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.
2.
3.
4.
5.
6.
The presence of Vibrio parahaemolyticus in 123 oyster samples collected from an estuary on the southern coast of Sao Paulo state, Brazil, was investigated. Of the 123 samples, 99.2% were positive with densities ranging from <3 to 105 most probable number (MPN)/g. Densities correlated significantly with water temperature (r = 0.48; P < 0.001) but not with salinity (r = −0.09; P = 0.34). The effect of harvest site on counts was not significant (P > 0.05). These data provide information for the assessment of exposure of V. parahaemolyticus in oysters at harvest.Infections caused by Vibrio parahaemolyticus have been reported in several countries (1, 3-5, 14, 15, 16, 17, 18, 19, 20, 22, 24, 26). Among other pathogenic features, V. parahaemolyticus strains produce a thermostable hemolysin, known as thermostable direct hemolysin (TDH), as well as TRH (a TDH-related hemolysin) (25, 29). However, not all strains are pathogenic, as less than 1% of food or environmental strains produce TDH or TRH (2, 7, 9, 10, 11).The most important vehicle for this microorganism is raw or partially cooked shellfish (8, 13, 25, 29). In this study, the densities of V. parahaemolyticus in oysters collected in six oyster bed sites in the estuary of Cananeia (25°S; 48°W) in the southern coastal area of Sao Paulo state, Brazil (Fig. (Fig.1)1) between May 2004 and June 2005 were determined using the most probable number (MPN) technique by the method of De Paola and Kaysner (12). Each sample consisted of 15 oysters, pooled in a plastic bag, and transported in a cold box to the laboratory located in the city of Sao Paulo, Brazil. The temperature during transportation did not exceed 13°C, and the travel time was around 5 h. In the laboratory, the oysters were kept under refrigeration (4 to 8°C) and analyzed within 24 h of collection. Oysters were cleaned and shucked by the method of Cook et al. (6). Identification of V. parahaemolyticus was based on traditional and API 20E strip biochemical tests (bioMérieux, France), using V. parahaemolyticus ATCC 17802 as the reference strain. The observed prevalence of this bacterium was high, as the microorganism was detected in 99.2% (122/123) of the samples and the densities varied between 0.78 and 5.04 log MPN/g.Open in a separate windowFIG. 1.Locations of oyster bed sites in the Cananeia estuary on the southern coast of Sao Paulo state, Brazil. (Courtesy of E. E. de Miranda and A. C. Coutinho [Embrapa Monitoramento por Satélite] [http://www.cdbrasil.cnpm.embrapa.br].)Strategies for the control of V. parahaemolyticus in oysters depend on understanding the seasonal and geographical distribution and the effects of environmental parameters on the growth of this pathogen. To verify the influence of salinity and temperature of seawater on the density of V. parahaemolyticus, samples of water (n = 123) were collected from the same depth of oyster beds using 250-ml plastic flasks. Salinity was determined using a salinometer (model RS10; Rosemount Analytical, Cedar Grove, NJ), and the temperature was determined at the time of collection using a digital thermometer (Hanna Instruments). The results are shown in Fig. Fig.22.Open in a separate windowFIG. 2.Total densities of Vibrio parahaemolyticus in oysters from the southern coast of Sao Paulo state, Brazil. Each bar or point represents the arithmetic mean of six sites, and each error bar represents the standard deviation.Total V. parahaemolyticus densities did not correlate significantly with water salinity, as determined by Pearson coefficient (r = −0.09; P = 0.34). However, the mean salinity varied significantly according to the sampling site and season (P < 0.05) (Table (Table1).1). The highest mean salinity (24.2 ppt) was detected at site 5 and was 1.4 times higher than at site 2 (17.3 ppt), the lowest mean salinity detected in this study.

TABLE 1.

Seasonal distribution of the total density of Vibrio parahaemolyticus in oysters, water temperature, and salinity in the southern coastal area of Sao Paulo state, Brazil
VariableSeasonNo. of samplesMeanaSDRange
Vibrio parahaemolyticusWinter352.44 A1.06<0.48-4.38
    density (log10Spring293.26 B1.171.04-5.04
    MPN/g)Summer243.47 B0.751.54-5.04
Fall353.48 B1.011.04-5.04
Temp (°C)Winter3520.1 A1.914.4-24.0
Spring2923.6 B1.820.0-26.0
Summer2426.7 C1.424.1-29.2
Fall3523.9 B2.220.6-28.3
Salinity (ppt)Winter3522.3 A4.512.2-29.8
Spring2920.2 AB4.411.2-29.4
Summer2418.2 B4.35.3-25.2
Fall3521.8 A3.98.7-28.2
Open in a separate windowaValues with different letters are significantly different (P < 0.05).The weak correlation between water salinity and V. parahaemolyticus densities in oysters suggests that salinity per se is a secondary factor for growth of this bacterium, as are turbidity and chlorophyll content in water (27, 30). These results agree with those obtained by Deepanjali et al. (9) and Martinez-Urtiga et al. (21), who did not find correlation between these two parameters. However, they are in contrast with the results reported by DePaola et al. (11), who observed correlation (P < 0.05) between salinity and total density of V. parahaemolyticus.The results of this study corroborate existing evidence (10, 11, 27, 30) indicating that the temperature of seawater has a significant correlation (r = 0.48; P < 0.001) on the densities of V. parahaemolyticus in oysters, but they are at odds with results reported by Deepanjali et al. (9), who observed no statistically significant correlation with seawater temperature. The temperature variations observed in the present study (15°C) were lower than those observed by DePaola et al. (22°C) (11) but higher than those reported by Deepanjali et al. (10°C) (9).The relationship between V. parahaemolyticus density and water temperature and salinity were analyzed by multiple linear regression. Results showed that salinity was not significant either for linear effects or for squared effects (P > 0.05). For temperature, while the parameter of linear effect was significant (P < 0.05), the squared effect was not (P > 0.05). Considering the goodness of fit of the model, the following linear regression described the density in oysters the best (Fig. (Fig.3):3): log10 MPN V. parahaemolyticus/g = −0.944 + (0.175 × temperature). The lack of model fitness test was not significant and was considered adequate to express the relationship between V. parahaemolyticus density and seawater temperature, in spite of the low R2 (0.23).Open in a separate windowFIG. 3.Goodness of fit regression model of V. parahaemolyticus density in oysters and water temperature.The effect of temperature was further summarized by rank correlation and the use of a smoothing technique (moving average) in which densities corresponding to temperatures within a range of 1°C were pooled to estimate an arithmetic mean of densities in successive intervals. The moving average was calculated using a length of three values. Although seawater temperature and V. parahaemolyticus densities were correlated in the present study, the mean densities reached a plateau at temperatures above 24°C and below 20°C (Fig. (Fig.4)4) where the density was not significantly influenced by temperature, consistent with observations reported also by DePaola et al. (11). Our findings could explain the lack of correlation among those parameters detected in tropical oysters by Deepanjali et al. (9) when the temperature varied from 25 to 35°C.Open in a separate windowFIG. 4.Relationship between the mean density of V. parahaemolyticus in oysters and seawater temperature in Cananeia estuary, Sao Paulo state, Brazil.The influence of the season of the year and site of collection on the mean densities was assessed by analysis of variance and Tukey''s test, when necessary. As shown in Table Table1,1, the V. parahaemolyticus densities were similar in the samples collected during spring, summer, and autumn but differed significantly (P < 0.05) in those collected during winter. Densities among samples collected during summer varied less compared to other seasons. Densities above 105 MPN/g were detected in six (4.9%) oyster samples (three samples during spring, two samples during summer, and one sample during autumn) collected when the temperature was higher than 24°C and the salinity was higher than 15 ppt. The effect of harvest site on densities was not significant (P > 0.05).Previous studies performed with oysters collected in the same region in Brazil have shown a low incidence of pathogenic V. parahaemolyticus (23, 28). Similar results were observed in the present study, as only one oyster sample (0.8%) and only one isolate among 2,243 isolates tested (0.044%) were Kanagawa and tdh positive. Besides the Kanagawa reaction (24), all strains have been tested for tlh, tdh, and trh genes using PCR (12). The pathogen-positive sample presented with a low density of V. parahaemolyticus (3 MPN/g) and was collected during winter, when the temperature was 21°C. Due to the low incidence of pathogenic strains in the samples, correlation between pathogenicity and water temperature or salinity could not be determined.This study indicates that the presence of V. parahaemolyticus in oysters cultivated in the southern coast of Sao Paulo state, Brazil, is high, but pathogenic strains are seldom detected. These results on the ecology and characteristics of V. parahaemolyticus are valuable for future risk assessments related to this pathogen in oysters at harvest.  相似文献   

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

TABLE 1.

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

TABLE 2.

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

10.
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   

11.
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   

12.
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   

13.
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   

14.
A fitness cost due to imbalanced replichores has been proposed to provoke chromosome rearrangements in Salmonella enterica serovars. To determine the impact of replichore imbalance on fitness, the relative fitness of isogenic Salmonella strains containing transposon-held duplications of various sizes and at various chromosomal locations was determined. Although duplication of certain genes influenced fitness, a replichore imbalance of up to 16° did not affect fitness.The bacterial chromosome is a dynamic molecule that can undergo various types of rearrangements, including inversions, translocations, and duplications. These rearrangements can alter gene order and change replichore length. Replichores are defined as the halves of the chromosome between the origin of replication and the terminus region in the vicinity of the dif site (4, 6, 10, 15). In most bacteria, both replichores are approximately the same length, with each comprising 180° around the circular chromosome. In this state, the replichores and DNA replication are balanced. Imbalance is introduced when one replichore becomes longer than the other. For example, if the replichores comprise 200° and 160° around the circular chromosome, the replichores are 20° imbalanced. Various studies over the years have investigated the effect of imbalanced DNA replication on fitness in Escherichia coli (8, 9, 16, 17). In a recent analysis, E. coli strains that contained asymmetrical interreplichore inversions were found to have growth defects when the imbalance was at least 50° (8). While these studies have demonstrated that imbalanced replichores can affect fitness, the approaches used to introduce the imbalance either do not occur or are extremely rare in nature.Duplications play important evolutionary roles because the increase in gene copy number can facilitate adaptation to certain growth conditions (20), as well as being a source for the evolution of new genes (3). Typically duplications occur at frequencies between 10−3 and 10−5 in unselected cultures (2) but are lost at rates of up to 1,000-fold more often if they do not provide a selective advantage (19). Duplications also result in replichore imbalance. However, the effect of duplications on fitness in relation to replichore balance has not been investigated.A major hurdle in studying the effects of duplications on fitness is that if the duplication is detrimental, haploid revertants will outgrow the parental strain containing the duplication. To circumvent this problem, we used a set of 11 isogenic Salmonella enterica strains with transposon-held duplications (5) (Fig. (Fig.11 and Table Table1).1). Culturing these strains in the presence of chloramphenicol selects for the duplication because if the duplication collapses, the transposon is lost and the cells become chloramphenicol sensitive. As the size of the duplications in these strains varies, so does the amount of introduced replichore imbalance, ranging from 5° to 23°. In addition, fitness effects due to the location of the duplication versus the size of the duplication were also discerned, as the collection includes duplications of similar sizes located in different regions of the chromosome.Open in a separate windowFIG. 1.Genetic map of S. enterica serovar Typhimurium LT2 showing genes used as endpoints in constructing transposon-held duplications of the regions between genes. Balanced replichores are indicated by the symmetry of the oriC-Ter axis. Duplications increase the length of one replichore relative to the other, imbalancing axis symmetry.

TABLE 1.

Properties of the S. enterica strains used in this study
StrainAliasGenotypeDuplication location (min)Duplication size (kbp)Replichore imbalance (°)Reference
MST1LT2Wild type1.911
MST3813SV4200Dup [trp248 MudP hisD9953]38-44332.111.55
MST3814SV3193Dup [hisH9962 MudP cysA1586]44-53399.713.75
MST3815SV4015Dup [cysA1586 MudP purG2149]53-56158.45.75
MST3816SV4193Dup [purG2149 MudP argA9001]56-64430.714.75
MST3817SV4194Dup [argA9000 MudP cysG1573]64-75484.616.35
MST3818SV1601Dup [cysG1573 MudP ilvA2642]75-85486.416.45
MST3819SV4195Dup [ilvA2648 MudP purA1881]85-95495.316.75
MST3820SV4142Dup [purA1881 MudP thr469]95-0248.78.85
MST3821SV1604Dup [thr469 MudP proA692]0-8367.112.65
MST3822SV1603Dup [proA692 MudQ purE2164]8-12230.28.15
MST3823SV1611Dup [purE2514 MudP purB1879]12-27723.223.35
MST1529TT11183srl-203::Tn10d(Cam)1
TYT4480rrfH::pCE36This work
MST5198rrfH::pCE36 srl-203::Tn10d(Cam)This work
Open in a separate windowThe relative fitness of these strains was investigated by measuring growth rates and by assaying growth in a mixed culture with an isogenic competitor strain. Single colony isolates streaked from frozen stocks were used to inoculate broth cultures. Growth was measured in E medium supplemented with 0.2% glucose (minimal medium), Luria-Bertani medium (LB), or LB supplemented with 1× E-salts and 0.2% glucose (LBEDO) (18). Competition assays were done in LB. Media were supplemented with chloramphenicol (20 mg/ml) to maintain the duplications, and the solid media used in competition experiments also contained 5-bromo-4-chloro-3-indolyl-β-d-galactopyranoside (X-Gal; 40 mg/ml) to differentiate between the duplication and competitor strains.In minimal medium, most of the strains had a generation time of approximately 40 min, except for MST3813, MST3823, and MST3819, which grew somewhat slower (Fig. (Fig.2).2). In LB, the average generation times of all of the duplication-bearing strains were longer than that of the wild type. When grown in LBEDO, all of the strains had generation times of 11 to 15 min. Observed differences in growth rate were not due to lower viability as determined by plate counts. There was no correlation between duplication size and generation time in any of the three media.Open in a separate windowFIG. 2.Generation times of strains as a function of duplication size. Strains were grown in minimal medium (♦), LB (▪), or LBEDO (▴) aerobically at 37°C. Error bars show standard deviations.Competition indices were determined from mixed cultures with the isogenic competitor strain MST5198 and either the wild-type strain or one of the duplication-bearing strains. The competitor strain contained an integrated plasmid encoding a promoterless lacZ gene (7) driven by the rrnH promoter and Tn10d(Cam) conferring chloramphenicol resistance integrated into the srl locus (Table (Table1).1). Strains were grown to saturation overnight, and 10−4 dilutions were used to inoculate mixed cultures. Samples taken at t = 0 (input) and subsequent time points (output) were diluted and spot plated in triplicate. The competition index (C.I.) was calculated as follows: C.I. = (no. output Lac CFU/no. output Lac+ CFU)/(no. input Lac CFU/no. input Lac+ CFU).Data are shown for 6 and 12 h, but readings were taken at time points of up to 1 week. There was no correlation between the duplication size and the C.I. (Fig. (Fig.3).3). Most of the duplication-bearing strains competed as well as or slightly better than the wild type against the competitor strain. Two exceptions were MST3819 and MST3823, which both competed significantly more poorly against the competitor strain than did the wild-type strain (Student''s t test P < 0.05). While these two strains contain the largest duplications, the size of the duplication in MST3819 was close to the size of the duplications in MST3817 and MST3818. Strains MST3817 and MST3818 were either similar or slightly better competitors than the wild type, indicating that the fitness defect in MST3819 is not due to replichore imbalance.Open in a separate windowFIG. 3.Competition indices of the wild-type and duplication-bearing strains after 6 (▪) and 12 h (░⃞) of growth in mixed cultures with MST5198 (n = 6). Strains are in order of increasing duplication size, with the locations of duplications indicated at the top. Indices statistically significantly different (P < 0.05) from that of the wild-type strain (MST1) are marked (*). Error bars show standard deviations.To confirm that the duplications in MST3819 and MST3823 were responsible for the observed growth defects, isolates of each strain with collapsed duplications were obtained by growing cultures of each strain without chloramphenicol and then screening for chloramphenicol-sensitive derivatives. The loss of the duplication restored wild-type growth to both strains. The frequencies of duplication collapse in cultures of MST3818 and MST3819 were also compared. Since these two strains have duplications of similar sizes, the collapse frequencies of the duplicated regions, and concomitant loss of chloramphenicol resistance, were expected to be similar. While the reversion frequency of MST3818 cells was 3.5 × 10−3 cells/generation, MST3819 cells reverted with a frequency of 2.8 × 10−2 cells/generation, resulting in 97 to 98% of MST3819 cells being chloramphenicol sensitive and haploid after reaching stationary phase.In conclusion, the results of this study show that a replichore imbalance of up to 16° introduced by transposon-held duplications in the chromosome does not have a measurable effect on the fitness of S. enterica. The duplicated regions in these strains are larger than the islands acquired via horizontal gene transfer, which have been purported to disrupt replichore balance sufficiently to promote chromosome rearrangements. A duplication that introduced a replichore imbalance of 23° did have a growth defect, but whether the defect is from replichore imbalance or the gene content of the duplication could not be distinguished. This study suggests that the disruption of replichore balance by acquisition of horizontally transferred genes is not the cause of chromosome rearrangements in host-specific S. enterica serovars as hypothesized (12-14).  相似文献   

15.
Aerobic growth conditions significantly influenced anaerobic succinate production in two-stage fermentation by Escherichia coli AFP111 with knockouts in rpoS, pflAB, ldhA, and ptsG genes. At a low cell growth rate limited by glucose, enzymes involved in the reductive arm of the tricarboxylic acid cycle and the glyoxylate shunt showed elevated activities, providing AFP111 with intracellular redox balance and increased succinic acid yield and productivity.Succinic acid is valued as one of the key basic chemicals used in the preparation of biodegradable polymers or as raw material for chemicals of the C4 family (8, 19). The fermentative production of succinic acid from renewable resources is environmentally acceptable and sustainable (3). A breakthrough in genetically engineering Escherichia coli (6, 7, 11, 18) for succinate production was the isolation of strain AFP111 (1, 4), a mutant of NZN111 with a spontaneous ptsG mutation (pflAB ldhA double mutant). The process involves a two-stage fermentation, with aerobic cell growth followed by anaerobic conditions for succinate production (16, 21, 22). The aerobically induced enzymes can maintain their activity during the anaerobic phase and significantly affect succinate fermentation (22, 23). Using the best transition time based on the activities of the key enzymes and other physiological states, a two-stage fermentation using the recombinant AFP111 strain harboring pTrc99A-pyc achieved a final succinic acid concentration and productivity of 99.2 g·liter−1 and 1.3 g·liter−1·h−1, respectively (21).Aerobic cell growth is essential for the subsequent anaerobic fermentation. However, few studies have focused on the regulation of aerobic cell growth. As a regulation method, gluconeogenic carbon sources were used instead of glucose for the aerobic growth of Escherichia coli NZN111 and the activities of enzymes that are favorable for the anaerobic synthesis of succinate were enhanced (23, 24). Unfortunately, a gluconeogenic carbon source (e.g., sodium acetate) might increase the osmotic pressure of culture media, which would be detrimental to succinate production (23). As another regulation method, a glucose feeding strategy controlling the glucose concentration at about 0.5 g·liter−1 up to 1 g·liter−1 was reported to prevent excessive formation of acetic acid (16).In this study, we investigated different glucose feeding strategies for the aerobic growth phase of the two-phase process for succinate production by E. coli AFP111. Specifically, we compared several growth rates by using glucose limitation in addition to maximum growth under conditions of excess glucose.E. coli AFP111 [F+ λ rpoS396(Am) rph-1 ΔpflAB::Cam ldhA::Kan ptsG] (4, 16), which was a kind gift from D. P. Clark (Southern Illinois University), was the only strain used in this study. Luria-Bertani (LB) medium (60 ml) was used for inoculum culture in 1,000-ml flasks, and 3 liters of chemically defined medium (13, 14) was used for two-stage culture in a 7-liter fermentor. Two-stage fermentations were divided into three types, based on the glucose feeding strategy used during the aerobic stage. For type I culture, the glucose concentration was maintained at about 20 g·liter−1 during aerobic cell growth. Type II and III cultures comprised a batch process and subsequent glucose-limited fed-batch process (Fig. (Fig.1).1). The batch process initially contained 13 g/liter of glucose. The fed-batch process began when the dry cell weight (DCW) reached about 6 g/liter, with type II and type III cultures using a 600 g/liter glucose feed to achieve cell growth rates of 0.15 h−1 and 0.07 h−1, respectively (10). When the DCW reached 12 g·liter−1, the aerobically grown cells were directly transferred to anaerobic conditions (Fig. (Fig.1).1). For the anaerobic process, oxygen-free CO2 was sparged at 0.5 liter·min−1, the pH was controlled between 6.4 and 6.8 with intermittent supplementation of solid magnesium carbonate hydroxide, and the glucose concentration was maintained at about 20 g·liter−1 by supplying glucose in an 800-g·liter−1 solution.Open in a separate windowFIG. 1.Concentrations of glucose (circles), DCW (triangles), and succinic acid (squares) in the three types of two-stage fermentation by AFP111. μ, growth rate.The optical density at 600 nm was used to monitor cell growth, and this value was correlated to DCW. The concentration of glucose was assayed with an enzyme electrode analyzer, and organic acids were quantified by high-performance liquid chromatography (HPLC). The intracellular concentrations of NADH and NAD+ were assayed with a cycling method (12). The activities of isocitrate lyase (ICL) (20), pyruvate kinase (PYK) (17), phosphoenolpyruvate (PEP) carboxykinase (PCK) (20, 23), PEP carboxylase (PPC) (23), and malate dehydrogenase (MDH) (23) were measured spectrophotometrically at the end of the aerobic phase and 12 h after the onset of the anaerobic phase.All three types of fermentations were terminated when the succinate concentration increased less than 1 g·liter−1 in 5 h. Type III fermentation was terminated at a final succinic acid concentration of 101.2 g·liter−1 and an anaerobic-phase productivity of 1.89 g·liter−1·h−1 (Fig. (Fig.1).1). Trace amounts of by-products (such as acetate, ethanol, and pyruvate) accumulated and did not follow any trend in the anaerobic phase (data not shown).At the end of the aerobic culture phase, the specific enzyme activities of PCK, PYK, and ICL in type III culture were 2.9, 2.5, and 11.4 times higher, respectively, than the activities in type I culture (Table (Table1)1) . This phenomenon is consistent with published reports that suggest that the expression of enzymes involved in anaplerotic metabolism and the glyoxylate shunt (5, 15) is elevated in E. coli grown under glucose-limited conditions. These enzymes maintained their activities in the subsequent anaerobic phase (Table (Table1)1) and would be central to succinate production (22, 23). The elevated levels of PCK and PPC would provide the reductive branch of the tricarboxylic acid (TCA) cycle with oxaloacetate (OAA) at a higher rate (9), thereby supplying both malate and citrate (Table (Table11).

TABLE 1.

Activities of enzymes at the end of the aerobic culture phase and 12 h after the onset of the anaerobic phase
Fermentation typeaStagebMean sp act of enzyme ± SD (U/mg protein)c
PCKPPCMDHPYKICL
IAerobic0.82 ± 0.050.22 ± 0.0521.97 ± 0.151,175 ± 11.380.12 ± 0.00
Anaerobic0.55 ± 0.020.19 ± 0.0018.27 ± 1.05978 ± 12.330.09 ± 0.00
IIAerobic1.46 ± 0.100.23 ± 0.0425.69 ± 0.372,053 ± 3.650.73 ± 0.03
Anaerobic1.09 ± 0.010.20 ± 0.0135.55 ± 0.781,430 ± 13.780.41 ± 0.02
IIIAerobic2.38 ± 0.110.16 ± 0.0023.5 ± 0.132,955 ± 8.771.37 ± 0.00
Anaerobic1.75 ± 0.030.21 ± 0.0143.8 ± 0.622,501 ± 10.151.02 ± 0.01
Open in a separate windowaFermentation types were mentioned in culture conditions section.b“Aerobic” represents the data obtained at the end of aerobic culture; “Anaerobic” represents those obtained 12 h after transition to anaerobic fermentation.cThe standard deviations (SD) were calculated from triplicate samples of the same run.The reductive branch of the TCA cycle consumes 4 mol of electrons to form 2 mol of succinate based on 1 mol of glucose (1, 4). Therefore, the conversion of glucose to succinate through the reductive arm of the TCA cycle alone will lead to an intracellular imbalance of reducing equivalents (2, 18). Fortunately, the glyoxylate shunt (2, 18, 22) is available to provide 10 mol of electrons by converting 1 mol of glucose to 1 mol of succinate and 2 mol of CO2 (22). In the case of the ptsG mutant strain AFP111, when the molar flux at the PEP branch point flowing to OAA versus flowing to pyruvate reaches a ratio of 5:2, the intracellular redox balance is satisfied and the maximum theoretical mass yield of 1.12 g·g−1 succinic acid is achieved (22). Based on the elevated activities of PCK, PYK, and ICL (Table (Table1),1), both pathways leading to succinate were enhanced after glucose-limited growth. The succinic acid yields of 1.03 to 1.07 g·g−1 in the two glucose-limited processes approached the maximum theoretical yield for AFP111 (22), and these yields were about two times greater than the yield in the type I fermentation (Table (Table22).

TABLE 2.

Succinic acid production during anaerobic fermentation phasea
Fermentation typeMean ± SD
Succinic acid (g·liter−1)Yield (g·g−1)Productivity (g·liter−1·h−1)Specific productivity at 12 h (mg·g−1·h−1)NADH at 12 h mmol·(g DCW)−1NADH/NAD+ ratio at 12 h
I35.0 ± 0.740.43 ± 0.050.98 ± 0.04105 ± 150.88 ± 0.070.55 ± 0.08
II74.3 ± 3.241.03 ± 0.011.32 ± 0.05160 ± 81.95 ± 0.111.05 ± 0.10
III101.2 ± 1.041.07 ± 0.021.89 ± 0.07227 ± 111.97 ± 0.151.27 ± 0.13
Open in a separate windowaThe data were calculated only for the anaerobic stage. The standard deviations (SD) were calculated from two independent two-stage fermentations.In addition to differences in succinic acid yields, the glucose-limited and type I fermentations each resulted in significantly different specific succinic acid productivities (Table (Table2).2). A specific succinic acid productivity of 227 mg·g−1·h−1 was obtained at 12 h in type III fermentation. Because two pathways are needed for succinate production due to redox constraints, and enzyme activities in both pathways were elevated by glucose limitation, the results suggest that operating with glucose limitation provides the cells with greater metabolic flexibility to achieve a redox balance. Furthermore, the results suggest that one or more of these enzymes are limiting succinate formation under batch conditions (type I fermentation). Considering the NADH/NAD+ assays (Table (Table2),2), the results would support the hypothesis that succinate production was limited by insufficient NADH (2, 18).In summary, our study presented an efficient method of aerobic cell cultivation for two-stage succinate fermentation by engineered E. coli. Since the physiological state of aerobically grown cells was essential for their subsequent anaerobic succinate fermentation, some other environmental and physiology factors in the aerobic growth phase may also play an important role in improving succinate production.  相似文献   

16.
17.
18.
Simian immunodeficiency virus (SIV) infection of natural-host species, such as sooty mangabeys (SMs), is characterized by a high level of viral replication and a low level of generalized immune activation, despite evidence of an adaptive immune response. Here the ability of SIV-infected SMs to mount neutralizing antibodies (Nab) against autologous virus was compared to that of human immunodeficiency virus type 1 (HIV-1) subtype C-infected subjects. While high levels of Nab were observed in HIV-1 infection, samples obtained at comparable time points from SM exhibited relatively low titers of autologous Nab. Nevertheless, SM plasma with higher Nab titers also contained elevated peripheral CD4+ T-cell levels, suggesting a potential immunologic benefit for SMs. These data indicate that AIDS resistance in these primates is not due to high Nab titers and raise the possibility that low levels of Nab might be an inherent feature of natural-host SIV infections.More than 40 species of African nonhuman primates (NHPs) naturally harbor CD4+-tropic lentiviruses that are collectively known as simian immunodeficiency viruses (SIVs) and represent the ancestors of the human pathogens human immunodeficiency virus type 1 (HIV-1) and HIV-2. Interestingly, African NHPs infected with their cognate SIV generally do not progress to AIDS, despite high levels of sustained virus replication, with the only known exception being chimpanzee SIV (SIVcpz)-infected chimpanzees (16). Among the natural hosts for SIV infection, the sooty mangabey ([SM] Cercocebus atys) is of particular interest, because cross-species transmission of SM SIV (SIVsm) from this natural host into humans initiated the HIV-2 epidemic in West Africa (17). In addition, SIVsm (herein referred to as SIV) is the ancestor of the rhesus macaque SIV (SIVmac) viruses that are used in disease pathogenesis and vaccination studies in the rhesus macaque model (17). Both naturally infected and experimentally inoculated SMs remain healthy, maintain CD4+ T cells, and do not progress to AIDS-like disease, despite sustained high levels of virus replication (31).Nonpathogenic infection of SMs is characterized by low levels of immune activation during the chronic phase of infection, which are reached after a transient immune activation that occurs during primary infection (reviewed in reference 31). These findings have led to the hypothesis that the absence of generalized immune activation in SIV-infected SMs during the chronic phase of infection is an important feature that favors the preservation of CD4+ T-cell homeostasis, thereby avoiding disease progression (31). However, most of these earlier studies focused on T cells and innate immune cells, with a significant gap existing in our understanding of whether humoral immunity might also differ between pathogenic and nonpathogenic infections. In HIV-1-infected patients, B cells produce neutralizing antibodies against the infecting (autologous) virus, which drives viral escape, continuous de novo antibody production (26-28, 32), and B-cell dysfunction (24). The striking differences in both the clinical outcomes of infection and the levels of immune activation between SIV-infected SMs and HIV-1-infected humans prompted us to compare the neutralizing antibody (Nab) response against the autologous virus in these two populations. To this end, we utilized a pseudovirus assay that has been used extensively by our group and others to evaluate Nab against HIV-1 and SIV envelope (Env) glycoproteins (15, 19, 22, 26, 28, 32, 33; also unpublished data). All SMs were housed at the Yerkes National Primate Research Center (Atlanta, GA) and maintained in accordance with National Institutes of Health guidelines. The Emory University Animal Care and Use Committee approved these studies. Details of the Zambia Emory HIV Research Project (ZEHRP) have been described elsewhere (2, 10, 21). The Emory University Institutional Review Board and the University of Zambia School of Medicine Research Ethics Committee approved informed-consent and human subject protocols. None of the subjects received antiretroviral therapy during the evaluation period.In HIV-1 infection, autologous Nabs develop to relatively high titers against the newly transmitted virus within the first few months (15, 19, 26-28, 32). Here we sought to test whether a similar increase in Nab titer occurs during nonpathogenic SIV infection of SMs. Samples were obtained from five animals that were inoculated intravenously with plasma from a naturally infected SM as part of a previous study (30). Multiple, biologically functional Envs were cloned from plasma collected at day 14 postinoculation (Table (Table1),1), and Nab activity was evaluated in plasma collected at 6 months postinoculation. To facilitate comparison with early HIV-1 infection, Nab activity in plasma was also evaluated between 2 and 9 months against Envs that were cloned between 31 and 88 estimated days after infection from four subtype C HIV-1-infected seroconverters in Zambia (Table (Table1).1). Figure Figure1A1A demonstrates that Nab activity in plasma diluted 1:100 was readily detectable in all HIV-1-infected subjects at levels approaching 100% neutralization. However, Nab activity in the SM plasma was significantly lower than in the human subjects (median, 10% versus 93%, respectively; P = 0.02). Binding antibody was detected in all five SMs at titers greater than 1:51,200 by enzyme-linked immunosorbent assay (ELISA), demonstrating that all monkeys had seroconverted by 6 months and maintained high titers of binding antibody throughout the evaluation period (Fig. (Fig.1B).1B). Thus, the low level of Nab was not due to a diminished humoral immune response.Open in a separate windowFIG. 1.Autologous Nab activity and B-cell proliferation during experimental infection of SMs. (A) Neutralization activity levels in plasma from five SMs (filled black circles), which were experimentally inoculated with plasma from a naturally SIV-infected SM, and four HIV-1-infected Zambian subjects (half-filled squares), who were recently infected through heterosexual contact, are shown. The horizontal bars represent the median for each group. To assess neutralizing activity, pseudoviruses were created by expressing each cloned Env with an HIV-1 env-deficient backbone (ΔSG3). JC53-BL (Tzm-bl) cells were infected with each pseudovirus in the presence or absence of serially diluted autologous plasma. Each point represents the average level of neutralization at a 1:100 dilution of plasma for at least two Env clones (see Table Table11 for number of Envs tested). Each neutralization assay was performed twice independently, using duplicate wells. Statistical significance between the groups was determined by a Mann-Whitney test, using GraphPad Prism 5. Longitudinal measurements of endpoint antibody ELISA titers in plasma (filled green circles) (23) (B), autologous neutralization activity in plasma (filled blue diamonds) (C), percentages of Ki-67+ CD20+ cells in blood (filled black triangles) (D), and percentages of CD20+ cells in blood (filled red squares) (E) are shown for the five experimentally inoculated SMs combined. In panel C, each point represents average neutralization at a 1:100 dilution of plasma over time for at least two day 14 Env clones from each SM. For panels D and E, PBMCs were gated by forward and side scatter, and the CD3 CD20+ population was assessed for Ki-67 staining (D) by flow cytometry. SP34-2 was used to stain CD3, L27 was used for CD20, and B56 was used for Ki-67 (all from BD Biosciences). Error bars represent the standard errors of the means (SEMs). Plasma viral load peaked at day 14 (data not shown). Filled symbols in panels A through E indicate data generated from experimentally infected SMs.

TABLE 1.

Autologous Nab activity in experimentally SIV-infected SM and acutely HIV-1-infected humans
Subject IDaVirusNo. of mo postinfection Nab activity was evaluatedNo. of days postinfection Envs were cloned from plasmaNo. of Envs tested% neutralization at a 1:100 dilution of plasma
FuvSIVsm-Fuo614416.3
FSsSIVsm-Fuo614310.6
FWvSIVsm-Fuo614510.5
FFsSIVsm-Fuo614210.3
FRsSIVsm-Fuo61439.3
185FHIV-1533494.6
153MHIV-1988594.3
221MHIV-1631691.5
205FHIV-1248587.1
Open in a separate windowaID, identification.The low level of Nab activity observed in the five experimentally inoculated SMs persisted for 16 months and did not exceed 50% at a 1:100 dilution of plasma at any time point tested (Fig. (Fig.1C).1C). In contrast, the high levels of Nab activity in the HIV-1-infected subjects persisted for over 2 years, often exceeding 50% inhibitory titers of 1:3,000 against the early virus, as is characteristic of early subtype C HIV-1 infection (15, 19, 26, 28). Figure Figure1D1D demonstrates that a transient increase in proliferating B cells, as measured by positive Ki-67 staining (12), occurred in the SMs and peaked around day 30 postinfection and then declined to a level just above baseline by day 60. Analysis using a Wilcoxon signed-rank test for paired samples showed that the percentages of Ki-67-positive (Ki-67+) B cells were higher at days 21 and 30 than at day −5, reaching borderline significance at both time points (P = 0.06). In contrast, the percentages of Ki-67+ B cells on days 60 and 475 were not significantly different from that on day −5 (P = 0.8 and 0.3, respectively). An early but transient decrease in the percentage of circulating CD20+ B cells was also observed during the initial 20 days of infection (Fig. (Fig.1E).1E). Thus, the B-cell compartment within the SM underwent changes consistent with immune activation followed by resolution. Based on these results, it does not appear that a global defect in the B-cell response in the SM can account for the low-level Nab response elicited.To investigate Nab responses during established infection, we extended this analysis to a panel of 11 naturally SIV-infected SMs in the Yerkes colony and 5 chronically HIV-1-infected subjects in Zambia. Envs were cloned from these monkeys and human subjects using peripheral blood mononuclear cell (PBMC) DNA or plasma samples, and sensitivity to Nab was evaluated. Because Nab activity against contemporaneous Env is often low or undetectable in HIV-1 infection (1, 5, 14, 25, 27, 28, 32), we evaluated plasma collected between 6 and 55 months after the Envs were cloned from each individual. Table Table22 shows that the SM Envs reflected the four SIV subtypes that circulate in the Yerkes colony (3). Figure Figure2A2A demonstrates that Nab activity in the chronically HIV-1-infected subjects was high (median, 91%), whereas in the naturally SIV-infected SMs it was again significantly lower (median, 14%; P = 0.003). Nevertheless, Nab activity in the naturally infected SMs exhibited a considerable range, from undetectable to 84% neutralization (Fig. (Fig.2A).2A). This observation prompted us to investigate whether parameters associated with disease progression in HIV-1 infection were correlated with the level of Nab activity. Figure Figure2B2B demonstrates that the number of CD4+ T cells was positively correlated with the potency of neutralization (r = 0.69; P = 0.02), while the plasma viral load showed a trend toward an inverse correlation with neutralization (Fig. (Fig.2C)2C) (r = −0.54; P = 0.08). A correlation between plasma viral load and autologous Nab titer in established HIV-1 infection has not been observed (9).Open in a separate windowFIG. 2.Autologous Nab activity and its correlation with CD4+ count and plasma viral load during established natural infection of SMs. (A) Neutralization activity levels in plasma from 11 naturally SIV-infected SMs in the Yerkes colony (open circles) and 5 chronically HIV-1-infected human subjects from Zambia (half-filled squares) are shown. Statistical significance between groups was determined by a Mann-Whitney test using GraphPad Prism 5. Correlation between Nab activity and CD3+ CD4+ T cell counts or plasma viral load in naturally infected SMs (open circles) is shown in panels (B) and (C), respectively. The percent neutralization at a 1:100 dilution of plasma (shown in panel A) is plotted along the x axis. Each CD4+ T cell count and viral load value represents the average of three measurements from samples collected from the 11 SMs approximately 1 year apart. The significance of each correlation was determined using a nonparametric Spearman test. Open circles indicate data from naturally infected SMs.

TABLE 2.

Autologous Nab activity in naturally SIV-infected SMs and HIV-1-infected humans with established infections
Subject IDaVirusEnv subtypeNo. of mo between plasma collection and Env cloningNo. of Envs tested% neutralization at 1:100 dilution of plasma
FWkSIVsm228584.4
FNnSIVsm131463.5
FFvSIVsm150459.7
FFmSIVsm130442.0
FNgSIVsm548428.0
FBnSIVsm349213.9
FDoSIVsm36512.0
FZoSIVsm12838.8
FOhSIVsm1624.7
FPnSIVsm13250.6
FFjSIVsm15420.0
109MHIV-1C6591.4
55MHIV-1C15891.3
135FHIV-1C16497.4
106MHIV-1C17579.4
153FHIV-1C55599.0
Open in a separate windowaID, identification.This study is the first to directly compare the Nab response against the autologous virus in nonpathogenic SIV versus HIV-1 infection, including evaluation of both the early, developing Nab response in acute infection and the mature response in chronic infection. A significant difference in the magnitude of Nab activity was apparent during both early and later time points, with relatively strong but ultimately ineffective neutralization activity developing and persisting into chronic infection in humans but not in SMs. Although the SIV and HIV-1 samples were obtained during similar stages of infection, the disparity in the magnitude of autologous Nab activity during early infection could in part reflect differences such as the route of infection (intravenous versus mucosal) or the complexity of the founder virus (a single variant in HIV-1 versus multiple variants in SIV). In addition, the production of SIV Env pseudoviruses in human 293T cells could have altered the glycosylation pattern or the proteins that are embedded within the virion, decreasing the neutralization susceptibility of the SIV Env pseudoviruses. However, production of a subset of these pseudoviruses in an African green monkey-derived cell line (COS-1) did not alter their Nab sensitivity (data not shown).Despite the lack of potent autologous Nab, both naturally and experimentally SIV-infected SMs produce antibodies that bind Env in ELISAs or Western blotting (4, 6, 13, 18, 23). It is possible that the SIV Env glycoproteins elicit a different profile of Nab than does HIV-1 Env. The potential for structural and biological differences between SIV and HIV-1 Envs has not been thoroughly investigated, although they would not be unexpected due to the low level of amino acid sequence conservation between them. SIVsm/HIV-2 lineage-derived Envs (i.e., the SIVmac series) show a “wide evolutionary distance” and lack of cross-reactivity with SIVcpz/HIV-1-derived Envs, with an overall sequence identity in gp120 of ∼25% across HIV-1, HIV-2, and SIVsm (7, 8). Clear biological differences in immunogenicity have been described for HIV-1 group M subtypes, which all derive from a common SIV ancestor (reviewed in reference 20). Furthermore, SM IgG antibody molecules have less flexibility in the hinge region than human IgG, which could lead to a failure of the SM antibodies to recognize recessed neutralization targets such as the receptor binding domains (29). Thus, HIV-1 Env could elicit neutralizing antibodies that are qualitatively different from those induced by SIV Env.Early resolution of immune activation could be a key feature that distinguishes nonpathogenic from pathogenic infection (12, 31). The data presented here are consistent with that hypothesis, in that signs of early B-cell proliferation were present in the experimentally infected SMs but were resolved and did not result in potent neutralizing activity. However, later in infection, the naturally infected SMs did develop low-to-moderate levels of Nab activity, and these levels were positively correlated with the number of peripheral CD4+ T cells. This finding suggests that synergy between CD4+ T cells and B cells is maintained in this nonpathogenic setting. Other biologic factors could contribute to this correlation; however, differences in age and viral subtype in this cohort of SMs could not explain this finding (data not shown).Taken together, these results indicate that a low level of autologous Nab activity is a novel and previously unappreciated feature of nonpathogenic SIV infection of SMs. The fact that high-titer Nabs are not necessary to avoid disease progression during SIV infection of SMs is consistent with the notion that the apathogenicity of natural SIV infections is not the result of particularly effective adaptive immune responses against the virus (11). It is possible that this low level of autologous Nab activity in SMs stems in part from antibody recognition of targets that are poorly exposed on the native SIV Env glycoproteins. A low level of neutralizing activity in SM may therefore have a protective effect because it does not drive viral escape or induce chronic immune activation in the B-cell compartment. Moreover, a low level of immune activation in B cells and/or preservation of CD4+ T cells could enhance the quality of the neutralizing antibody response. It will be important, in future work, to assess how this low level of autologous Nab activity in SIV-infected SMs meshes with the lower levels of immune activation and dysregulation observed in these animals. Understanding the qualitative and quantitative differences in the Nab response during pathogenic versus nonpathogenic infection could provide critical information regarding protection from AIDS.  相似文献   

19.
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   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

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