4b strains account for approximately 40% of sporadic listeriosis and also are responsible for the majority of listeriosis outbreaks despite being relatively rare contaminants of food products (9, 13, 17, 30, 34). In addition, serogroup 4b strains are associated with more severe clinical presentations and higher mortality rates than other serogroups (11, 16, 20, 31, 34). Serogroups 1/2a and 1/2b are overrepresented among food isolates but also contribute significantly to human listeriosis, whereas serogroup 1/2c rarely causes human illness and may pose a lower risk of listeriosis for humans (16). Serogroup-specific differences in association with human listeriosis are consistent with the prevalence of virulence-attenuating mutations in inlA within these serogroups (32, 34); however, a number of additional factors likely contribute to these differences.Four previously described epidemic clones (ECs; ECI, ECIa, ECII, and ECIII) of L. monocytogenes have been implicated in numerous listeriosis outbreaks and have contributed significantly to sporadic illness (15, 34). ECI, ECIa, and ECII are distinct groups within serogroup 4b that were each responsible for repeated outbreaks of listeriosis in the United States and Europe. ECIII is a lineage II clone of serotype 1/2a that persisted in the same processing facility for more than a decade prior to causing a multistate outbreak linked to contaminated turkey (15, 25). While there has been speculation that epidemic clones possess unique adaptations that explain their frequent involvement in listeriosis outbreaks (9, 34, 37), it is not clear that epidemic clones are more virulent than other strains with the same serotype. However, contamination of RTE food with EC strains would be cause for increased concern due to the previous involvement of these clones in major outbreaks of listeriosis (16).As a result of the L. monocytogenes subtype-specific differences in ecology, virulence, and association with human illness, molecular subtyping technologies have the potential to inform assessments of relative risk and to improve risk-based inspection programs. The objective of the present study was to develop a single assay for rapid and accurate classification of L. monocytogenes isolates by lineage, major serogroup, and epidemic clone in order to facilitate strain characterization and the integration of subtype data into inspection programs that are based on assessment of relative risk.A database of more than 5.3 Mb of comparative DNA sequences from 238 L. monocytogenes isolates (9, 33-35) was scanned for single nucleotide polymorphisms that could be used to differentiate lineages, major serogroups, and epidemic clones via a targeted multilocus genotyping (TMLGT) approach. The acronym TMLGT is used to distinguish this approach from previously published multilocus genotyping (MLGT) assays that were lineage specific and designed for haplotype discrimination (9, 33). To provide for simultaneous interrogation of the selected polymorphisms via TMLGT, six genomic regions (Table (Table1)1) were coamplified in a multiplex PCR. While the previous MLGT assays were based on three lineage-specific multiplexes and required prior identification of lineage identity, TMLGT was designed to target variation across all of the lineages simultaneously and is based on a unique set of amplicons. PCR was performed in 50-μl volumes with 1× High Fidelity PCR buffer (Invitrogen Life Technologies), 2 mM MgSO4, 100 μM deoxynucleoside triphosphate (dNTP), 300 nM primer, 1.5 U Platinum Taq high-fidelity DNA polymerase (Invitrogen Life Technologies), and 100 ng of genomic DNA. PCR consisted of an initial denaturation of 90 s at 96°C, followed by 40 cycles of 30 s at 94°C, 30 s at 50°C, and 90 s at 68°C. Amplification products were purified using Montage PCR cleanup filter plates (Millipore) and served as a template for allele-specific primer extension (ASPE) reactions utilizing subtype-specific probes.

TABLE 1.

Primers used in multiplex amplification for the TMLGT assay
AmpliconPositionaGene(s)PrimerSequence (5′-3′)b
INLa455381-456505inlAinl2-a1GTCCTTGATAGTCTACTG
inl2-a2ACCAAATTAGTAATCTAGCAC
INLb457726-458752inlBinl-f1dGAATTRTTTAGYCAAGAATGT
inlb-rCTACCGGRACTTTATAGTAYG
LMO325116-326096lmo0298-lmo0300lmo-a1AAGGCTTACAAGATGGCT
lmo1a-1rAAATAATAYGTGATACCGAC
VGCa205366-206622plcA, hlyplca-fCTCATCGTATCRTGTGTACC
hly-rTCTGGAAGGTCKTGTAGGTTC
VGCb208447-209465mplra_mpl-fGTGGAYAGAACTCATAAAGG
ra_mpl-rACTCCCTCCTYGTGATASGCT
VGCc209728-211239actAvgc1a-2fTTCMATRCCAGCAGAACG
vgc1a-2rGCAGACCTAATAGCAATGTTG
Open in a separate windowaCorresponding nucleotide positions in the complete genome sequence of L. monocytogenes strain EGD-e (GenBank accession number NC_003210).bSee IUPAC codes for definition of degenerate bases.ASPE was performed in multiplex reactions including 30 probes, with each lineage (I to IV), major serogroup (4b, 1/2b, 1/2a, and 1/2c), and epidemic clone (ECI, ECIa, ECII, and ECIII) targeted by two different probes (Table (Table2).2). In addition, positive-control probes were included to confirm the presence of each amplicon in the multiplex PCR. As serogroups and epidemic clones are nested within a particular lineage, probes for these groups were designed to be specific within the appropriate lineage and values for these probes were evaluated only for isolates of the appropriate lineage. For example, serogroup 1/2a probes were evaluated only for isolates that were positive for lineage II probes. ASPE probes were designed with a unique 5′ sequence tag specific to individual sets of xMAP fluorescent polystyrene microspheres (Luminex Corporation) used to sort extension products. Extension and hybridization reactions were performed as described previously (9) except microspheres were twice pelleted by centrifugation (4 min at 2,250 × g) and resuspended in 75 μl 1× TM buffer prior to being pelleted and resuspended in 100 μl 1× TM buffer containing 2 μg/ml streptavidin-R-phycoerythrin (Invitrogen Life Technologies). Samples were incubated for 15 min at 37°C prior to detecting the microsphere complexes with a Luminex 100 flow cytometer (Luminex Corporation). The median fluorescence intensity (MFI) from biotinylated extension products attached to 100 microspheres was measured for each probe. The average MFI from three template-free control samples was also determined and subtracted from the raw MFI of each sample to account for background fluorescence. Probe performance was initially evaluated via the index of discrimination (ID) as described by Ducey et al. (9), and probes with ID values less than 2.0 were redesigned.

TABLE 2.

TMLGT probes and probe performance data
ProbebTarget (n)cProbe sequencedIDeSensitivity (%)Specificity (%)
VGCb-21Lineage I (506)AATCCTTTCTTTAATCTCAAATCAgcggaagcttgggaagcggtc7.3100100
VGCa-94Lineage ICTTTCTATCTTTCTACTCAATAATcaacccgatgttcttcctgtc51.7100100
VGCc-8Lineage II (340)AATCCTTTTACATTCATTACTTACattagctgattcgctttcct14.1100100
INLb-51Lineage IITCATTTCAATCAATCATCAACAATagcgccaataaagctggc21.9100100
VGCb-19Lineage III (50)TCAATCAATTACTTACTCAAATACccgctattaaaatgtactcca31.0100100
VGCb-29Lineage IIIAATCTTACTACAAATCCTTTCTTTggtataccgctattaaaatgt45.1100100
LMO-17Lineage IV (10)CTTTAATCCTTTATCACTTTATCAgaaccaaacaatgttattggt11.8100100
VGCa-27Lineage IVCTTTTCAAATCAATACTCAACTTTttaacgacggtaacgtgccac58.3100100
INLb-84Serogroup 4b (213)TCAACTAACTAATCATCTATCAATggtaaaaatatgcgaatattg9.7100100
INLb-85Serogroup 4bATACTACATCATAATCAAACATCActcgtgaacaagctttcc5.5100100
INLb-16Serogroup 1/2b (293)AATCAATCTTCATTCAAATCATCAggtaaaaatatgcgtatctta11.7100100
INLb-100Serogroup 1/2bCTATCTTTAAACTACAAATCTAACgtgaataagctatcggtctat13.0100100
LMO-42Serogroup 1/2a (268)CTATCTTCATATTTCACTATAAACtggcgttgctgrctaagtttg6.6100100
VGCb-40Serogroup 1/2aCTTTCTACATTATTCACAACATTAaatcaagcsgctcatatgaag10.410098.6
LMO-9Serogroup 1/2c (72)TAATCTTCTATATCAACATCTTACtttactggtgaaatggcg13.5100100
VGCb-5Serogroup 1/2cCAATTCAAATCACAATAATCAATCaagattacgaatcgcttccac20.898.6100
LMO-10ECI (111)ATCATACATACATACAAATCTACAatgattaaaagtcagggaaag19.0100100
LMO-28ECICTACAAACAAACAAACATTATCAAaatcgaggcttacgaacgt23.7100100
VGCc-80ECIa (44)CTAACTAACAATAATCTAACTAACactacaacgaaaacagcgc10.7100100
VGCa-35ECIaCAATTTCATCATTCATTCATTTCAgttacttttatgtcgagt9.2100100
LMO-12ECII (35)TACACTTTCTTTCTTTCTTTCTTTataccgattatttggacggtt3.8100100
LMO-30ECIITTACCTTTATACCTTTCTTTTTACgacttgtagcagttgatttcaa7.5100100
VGCc-45ECIII (10)TCATTTCACAATTCAATTACTCAActcttatttgcttttgttggtc21.110099.4
INLa-3ECIIITACACTTTATCAAATCTTACAATCgagcttaatgaaaatcagcta17.010099.4
INLa-1INLa controlCTTTAATCTCAATCAATACAAATCagaagtggaagctgggaaNAaNANA
INLb-13INLb controlCAATAAACTATACTTCTTCACTAAtgcacctaaacctccgacNANANA
LMO-88LMO controlTTACTTCACTTTCTATTTACAATCccgtttccttatgccacaNANANA
VGCa-23VGCa controlTTCAATCATTCAAATCTCAACTTTcaagycctaagacgccaatcgNANANA
VGCb-25VGCb controlCTTTTCAATTACTTCAAATCTTCAgcatgcgttagttcatgrccaNANANA
VGCc-82VGCc controlTACATACACTAATAACATACTCATgactgcatgctagaatctaagNANANA
Open in a separate windowaNA, not applicable for positive amplicon control probes.bLuminex microsphere sets (Luminex Corporation) used for hybridization reactions are indicated following the hyphen.cn, number of isolates representing the target subtype among the 906 tested isolates.dThe 5′ sequence tag portions of extension probes are capitalized. See IUPAC codes for definitions of degenerate bases.eID, index of discrimination.Validation of the TMLGT assay was performed using 906 L. monocytogenes isolates for which the lineage, major serogroup, and epidemic clone type had been determined independently (see Table S1 in the supplemental material). A subset of 92 isolates, including at least five isolates from each lineage, serogroup, and epidemic clone type, was used to evaluate the discriminatory power of subtype-specific probes and the repeatability of the assay (see Table S1). Two independent runs of the 30-probe TMLGT assay produced identical results for these 92 isolates. In addition, genotypes matched expectations for all isolate/probe combinations, and the fluorescence intensities for positive genotypes (those targeted by a particular probe) were 3.8 to 58.3 (mean, 18.5) times as high as background values for isolates with negative genotypes (those not targeted by a particular probe) (Table (Table2).2). The performances of individual probes also were assessed in terms of sensitivity and specificity, where sensitivity is defined as the percentage of positive samples that produced positive results and specificity indicates the percentage of negative samples that produce negative results (5). Based on results from all 906 isolates analyzed by TMLGT, probe sensitivity was at least 98.6% and 23 of the 24 subtype-specific probes exhibited 100% sensitivity (Table (Table2).2). The specificities for all probes were also greater than 98.6%, and 21 of the 24 subtype-specific probes exhibited 100% specificity (Table (Table22).All but three of the 906 isolates in the validation panel were fully and accurately typed relative to lineage, serogroup, and epidemic clone by using the TMLGT assay (typeability, 99.9%; accuracy of isolate assignment, 99.8%). One of the lineage II isolates, NRRL B-33880, could not be assigned to a serogroup based on the TMLGT results because this isolate was positive for one of the serogroup 1/2a probes (VGCb-40) and one of the serogroup 1/2c probes (LMO-9). This isolate was previously identified as a member of serogroup 1/2c based on mapping lineage-specific MLGT data onto a multilocus phylogeny (34) but produced a serogroup 1/2a-specific banding pattern (data not shown) with the multiplex PCR assay described by Doumith et al. (7). Similar strains, including the common laboratory strain EGD-e, were found to have genomes that are more similar to serogroup 1/2c strains than to strains from the 1/2a serogroup (8, 33) and likely represent intermediates in the evolution of the 1/2c clade from 1/2a ancestors. There is a poor correlation between genomic and antigenic variation for such isolates (34), consistent with the ambiguous results produced by application of the TMLGT assay to NRRL B-33880. The two other problematic isolates, NRRL B-33555 and NRRL B-33559, were accurately identified based on TMLGT data as lineage II isolates from the 1/2a serogroup. However, these two isolates were positive for both ECIII-specific probes in the TMLGT assay but have lineage-specific MLGT haplotypes (Lm2.46), indicating that they are representatives of a sister group closely related to ECIII (33).In 2005, the Food Safety and Inspection Service (FSIS) implemented an approach to inspection that includes consideration of relative risk in order to determine L. monocytogenes sampling frequency among establishments that produce certain RTE products. This approach incorporates information on production volume, outgrowth potential in the product, steps taken to prevent postlethality contamination, and FSIS sampling history. However, L. monocytogenes subtype-specific variation in ecology and virulence indicates that information on the lineage, major serogroup, and epidemic clone identities of isolates could be used to inform assessments of relative risk and to improve inspection programs that are based on consideration of risk. Several PCR-based methods have been described for differentiation of various combinations of these subgroups (1-3, 5, 7, 10, 35, 37); however, these approaches have focused on a single subgroup or a smaller set of subgroups than is differentiated by TMLGT analysis. Although we previously developed a set of three MLGT assays that can be used to differentiate all of the major serogroups and epidemic clones of L. monocytogenes (9, 33, 34), those assays did not include probes for lineage discrimination and require identification of the lineage prior to application of one of three unique sets of probes. In addition, the MLGT assays were designed to maximize strain discrimination, as opposed to subgroup identification, and require the use of at least twice as many probes as is needed for TMLGT analysis. MLGT data analysis is also more complicated than analysis of TMLGT data, and serogroup or epidemic clone type identification via MLGT requires phylogenetic analyses to place novel haplotypes within an established phylogenetic framework.In the present study, we developed the first assay for simultaneous discrimination of the four lineages, the four major serogroups, and the four previously described epidemic clones of L. monocytogenes. The assay includes multiple markers for each of these subtype probes as well as control probes to ensure that negative probe data were not the result of amplification failure, providing a high degree of internal validation required for use in inspection programs that consider risk in making sampling decisions. In addition, the utility of the assay has been validated with a large and diverse panel of 906 isolates, including 567 isolates from FSIS surveillance of RTE products and processing facilities (see Table S1 in the supplemental material). Data produced by the TMLGT assay are amenable to high-throughput analysis, and a simple spreadsheet utility has been developed to semiautomate subtype identifications and to alert investigators to potentially conflicting probe data (available upon request). In addition to having a potential application in inspection programs, the TMLGT assay provides a rapid and accurate means of characterizing L. monocytogenes isolates from different environments, which would facilitate pathogen tracking and improve understanding of L. monocytogenes ecology.   相似文献   

10.
The biological, serological, and genomic characterization of a paramyxovirus recently isolated from rockhopper penguins (Eudyptes chrysocome) suggested that this virus represented a new avian paramyxovirus (APMV) group, APMV10. This penguin virus resembled other APMVs by electron microscopy; however, its viral hemagglutination (HA) activity was not inhibited by antisera against any of the nine defined APMV serotypes. In addition, antiserum generated against this penguin virus did not inhibit the HA of representative viruses of the other APMV serotypes. Sequence data produced using random priming methods revealed a genomic structure typical of APMV. Phylogenetic evaluation of coding regions revealed that amino acid sequences of all six proteins were most closely related to APMV2 and APMV8. The calculation of evolutionary distances among proteins and distances at the nucleotide level confirmed that APMV2, APMV8, and the penguin virus all were sufficiently divergent from each other to be considered different serotypes. We propose that this isolate, named APMV10/penguin/Falkland Islands/324/2007, be the prototype virus for APMV10. Because of the known problems associated with serology, such as antiserum cross-reactivity and one-way immunogenicity, in addition to the reliance on the immune response to a single protein, the hemagglutinin-neuraminidase, as the sole base for viral classification, we suggest the need for new classification guidelines that incorporate genome sequence comparisons.Viruses from the Paramyxoviridae family have caused disease in humans and animals for centuries. Over the last 40 years, many paramyxoviruses isolated from animals and people have been newly described (16, 17, 22, 29, 31, 32, 36, 42, 44, 46, 49, 58, 59, 62-64). Viruses from this family are pleomorphic, enveloped, single-stranded, nonsegmented, negative-sense RNA viruses that demonstrate serological cross-reactivity with other paramyxoviruses related to them (30, 46). The subfamily Paramyxovirinae is divided into five genera: Respirovirus, Morbillivirus, Rubulavirus, Henipavirus, and Avulavirus (30). The Avulavirus genus contains nine distinct avian paramyxovirus (APMV) serotypes (Table (Table1),1), and information on the discovery of each has been reported elsewhere (4, 6, 7, 9, 12, 34, 41, 50, 51, 60, 68).

TABLE 1.

Characteristics of prototype viruses APMV1 to APMV9 and the penguin virus
StrainHostDiseaseDistributionFusion cleavagecGI accession no.
APMV1/Newcastle disease virus>250 speciesHigh mortalityWorldwideGRRQKRF45511218
InapparentWorldwideGGRQGRLa11545722
APMV2/Chicken/CA/Yucaipa/1956Turkey, chickens, psittacines, rails, passerinesDecrease in egg production and respiratory diseaseWorldwideDKPASRF169144527
APMV3/Turkey/WI/1968TurkeyMild respiratory disease and moderate egg decreaseWorldwidePRPSGRLa209484147
APMV3/Parakeet/Netherlands/449/1975Psittacines, passerines, flamingosNeurological, enteric, and respiratory diseaseWorldwideARPRGRLa171472314
APMV4/Duck/Hong Kong/D3/1975Duck, geese, chickensNone knownWorldwideVDIQPRF210076708
APMV5/Budgerigar/Japan/Kunitachi/1974Budgerigars, lorikeetsHigh mortality, enteric diseaseJapan, United Kingdom, AustraliaGKRKKRFa290563909
APMV6/Duck/Hong Kong/199/1977Ducks, geese, turkeysMild respiratory disease and increased mortality in turkeysWorldwidePAPEPRLb15081567
APMV7/Dove/TN/4/1975Pigeons, doves, turkeysMild respiratory disease in turkeysUnited States, England, JapanTLPSSRF224979458
APMV8/Goose/DE/1053/1976Ducks, geeseNone knownUnited States, JapanTYPQTRLa226343050
APMV9/Duck/NY/22/1978DucksNone knownWorldwideRIREGRIa217068693
APMV10/Penguin/Falkland Islands/324/2007Rockhopper penguinsNone KnownFalkland IslandsDKPSQRIa300432141
Open in a separate windowaRequires the addition of an exogenous protease.bProtease requirement depends on the isolate examined.cPutative.Six of these serotypes were classified in the latter half of the 1970s, when the most reliable assay available to classify paramyxoviruses was the hemagglutination inhibition (HI) assay (61). However, there are multiple problems associated with the use of serology, including the inability to classify some APMVs by comparing them to the sera of the nine defined APMVs alone (2, 8). In addition, one-way antigenicity and cross-reactivity between different serotypes have been documented for many years (4, 5, 14, 25, 29, 33, 34, 41, 51, 52, 60). The ability of APMVs, like other viruses, to show antigenic drift as it evolves over time (37, 43, 54) and the wide use and availability of precise molecular methods, such as PCR and genome sequencing, demonstrate the need for a more practical classification system.The genetic diversity of APMVs is still largely unexplored, as hundreds of avian species have never been surveyed for the presence of viruses that do not cause significant signs of disease or are not economically important. The emergence of H5N1 highly pathogenic avian influenza (HPAI) virus as the cause of the largest outbreak of a virulent virus in poultry in the past 100 years has spurred the development of surveillance programs to better understand the ecology of avian influenza (AI) viruses in aquatic birds around the globe, and in some instances it has provided opportunities for observing other viruses in wild bird populations (15, 53). In 2007, as part of a seabird health surveillance program in the Falkland Islands (Islas Malvinas), oral and cloacal swabs and serum were collected from rockhopper penguins (Eudyptes chrysocome) and environmental/fecal swab pools were collected from other seabirds.While AI virus has not yet been isolated from penguins in the sub-Antarctic and Antarctic areas, there have been two reports of serum antibodies positive to H7 and H10 from the Adélie species (11, 40). Rare isolations of APMV1, both virulent (45) and of low virulence (8), have been reported from Antarctic penguins. Sera positive for APMV1 and AMPV2 have also been reported (21, 24, 38, 40, 53). Since 1981, paramyxoviruses have been isolated from king penguins (Aptenodytes patagonicus), royal penguins (Eudyptes schlegeli), and Adélie penguins (Pygoscelis adeliae) from Antarctica and little blue penguins (Eudyptula minor) from Australia that cannot be identified as belonging to APMV1 to -9 and have not yet been classified (8, 11, 38-40). The morphology, biological and genomic characteristics, and antigenic relatedness of an APMV recently isolated from multiple penguin colonies on the Falkland Islands are reported here. Evidence that the virus belongs to a new serotype (APMV10) and a demonstration of the advantages of a whole genome system of analysis based on random sequencing followed by comparison of genetic distances are presented. Only after all APMVs are reported and classified will epidemiological information be known as to how the viruses are moving and spreading as the birds travel and interact with other avian species.  相似文献   

11.
The three-dimensional structure of adeno-associated virus (AAV) serotype 6 (AAV6) was determined using cryo-electron microscopy and image reconstruction and using X-ray crystallography to 9.7- and 3.0-Å resolution, respectively. The AAV6 capsid contains a highly conserved, eight-stranded (βB to βI) β-barrel core and large loop regions between the strands which form the capsid surface, as observed in other AAV structures. The loops show conformational variation compared to other AAVs, consistent with previous reports that amino acids in these loop regions are involved in differentiating AAV receptor binding, transduction efficiency, and antigenicity properties. Toward structure-function annotation of AAV6 with respect to its unique dual glycan receptor (heparan sulfate and sialic acid) utilization for cellular recognition, and its enhanced lung epithelial transduction compared to other AAVs, the capsid structure was compared to that of AAV1, which binds sialic acid and differs from AAV6 in only 6 out of 736 amino acids. Five of these residues are located at or close to the icosahedral 3-fold axis of the capsid, thereby identifying this region as imparting important functions, such as receptor attachment and transduction phenotype. Two of the five observed amino acids are located in the capsid interior, suggesting that differential AAV infection properties are also controlled by postentry intracellular events. Density ordered inside the capsid, under the 3-fold axis in a previously reported, conserved AAV DNA binding pocket, was modeled as a nucleotide and a base, further implicating this capsid region in AAV genome recognition and/or stabilization.Adeno-associated viruses (AAVs) are nonpathogenic single-stranded DNA (ssDNA) parvoviruses that belong to the Dependovirus genus and require helper viruses, such as Adenovirus or Herpesvirus, for lytic infection (4, 8, 22, 67). These viruses package a genome of ∼4.7 kb inside an icosahedral capsid (∼260 Å in diameter) with a triangulation number equal to 1 assembled from a total of 60 copies of their overlapping capsid viral protein (VP) 1 (VP1), VP2, and VP3 in a predicted ratio of 1:1:8/10 (10). The VPs are encoded from a cap open reading frame (ORF). VP3 is 61 kDa and constitutes 90% of the capsid''s protein composition. The less abundant VPs, VP1 (87 kDa) and VP2 (73 kDa), share the same C-terminal amino acid sequence with VP3 but have additional N-terminal sequences. A rep ORF codes for four overlapping proteins required for replication and DNA packaging.To date, more than 100 AAV isolates have been identified (21). Among the human and nonhuman primate AAVs isolated, 12 serotypes (AAV serotype 1 [AAV1] to AAV12) have been described and are classified into six phylogenetic clades on the basis of their VP sequences and antigenic reactivities, with AAV4 and AAV5 considered to be clonal isolates (21). AAV1 and AAV6, which represent clade A, differ by only 6 out of 736 VP1 amino acids (5 amino acids within VP3) and are antigenically cross-reactive. Other clade representatives include AAV2 (clade B), AAV2-AAV3 hybrid (clade C), AAV7 (clade D), AAV8 (clade E), and AAV9 (clade F) (21).The AAVs are under development as clinical gene delivery vectors (e.g., see references 5, 9, 12, 13, 24, 25, 53, and 61), with AAV2, the prototype member of the genus, being the most extensively studied serotype for this application. AAV2 has been successfully used to treat several disorders, but its broad tissue tropism makes it less effective for tissue-specific applications and the prevalence of preexisting neutralizing antibodies in the human population (11, 43) limits its utilization, especially when readministration is required to achieve a therapeutic outcome. Efforts have thus focused on characterizing the capsid-associated tissue tropism and transduction properties conferred by the capsid of representative serotypes of other clades (21). Outcomes of these studies include the observation that AAV1 and AAV6, for example, transduce liver, muscle, and airway epithelial cells more efficiently (e.g., up to 200-fold) than AAV2 (27, 28, 30). In addition, the six residues (Table (Table1)1) that differ between the VPs of AAV1 and AAV6 (a natural recombinant of AAV1 and AAV2 [56]) confer functional disparity between these two viruses. For example, AAV6 shows ∼3-fold higher lung cell epithelium transduction than AAV1 (27), and AAV1 and AAV6 bind terminally sialylated proteoglycans as their primary receptor, whereas AAV6 additionally binds to heparan sulfate (HS) proteoglycans with moderate affinity (70, 71). Therefore, a comparison of the AAV1 and AAV6 serotypes and, in particular, their capsid structures can help pinpoint the capsid regions that confer differences in cellular recognition and tissue transduction.

TABLE 1.

Amino acid differences between AAV1 and AAV6 and their reported mutants
AAVAmino acid at positiona:
Glycan targetbReference
129418531532584598642
AAV1LEEDFANS70
AAV1-E/KLEKDFANHS+ (and S)c70
AAV6FDKDLVHHS and S70
AAV6.1FDEDLVHHS (and S)c40, 70
AAV6.2LDKDLVHHS (and S)c40, 70
AAV6R2LDEDLVHHS (and S)c40
HAE1LEEDLVN(HS and S)d39
HAE2LDKDLVN(HS and S)d39
shH10FDKNLVNHS (and S-inde)33
Open in a separate windowaMutant residues in boldface have an AAV6 parental original; those underlined have an AAV1 parental origin.bS, sialic acid; HS, heparan sulfate; HS+, HS positive.cThe sialic acid binding phenotypes of these mutants were not discussed in the respective publications but are assumed to be still present.dThe glycan targets for these mutants were not discussed in this publication; thus, the phenotypes indicated are assumed.eThis mutant is sialic acid independent (S-ind) for cellular transduction.The structures of AAV1 to AAV5 and AAV8 have been determined by X-ray crystallography and/or cryo-electron microscopy and image reconstruction (cryo-EM) (23, 36, 47, 52, 66, 73; unpublished data), and preliminary characterization of crystals has also been reported for AAV1, AAV5, AAV7, and AAV9 (15, 45, 46, 55). The capsid VP structures contain a conserved eight-stranded (βB to βI) β-barrel core and large loop regions between the strands that form the capsid surface. The capsid surface is characterized by depressions at the icosahedral 2-fold axes of symmetry, finger-like projections surrounding the 3-fold axes, and canyon-like depressions surrounding the 5-fold axes. A total of nine variable regions (VRs; VRI to VRIX) were defined when the two most disparate structures, AAV2 and AAV4, were compared (23). The VRs contain amino acids that contribute to slight differences in surface topologies and distinct functional phenotypes, such as in receptor binding, transduction efficiency, and antigenic reactivity (10, 23, 37, 47).The structure of virus-like particles (VLPs) of AAV6, produced in a baculovirus/Sf9 insect cell expression system, has been determined by two highly complementary approaches, cryo-EM and X-ray crystallography. The AAV6 VP structure contains the general features already described for the AAVs and has conformational differences in the VRs compared to the VRs of other AAVs. The 9.7-Å-resolution cryoreconstructed structure enabled the localization of the C-α positions of five of the six amino acids that differ between highly homologous AAV6 and AAV1 but did not provide information on the positions of the side chains or their orientations. The X-ray crystal structure determined to 3.0-Å resolution enabled us to precisely map the atomic positions of these five residues at or close to the icosahedral 3-fold axes of the capsid. Reported mutagenesis and biochemical studies had functionally annotated the six residues differing between AAV1 and AAV6 with respect to their roles in receptor attachment and differential cellular transduction. Their disposition identifies the 3-fold capsid region as playing essential roles in AAV infection.  相似文献   

12.
13.
14.
15.
Recent studies indicate that sexual transmission of human immunodeficiency virus type 1 (HIV-1) generally results from productive infection by only one virus, a finding attributable to the mucosal barrier. Surprisingly, a recent study of injection drug users (IDUs) from St. Petersburg, Russia, also found most subjects to be acutely infected by a single virus. Here, we show by single-genome amplification and sequencing in a different IDU cohort that 60% of IDU subjects were infected by more than one virus, including one subject who was acutely infected by at least 16 viruses. Multivariant transmission was more common in IDUs than in heterosexuals (60% versus 19%; odds ratio, 6.14; 95% confidence interval [CI], 1.37 to 31.27; P = 0.008). These findings highlight the diversity in HIV-1 infection risks among different IDU cohorts and the challenges faced by vaccines in protecting against this mode of infection.Elucidation of virus-host interactions during and immediately following the transmission event is one of the great challenges and opportunities in human immunodeficiency virus (HIV)/AIDS prevention research (14-16, 31, 34, 45). Recent innovations involving single-genome amplification (SGA), direct amplicon sequencing, and phylogenetic inference based on a model of random virus evolution (18-20, 43) have allowed for the identification of transmitted/founder viruses that actually cross from donor to recipient, leading to productive HIV type 1 (HIV-1) infection. Our laboratory and others have made the surprising finding that HIV-1 transmission results from productive infection by a single transmitted/founder virus (or virally infected cell) in ∼80% of HIV-infected heterosexuals and in ∼60% of HIV-infected men who have sex with men (MSM) (1, 13, 18, 24). These studies thus provided a precise quantitative estimate for the long-recognized genetic bottleneck in HIV-1 transmission (6, 11-13, 17, 25, 28, 30, 35, 38, 42, 47-49) and a plausible explanation for the low acquisition rate per coital act and for graded infection risks associated with different exposure routes and behaviors (15, 36).In contrast to sexual transmission of HIV-1, virus transmission resulting from injection drug use has received relatively little attention (2, 3, 29, 42) despite the fact that injection drug use-associated transmission accounts for as many as 10% of new infections globally (26, 46). We hypothesized that SGA strategies developed for identifying transmitted/founder viruses following mucosal acquisition are applicable to deciphering transmission events following intravenous inoculation and that, due to the absence of a mucosal barrier, injection drug users (IDUs) exhibit a higher frequency of multiple-variant transmission and a wider range in numbers of transmitted viruses than do acutely infected heterosexual subjects. We obtained evidence in support of these hypotheses from the simian immunodeficiency virus (SIV)-Indian rhesus macaque infection model, where we showed that discrete low-diversity viral lineages emanating from single or multiple transmitted/founder viruses could be identified following intravenous inoculation and that the rectal mucosal barrier to infection was 2,000- to 20,000-fold greater than with intravenous inoculation (19). However, we also recognized potentially important differences between virus transmission in Indian rhesus macaques and virus transmission in humans that could complicate an IDU acquisition study. For example, in the SIV macaque model, the virus inocula can be well characterized genetically and the route and timing of virus exposure in relation to plasma sampling precisely defined, whereas in IDUs, the virus inoculum is generally undefined and the timing of virus infection only approximated based on clinical history and seroconversion testing (8). In addition, IDUs may have additional routes of potential virus acquisition due to concomitant sexual activity. Finally, there is a paucity of IDU cohorts for whom incident infection is monitored sufficiently frequently and clinical samples are collected often enough to allow for the identification and enumeration of transmitted/founder viruses. To address these special challenges, we proposed a pilot study of 10 IDU subjects designed to determine with 95% confidence if the proportion of multivariant transmissions in IDUs was more than 2-fold greater than the 20% frequency established for heterosexual transmission (1, 13, 18, 24). A secondary objective of the study was to determine whether the range in numbers of transmitted/founder viruses in IDUs exceeded the 1-to-6 range observed in heterosexuals (1, 13, 18, 24). To ensure comparability among the studies, we employed SGA-direct amplicon sequencing approaches, statistical methods, and power calculations identical to those that we had used previously to enumerate transmitted/founder viruses in heterosexual and MSM cohorts (1, 13, 18, 20, 24).We first surveyed investigators representing acute-infection cohorts in the United States, Canada, Russia, and China; only one cohort—the Montreal Primary HIV Infection Cohort (41)—had IDU clinical samples and clinical data available for study. The Montreal cohort of subjects with acute and early-stage HIV-1 infection was established in 1996 and recruits subjects from both academic and private medical centers throughout the city. Injection drug use is an important contributing factor to Montreal''s HIV burden, with IDUs comprising approximately 20% of the city''s AIDS cases and 35% of the cohort (21, 40, 41). A large proportion of Montreal''s IDUs use injection cocaine, with 50 to 69% of subjects reporting cocaine as their injection drug of choice (4, 5, 9, 22, 23).Subjects with documented serological evidence of recent HIV-1 infection and a concurrent history of injection drug use were selected for study. These individuals had few or no reported risk factors for sexual HIV-1 acquisition. Clinical history and laboratory tests of HIV-1 viremia and antibody seroconversion were used to determine the Fiebig clinical stage (8) and to estimate the date of infection (Table (Table1).1). One subject was determined to be in Fiebig stage III, one subject was in Fiebig stage IV, five subjects were in Fiebig stage V, and three subjects were in Fiebig stage VI. We performed SGA-direct amplicon sequencing on stored plasma samples and obtained a total of 391 3′ half-genomes (median, 25 per subject; range, 19 to 167). Nine of these sequences contained large deletions or were G-to-A hypermutated and were excluded from subsequent analysis. Sequences were aligned, visually inspected using the Highlighter tool (www.hiv.lanl.gov/content/sequence/HIGHLIGHT/highlighter.html), and analyzed by neighbor-joining (NJ) phylogenetic-tree construction. A composite NJ tree of full-length gp160 env sequences from all 10 subjects (Fig. (Fig.1A)1A) revealed distinct patient-specific monophyletic lineages, each with high bootstrap support and separated from the others by a mean genetic distance of 10.79% (median, 11.29%; range, 3.00 to 13.42%). Maximum within-patient env gene diversity ranged from 0.23% to 3.34% (Table (Table1).1). Four subjects displayed distinctly lower within-patient maximum env diversities (0.23 to 0.49%) than the other six subjects (1.48% to 3.34%). The lower maximum env diversities in the former group are consistent with infection either by a single virus or by multiple closely related viruses, while the higher diversities can be explained only by transmission of more than one virus based on empirical observations (1, 13, 18, 24) and mathematical modeling (18, 20).Open in a separate windowFIG. 1.NJ trees and Highlighter plots of HIV-1 gp160 env sequences. (A) Composite tree of 382 gp160 env sequences from all study subjects. The numerals at the nodes indicate bootstrap values for which statistical support exceeded 70%. (B) Subject ACT54869022 sequences suggest productive infection by a single virus (V1). (C) Subject HDNDRPI032 sequences suggest productive infection by as many as three viruses. (D) Subject HDNDRPI001 sequences suggest productive infection by at least five viruses with extensive interlineage recombination. Sequences are color coded to indicate viral progeny from distinct transmitted/founder viruses. Recombinant virus sequences are depicted in black. Methods for SGA, sequencing, model analysis, Highlighter plotting, and identification of transmitted/founder virus lineages are described elsewhere (18, 20, 24, 44). The horizontal scale bars represent genetic distance. nt, nucleotide.

TABLE 1.

Subject demographics and HIV-1 envelope analysis results
Subject identifierAge (yr)SexaFiebig stageEstimated no. of days postinfectionbCD4 countPlasma viral load (log)No. of SGA ampliconsDiversity of env genes (%)c
No. of transmitted/ founder viruses
MeanInterquartile rangeMaximumdModel predictionePhylogenetic estimatef
HDNDRPI03447MIII292407.881631.070.553.34>116
HDNDRPI02918FIV484404.34290.160.150.4911
HTM38524MV624065.37220.120.080.2711
CQLDR0342MV66NDg5.01210.080.080.2311
HDNDRPI00136MV286905.94250.900.631.91>15
HTM31939MV685204.43250.770.461.54>13
HDNDRPI03237MV731,0403.53191.482.993.34>13
ACTDM58020839MVI933874.53301.170.972.64>13
ACT5486902228MVI687233.43270.070.040.2411
PSL02446MVI823404.46210.820.631.57>13
Open in a separate windowaM, male; F, female.bNumbers of days postinfection were estimated on the basis of serological markers, clinical symptoms, or a history of a high-risk behavior leading to virus exposure.cDiversity measurements determined by PAUP* analysis.dThe model prediction of the maximum achievable env diversity 100 days after transmission is 0.60% (95% CI, 0.54 to 0.68%). Diversity values exceeding this range imply transmission and productive infection by more than one virus. Diversity values less than 0.54% can be explained by transmission of one virus or of multiple closely related viruses (18).eModel described in Keele et al. (18).fMinimum estimate of transmitted/founder viruses.gND, not determined.An example of productive clinical infection by a single virus is shown in phylogenetic tree and Highlighter plots from subject ACT54869022 (Fig. (Fig.1B).1B). A similar phylogenetic pattern of single-variant transmission was found in 4 of 10 IDU subjects (Table (Table1).1). Examples of multivariant transmission are shown for subject HDNDRPI032, for whom there was evidence of infection by 3 transmitted/founder viruses (Fig. (Fig.1C)1C) and for subject HDNDRPI001, for whom there was evidence of infection by at least 5 transmitted/founder viruses (Fig. (Fig.1D).1D). One IDU subject, HDNDRPI034, had evidence of multivariant transmission to an extent not previously seen in any of 225 subjects who acquired their infection by mucosal routes (1, 13, 18, 24) or in any of 13 IDUs, as recently reported by Masharsky and colleagues (29). We greatly extended the depth of our analysis in this subject to include 163 3′ half-genome sequences in order to increase the sensitivity of detection of low-frequency viral variants. Power calculations indicated that a sample size of 163 sequences gave us a >95% probability of sampling minor variants comprising as little as 2% of the virus population. By this approach, we found evidence of productive infection by at least 16 genetically distinct viruses (Fig. (Fig.2).2). Fourteen of these could be identified unambiguously based on the presence of discrete low-diversity viral lineages, each consisting of between 2 and 48 sequences. Two additional unique viral sequences with long branch lengths (3F8 and G10) exhibited diversity that was sufficiently great to indicate a distinct transmission event as opposed to divergence from other transmitted/founder lineages (see the legend to Fig. Fig.2).2). It is possible that still other unique sequences from this subject also represented transmitted/founder viruses, but we could not demonstrate this formally. We also could not determine if all 16 (or more) transmission events resulted from a single intravenous inoculation or from a series of inoculations separated by hours or days; however, it is likely that all transmitted viruses in this subject resulted from exposure to plasma from a single infected individual, since the maximum env diversity was only 3.34% (Fig. (Fig.1A).1A). It is also likely that transmission occurred within a brief window of time, since the period from transmission to the end of Fiebig stage III is typically only about 25 days (95% CI, 22 to 37 days) (18, 20) and the diversity observed in all transmitted/founder viral lineages in subject HDNDRPI034 was exceedingly low, consistent with model predictions for subjects with very recent infections (18, 20).Open in a separate windowFIG. 2.NJ tree and Highlighter plot of HIV-1 3′ half-genome sequences from subject HDNDRPI034. Sequences emanating from 16 transmitted/founder viruses are color coded. Fourteen transmitted/founder viral lineages comprised of 2 or more identical or nearly identical sequences could be readily distinguished from recombinant sequences (depicted in black), which invariably appeared as unique sequences containing interspersed segments shared with other transmitted/founder virus lineages. The two sequences with the longest branch lengths (3F8 and G10) were interpreted to represent rare progeny of discrete transmitted/founder viruses because their unique polymorphisms far exceeded the maximum diversity estimated to occur in the first 30 days of infection (0.22%; CI, 0.15 to 0.31%) (18) and far exceeded the diversity observed within the other transmitted/founder virus lineages. The horizontal scale bar represents genetic distance.Lastly, we compared the multiplicity of HIV-1 transmission in the Montreal IDU subjects with that of non-IDU subjects for whom identical SGA methods had been employed. In this combined-cohort analysis, we found the frequency of multiple-variant transmission in heterosexuals to be 19% (34 of 175) and in MSM 38% (19 of 50) (Table (Table2)2) (24). The current study was powered to detect a >2-fold difference in multivariant transmission between IDUs and heterosexual subjects; in fact, we observed a 3-fold-higher frequency of multiple-variant transmission in Montreal IDUs (6 of 10 subjects [60%]) than in heterosexuals (odds ratio, 6.14; 95% CI, 1.37 to 31.27; Fisher exact test, P = 0.008) and a 1.5-fold-higher frequency in Montreal IDUs than in MSM (odds ratio, 2.41; 95% CI, 0.50 to 13.20; P = 0.294, not significant). In addition, we found that the range of numbers of transmitted/founder viruses was greater in IDUs (range, 1 to 16 viruses; median, 3) than in either heterosexuals (range, 1 to 6 viruses; median, 1) or MSM (range, 1 to 10 viruses; median, 1). The finding of larger numbers of transmitted/founder viruses in IDUs was not simply the result of more intensive sampling, since the numbers of sequences analyzed in all studies were comparable. Moreover, it is notable that in studies reported elsewhere, we sampled as many as 239 sequences by SGA or as many as 500,000 sequences by 454 pyrosequencing from four acutely infected MSM subjects and in each case found evidence of productive clinical infection by only a single virus (24; W. Fischer, B. Keele, G. Shaw, and B. Korber, unpublished). These results thus suggest that IDUs may be infected by more viruses and by a greater range of viruses than is the case following mucosal transmission. On this count, our findings differ from those reported by Masharsky and coworkers for an IDU cohort from St. Petersburg, Russia (29). Their study found a low frequency of multiple virus transmissions (31%), not significantly different from that of acutely infected heterosexuals, and a low number of transmitted/founder viruses (range, 1 to 3 viruses; median, 1). Because the SGA methods employed in both studies were identical, the numbers of sequences analyzed per subject were comparable (median of 25 sequences in Montreal versus 33 in St. Petersburg), and because the discriminating power of the SGA-direct sequencing method was sufficient to distinguish transmitted/founder viruses differing by as few as 3 nucleotides, or <0.1% of nucleotides (Fig. (Fig.2,2, compare lineages V4 and V5), it is unlikely that differences in the genetic diversity of HIV-1 in the two IDU populations explain the differences in findings between the two studies. Instead, we suspect that the explanation lies in the small cohort sizes (10 versus 13 subjects) and the particular risk behaviors of the IDUs in each cohort. The Russian cohort is heavily weighted toward heroine use, whereas the Montreal cohort is weighted toward injection cocaine use, the latter being associated with more frequent drug administration and the attendant infection risks of needle sharing (4).

TABLE 2.

Multiplicity of HIV-1 infection in IDU, heterosexual, and MSM subjects
CohortReferenceVirus subtypeTotal no. of subjectsSingle-variant transmission
Multiple-variant transmission
P valueOdds ratio95% CIMedianRange
No. of subjects% of totalNo. of subjects% of total
HeterosexualsKeele et al. (18)B796582.301417.7011-4
Abrahams et al. (1)C695478.301521.7011-5
Haaland et al. (13)A or C272281.50518.5011-6
Total17514180.603419.400.008a6.141.37-31.2711-6
MSMKeele et al. (18)B221359.10940.9011-6
Li et al. (24)B281864.301035.7011-10
Total503162.001938.000.294b2.410.50-13.2011-10
IDUsBarB10440.00660.0031-16
Open in a separate windowaFisher''s exact test of multiple-variant transmission in heterosexuals versus in IDUs.bFisher''s exact test of multiple-variant transmission in MSM versus in IDUs.The results from the present study indicate that transmission of HIV-1 to IDUs can be associated with a high frequency of multiple-variant transmission and a broad range in the numbers of transmitted viruses. This wide variation in the multiplicity of HIV-1 infection in IDUs is likely due to the absence of a mucosal barrier to virus transmission (12, 19) and differences in the virus inocula (27, 29, 32, 39). The findings substantiate concerns raised in recent HIV-1 vaccine efficacy trials that different vaccine candidates may be more efficacious in preventing infection by some exposure routes than by others (7, 10, 33, 37). They further suggest that biological comparisons of molecularly cloned transmitted/founder viruses responsible for vaginal, rectal, penile, and intravenous infection could facilitate a mechanistic understanding of HIV-1 transmission and vaccine prevention (24, 44).  相似文献   

16.
A total of 210 Salmonella isolates, representing 64 different serovars, were isolated from imported seafood samples, and 55/210 isolates were found to be resistant to at least one antibiotic. Class 1 integrons from three multidrug-resistant Salmonella enterica strains (Salmonella enterica serovars Newport [strain 62], Typhimurium var. Copenhagen [strain 629], and Lansing [strain 803], originating from Hong Kong, the Philippines, and Taiwan, respectively) were characterized. Southern hybridization of plasmids isolated from these strains, using a class 1 integron probe, showed that trimethoprim-sulfamethoxazole and streptomycin resistance genes were located on a megaplasmid in strain 629. Our study indicates that imported seafood could be a reservoir for Salmonella isolates resistant to multiple antibiotics.Salmonella spp. are recognized as major food-borne pathogens of humans worldwide. In the United States, there are an estimated 800,000 to 4 million Salmonella infections annually, and approximately 500 of the cases are fatal (8, 26). A variety of foods have been implicated as vehicles transmitting salmonellosis to humans, including poultry, beef, pork, eggs, milk, cheese, fish, shellfish, fruits, juice, and vegetables (1, 4, 9, 12, 23). Previous studies by field laboratories of the U.S. Food and Drug Administration have shown the prevalences of Salmonella isolates in imported and domestic seafood as 7.2% and 1.3%, respectively (6, 11, 27).Mobile genetic elements, such as plasmids, transposons, and integrons, which disseminate antibiotic resistance genes by horizontal or vertical transfer, as part of either resistance plasmids or conjugative transposons, play an important role in the evolution and dissemination of multidrug resistance (2, 3, 10, 17). Salmonella genomic island 1 (SGI1), the first genomic island reported to contain an antibiotic resistance gene cluster, was identified in the multidrug-resistant Salmonella enterica serovar Typhimurium strain DT 104 (21).Most studies of the prevalence and characterization of antimicrobial resistance genes and integrons in Salmonella spp. have focused on strains from clinical and veterinary sources. However, little is known about the occurrence of SGI1 and its variants in Salmonella spp. isolated from seafood. We have screened a set of drug-resistant S. enterica strains from seafood belonging to 64 different serovars for SGI1 and class 1 integron conserved sequences (CS). We report the presence of a class I variant integron carrying the dfrXII and aadA2 genes on a megaplasmid in serovar Typhimurium var. Copenhagen and on the chromosome in Salmonella enterica serovar Lansing. We also found the variant class 1 integron carrying the dfrA1 and orfC genes in Salmonella enterica serovar Newport strains from seafood.A total of 210 Salmonella enterica strains isolated from seafood imported into the United States between 2000 and 2005 were identified and serotyped by the Pacific Regional Laboratory-Southwest of the FDA, Irvine, CA. The Salmonella strains represented 20 serogroups (Table (Table1)1) from various imported seafood items. The Salmonella strains were tested with 16 antibiotics (14) commonly used in either human or veterinary medicine on Mueller-Hinton agar (Difco Laboratories, Detroit, MI), using a disk diffusion method. The sensitivity and resistance were determined by the criteria of the Clinical and Laboratory Standards Institute (1999).

TABLE 1.

Salmonella serotypes isolated from imported foods
No. of strainsS. enterica serovar(s) or Salmonella group(s)
39Weltevreden
16Newport
13Saintpaul
10Senftenberg
8Lexington
7Virchow
6Enteritidis, Bareily
5Bovismorbificans, Brunei, Java, Hvittingfoss
4Paratyphi B var. Java, Thompson
3Aberdeen, Cubana, Stanley, Derby, Lansing
2Montevideo, Hadar, Agona, San Diego, Braenderup, Lanka, Salmonella enterica subsp. diarizonae, Oslo, Bareily variant, Salmonella monophasic group C2
1Ouakam, Cannstatt, Albany, Newport/Bardo, Adelaide, S. enterica subsp. diarizonae, Houten, Giza, Miami, Onderstepoof, Infantis, Salmonella monophasic group D1, Mbandaka, Salmonella monophasic group G2, Ohio, Rutgers, Salmonella monophasic group D2, Amsterdam, Salmonella enterica subsp. IV serotype 43:z4z23, Paratyphi B var. Java, Wentworth, Potsdam, Muenster var. 15+, 34+, Lexington var. 15+, Weltevreden var. 15+, S. enterica subsp. I, Madella, Alachua, London, Singapore, Uphill, Thielallee, Typhimurium var. Copenhagen
Open in a separate windowAll Salmonella strains that were resistant to three or four antibiotics and trimethoprim were screened by PCR for the presence of class 1 integrons, using the CSL1 and CSR1 primers (Table (Table2)2) (14). To confirm other antibiotic resistance genes, we used primers and PCR methods described previously (13, 14, 16). To identify SGI1 in multidrug-resistant strains, PCR was performed by using primers U7-L12/LJ-R1 and 104-RJ/104-D (Table (Table2),2), corresponding to the left and right junctions of SGI1 in the Salmonella chromosome, respectively (16). For a positive control, serovar Typhimurium DT104 strain DT7 (13) was used. As a negative control, Escherichia coli cells or DNA was used. A reagent blank included in each PCR contained distilled water instead of template DNA. For sequencing, the PCR-amplified integrons were purified and cloned into plasmid vector pCR2.1 (Invitrogen Corp., Carlsbad, CA). The clones were investigated for the presence of inserts by isolating the recombinant plasmid, which was confirmed by digestion with the restriction enzyme EcoRI. Sequencing of both strands was performed. DNA sequences were analyzed with Lasergene (DNASTAR, Inc., Madison, WI) software. Oligonucleotide primers and probes were purchased from MWG Biotech (High Point, NC).

TABLE 2.

Primer pairs for integron PCR and sequencing
PrimerSequence (5′-3′)LocationPCR product size (bp)
CSL1GGC ATC CAA GCA GCA AGC5′ CS
CSR1AAG CAG ACT TGA CCT GAT3′ CS
U7-L12ACA CCT TGA GCA GGG CAA AGthdF500
LJ-R1AGT TCT AAA GGT TCG TAG TCG
104-RJTGA CGA GCT GAA GCG AAT TGS044
104DACC AGG GCA AAA CTA CAC AGyidY
aadA2FTGT TGG TTA CTG TGG CCG TAaadA2380
aadA2RGCT GCG AGT TCC ATA GCT TC
Open in a separate windowPlasmid DNA was isolated using an alkaline lysis method with modifications described previously (19). Plasmids were separated by electrophoresis in 1× Tris-acetate-EDTA buffer at 64 V for 2 h on 1.0% agarose gels, stained with 40 μl of ethidium bromide (0.625 mg/ml) for visualization, and then transferred and cross-linked to positively charged nylon membranes (Roche, Indianapolis, IN). The resulting blots were hybridized at 65°C for 18 h with digoxigenin-labeled DNA probes (1.2-kb and 1.9-kb PCR-amplified products), using CSL1 and CSR1 primers specific for class 1 integrons (22).  相似文献   

17.
18.
Amino acid modifications of the Thermobifida fusca Cel9A-68 catalytic domain or carbohydrate binding module 3c (CBM3c) were combined to create enzymes with changed amino acids in both domains. Bacterial crystalline cellulose (BC) and swollen cellulose (SWC) assays of the expressed and purified enzymes showed that three combinations resulted in 150% and 200% increased activity, respectively, and also increased synergistic activity with other cellulases. Several other combinations resulted in drastically lowered activity, giving insight into the need for a balance between the binding in the catalytic cleft on either side of the cleavage site, as well as coordination between binding affinity for the catalytic domain and CBM3c. The same combinations of amino acid variants in the whole enzyme, Cel9A-90, did not increase BC or SWC activity but did have higher filter paper (FP) activity at 12% digestion.Cellulases catalyze the breakdown of cellulose into simple sugars that can be fermented to ethanol. The large amount of natural cellulose available is an exciting potential source of fuels and chemicals. However, the detailed molecular mechanisms of crystalline cellulose degradation by glycoside hydrolases are still not well understood and their low efficiency is a major barrier to cellulosic ethanol production.Thermobifida fusca is a filamentous soil bacterium that grows at 50°C in defined medium and can utilize cellulose as its sole carbon source. It is a major degrader of plant cell walls in heated organic materials such as compost piles and rotting hay and produces a set of enzymes that includes six different cellulases, three xylanases, a xyloglucanase, and two CBM33 binding proteins (12). Among them are three endocellulases, Cel9B, Cel6A, and Cel5A (7, 8), two exocellulases, Cel48A and Cel6B (6, 19), and a processive endocellulase, Cel9A (5, 7).T. fusca Cel9A-90 (Uniprot P26221 and YP_290232) is a multidomain enzyme consisting of a family 9 catalytic domain (CD) rigidly attached by a short linker to a family 3c cellulose binding module (CBM3c), followed by a fibronectin III-like domain and a family 2 CBM (CBM2). Cel9A-68 consists of the family 9 CD and CBM3c. The crystal structure of this species (Fig. (Fig.1)1) was determined by X-ray crystallography at 1.9 Å resolution (Protein Data Bank [PDB] code 4tf4) (15). Previous work has shown that E424 is the catalytic acid and D58 is the catalytic base (11, 20). H125 and Y206 were shown to play an important role in activity by forming a hydrogen bonding network with D58, an important supporting residue, D55, and Glc(−1)O1. Several enzymes with amino acid changes in subsites Glc(−1) to Glc(−4) had less than 20% activity on bacterial cellulose (BC) and markedly reduced processivity. It was proposed that these modifications disturb the coordination between product release and the subsequent binding of a cellulose chain into subsites Glc(−1) to Glc(−4) (11). Another variant enzyme with a deletion of a group of amino acids forming a block at the end of the catalytic cleft, Cel9A-68 Δ(T245-L251)R252K (DEL), showed slightly improved filter paper (FP) activity and binding to BC (20).Open in a separate windowFIG. 1.Crystal structure of Cel9A-68 (PDB code 4tf4) showing the locations of the variant residues, catalytic acid E424, catalytic base D58, hydrogen bonding network residues D55, H125, and Y206, and six glucose residues, Glc(−4) to Glc(+2). Part of the linker is visible in dark blue.The CBM3c domain is critical for hydrolysis and processivity. Cel9A-51, an enzyme with the family 9 CD and the linker but without CBM3c, had low activity on carboxymethyl cellulose (CMC), BC, and swollen cellulose (SWC) and showed no processivity (4). The role of CBM3c was investigated by mutagenesis, and one modified enzyme, R557A/E559A, had impaired activity on all of these substrates but normal binding and processivity (11). Variants with changes at five other CBM3c residues were found to slightly lower the activity of the modified enzymes, while Cel9A-68 enzymes containing either F476A, D513A, or I514H were found to have slightly increased binding and processivity (11) (see Table Table1).1). In the present work, CBM3c has been investigated more extensively to identify residues involved in substrate binding and processivity, understand the role of CBM3c more clearly, and study the coordination between the CD and CBM3c. An additional goal was to combine amino acid variants showing increased crystalline cellulose activity to see if this further increased activity. Finally, we have investigated whether the changes that improved the activity of Cel9A-68 also enhanced the activity of intact Cel9A-90.

TABLE 1.

Activities of Cel9A-68 CBM3c variant enzymes and CD variant enzymes used to create the double variants
EnzymeActivity (% of wild type) on:
% Processivity% BC bindingReference
CMCSWCBCFPa
Wild type10010010010010015This work
R378K9891103931392011
DELb981011011289620
F476A97105791001452111
D513A1001151211071192011
I514H104911121041102311
Y520A1087833a79871411
R557A1039860a9390This work
E559A869030a7094This work
R557A+E559A907515a751061511
Q561A1035651a7874This work
R563A977052a931292011
Open in a separate windowaThe target percent digestion could not be reached; activity was calculated using 1.5 μM enzyme.bDEL refers to deletion of T245 to L251 and R252K.  相似文献   

19.
20.
Angiostrongylus cantonensis is the most common cause of human eosinophilic meningitis. Humans become infected by ingesting food items contaminated with third-stage larvae that develop in mollusks. We report the development of a real-time PCR assay for the species-specific identification of A. cantonensis in mollusk tissue.Angiostrongylus cantonensis is the most common agent associated with eosinophilic meningitis in humans. Young adult worms develop in the brains of rodents and are carried to pulmonary arteries to reach sexual maturity. Eggs are laid in lung tissues, and first-stage (L1) larvae break into air spaces, migrate to the trachea, are swallowed, and are passed with rodent feces. The L1 larvae must infect mollusks to develop into third-stage (L3) larvae; L3 is the infective stage for rodents and other mammals. Humans become infected by ingesting raw produce contaminated with L3 larvae or infected raw or undercooked mollusks or paratenic hosts. The immature worms remain in the human brain, creating tissue damage and inflammation (2, 19, 21).A. cantonensis is endemic in Southeast Asia, parts of the Caribbean, and the Pacific Islands, including Hawaii (7, 12, 15-17). The worm has been detected in host animals in Louisiana (5, 14) and in one human patient from New Orleans (18), but it is currently unclear to what extent the nematode has spread into other U.S. states (8, 9). Ascertaining the geographic presence of the parasite is important to manage and prevent new cases of eosinophilic meningitis associated with ingestion of infective larvae (12, 18).Detection of A. cantonensis in mollusks can be performed by releasing the larvae from the tissue with pepsin digestion (11). However, that procedure requires access to living mollusks, which complicates analysis of large numbers of samples. After a recent outbreak of angiostrongyliasis in Hawaii (12), we developed a conventional PCR assay and applied it to survey the Hawaiian mollusk population using frozen tissue (20). That PCR assay, as well as morphological identification using pepsin digestion, can only identify the larvae on the superfamily level, so additional molecular work is required for species-specific classification. Here we describe a new real-time PCR assay that allows for a direct detection of A. cantonensis at the species level.The 18S rRNA gene is too conserved among nematode species to allow species-specific detection. The first and second internal transcribed spacers (ITS1 and ITS2) are comparatively more variable than the rRNA coding regions and have thus been used for differentiation of closely related species (1, 4, 6, 10, 22, 23). We PCR amplified and sequenced ITS1 from A. costaricensis (two laboratory strains from Costa Rica and Brazil), A. vasorum (from naturally infected hosts in United Kingdom), and A. cantonensis from three geographical regions (one laboratory strain from Japan plus nine environmental isolates from Hawaii and New Orleans, LA) to assess the variability of this potential PCR target. The oligonucleotide primers used were AngioF1674 (5′-GTCGTAACAAGGTATCTGTAGGTG-3′) and 58SR4 (5′-TAGCTGCGTTTTTCATCGATA-3′). The reaction mixtures contained 0.4 μM each primer and AmpliTaq Gold PCR master mix (Applied Biosystems, Foster City, CA) and were cycled 45 times at 94°C for 30 s, 65°C for 30 s, and 72°C for 1 min. PCR products were cloned into pCR2.1 vectors using the TOPO cloning technique (Invitrogen, Carlsbad, CA) and sequenced on both strands as described elsewhere (20).The sequence analysis revealed high interspecific and low intraspecific variability. A TaqMan assay targeting ITS1 was then designed using Primer Express version 2.3 (Applied Biosystems, Foster City, CA). The real-time PCR assay was performed in a 20-μl total volume containing Platinum qPCR Supermix (Invitrogen, Carlsbad, CA), 0.2 μM (each) primers AcanITS1F1 (5′-TTCATGGATGGCGAACTGATAG-3′) and AcanITS1R1 (5′-GCGCCCATTGAAACATTATACTT-3′), and 0.05 μM the TaqMan probe AcanITS1P1 (5′-6-carboxyfluorescein-ATCGCATATCTACTATACGCATGTGACACCTG-BHQ-3′). The standard cycling conditions for TaqMan assays were used (i.e., 40 cycles of 95°C for 15 s and 60°C for 1 min).We evaluated the real-time PCR assay with a set of 26 Parmarion martensi slugs from Hawaii. Seventeen slugs were positive for L3 larvae as determined by pepsin digestion, and nine slugs were negative. DNA was extracted from approximately 25 mg of tissue of each slug using the DNeasy tissue and blood DNA extraction kit (Qiagen, Inc., Valencia, CA). The real-time PCR performed on this set of samples returned an identical result to the morphological analysis. The real-time PCR amplified only DNA from A. cantonensis and did not react with DNA from other nematode species (Table (Table1).1). The detection limit of the assay was determined by serially diluting a recombinant plasmid containing the ITS1 sequence to less than 1 copy per μl of sample. The real-time PCR reliably detected down to 10 plasmid copies in the reaction.

TABLE 1.

Comparison of conventional and real-time PCR for detection of Angiostrongylus cantonensis in mollusks and nematode samples
Biological origin of DNA sampleGeographic originNo. of samples testedNo. of samples positive by:
18S rRNA-based conventional PCRITS1-based TaqMan PCR
Parmarion martensiHawaii1127583
Veronicella cubensisHawaii5023a22
Laevicaulis alteHawaii534
Achatina fulicaHawaii645
Other/unidentified mollusksHawaii1645
FlatwormsHawaii222
Slime from infected slugsHawaii1311
Pomacea insularumLouisiana3155
A. costaricensisBrazil, Costa Rica22b0
A. vasorumUnited Kingdom22b0
Other nematodescCDC collection1400
Total253121127
Open in a separate windowaThis number includes three samples positive by PCR but later identified as non-Angiostrongylus nematodes by DNA sequencing analysis of the amplicons (20). These three samples were negative in the real-time PCR assay.bThe conventional PCR detects other Angiostrongylus species besides A. cantonensis.cTwo stool samples containing Strongyloides worms, eight environmental samples containing unclassified free-living nematodes and one of each of the following parasitic nematodes: Dipetalonema sp., Toxocara cati, Dracunculus medinensis, and Ascaris lumbricoides.The real-time PCR assay was then used to analyze a larger set of naturally infected host animals from Hawaii, partly described elsewhere (13, 20), and Island Apple snails (Pomacea insularum) from New Orleans, LA. All samples had previously been characterized by the conventional PCR followed by DNA sequencing analysis (20).Table Table11 summarizes the PCR findings and highlights the enhanced performance of the real-time PCR in comparison to the conventional PCR. In addition, the real-time PCR assay was more practical to use since it did not require DNA sequence confirmation to rule out false positives.The findings from Island Apple snails from New Orleans infected with A. cantonensis concur with previous reports about the potential for angiostrongyliasis transmission in this area (5, 14). Another interesting finding was the positive PCR results in two samples of flatworms from Hawaii. Predatory flatworms that ingest infected mollusks are known to be paratenic hosts of A. cantonensis and have been suspected to be an important source of infection for humans in Japan because they hide in leafy vegetables (3).In conclusion, this real-time PCR assay can be a useful tool for environmental surveys of local wildlife to determine the geographic distribution of this reemerging human parasite.  相似文献   

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