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 415 毫秒
1.
2.
Escherichia coli isolates (72 commensal and 10 O157:H7 isolates) were compared with regard to physiological and growth parameters related to their ability to survive and persist in the gastrointestinal tract and found to be similar. We propose that nonhuman hosts in E. coli O157:H7 strains function similarly to other E. coli strains in regard to attributes relevant to gastrointestinal colonization.Escherichia coli is well known for its ecological versatility (15). A life cycle which includes both gastrointestinal and environmental stages has been stressed by both Savageau (15) and Adamowicz et al. (1). The gastrointestinal stage would be subjected to acid and detergent stress. The environmental stage is implicit in E. coli having transport systems for fungal siderophores (4) as well as pyrroloquinoline quinone-dependent periplasmic glucose utilization (1) because their presence indicates evolution in a location containing fungal siderophores and pyrroloquinoline quinone (1).Since its recognition as a food-borne pathogen, there have been numerous outbreaks of food-borne infection due to E. coli O157:H7, in both ground beef and vegetable crops (6, 13). Cattle are widely considered to be the primary reservoir of E. coli O157:H7 (14), but E. coli O157:H7 does not appear to cause disease in cattle. To what extent is E. coli O157:H7 physiologically unique compared to the other naturally occurring E. coli strains? We feel that the uniqueness of E. coli O157:H7 should be evaluated against a backdrop of other wild-type E. coli strains, and in this regard, we chose the 72-strain ECOR reference collection originally described by Ochman and Selander (10). These strains were chosen from a collection of 2,600 E. coli isolates to provide diversity with regard to host species, geographical distribution, and electromorph profiles at 11 enzyme loci (10).In our study we compared the 72 strains of the ECOR collection against 10 strains of E. coli O157:H7 and six strains of E. coli which had been in laboratory use for many years (Table (Table1).1). The in vitro comparisons were made with regard to factors potentially relevant to the bacteria''s ability to colonize animal guts, i.e., acid tolerance, detergent tolerance, and the presence of the Entner-Doudoroff (ED) pathway (Table (Table2).2). Our longstanding interest in the ED pathway (11) derives in part from work by Paul Cohen''s group (16, 17) showing that the ED pathway is important for E. coli colonization of the mouse large intestine. Growth was assessed by replica plating 88 strains of E. coli under 40 conditions (Table (Table2).2). These included two LB controls (aerobic and anaerobic), 14 for detergent stress (sodium dodecyl sulfate [SDS], hexadecyltrimethylammonium bromide [CTAB], and benzalkonium chloride, both aerobic and anaerobic), 16 for acid stress (pH 6.5, 6.0, 5.0, 4.6, 4.3, 4.2, 4.1, and 4.0), four for the ability to grow in a defined minimal medium (M63 glucose salts with and without thiamine), and four for the presence or absence of a functional ED pathway (M63 with gluconate or glucuronate). All tests were done with duplicate plates in two or three separate trials. The data are available in Tables S1 to S14 in the supplemental material, and they are summarized in Table Table22.

TABLE 1.

E. coli strains used in this study
E. coli strain (n)Source
ECOR strains (72)Thomas Whittman
Laboratory adapted (6)
    K-12 DavisPaul Blum
    CG5C 4401Paul Blum
    K-12 StanfordPaul Blum
    W3110Paul Blum
    BTyler Kokjohn
    AB 1157Tyler Kokjohn
O157:H7 (10)
    FRIK 528Andrew Benson
    ATCC 43895Andrew Benson
    MC 1061Andrew Benson
    C536Tim Cebula
    C503Tim Cebula
    C535Tim Cebula
    ATCC 43889William Cray, Jr.
    ATCC 43890William Cray, Jr.
    ATCC 43888Willaim Cray, Jr.
    ATCC 43894William Cray, Jr.
Open in a separate window

TABLE 2.

Physiological comparison of 88 strains of Escherichia coli
Growth medium or conditionOxygencNo. of strains with type of growthb
ECOR strains (n = 72)
Laboratory strains (n = 6)
O157:H7 strains (n = 10)
GoodPoorNoneVariableGoodPoorNoneVariableGoodPoorNoneVariable
LB controlaBoth72000600010000
1% SDSAerobic6930060008002
5% SDSAerobic6840060008200
1% SDSAnaerobic53154023101702
5% SDSAnaerobic0684004200704
CTABd (all)Both00720006000100
0.05% BACAerobic31158202220091
0.2% BACAerobic01710105000100
0.05% BACAnaerobic2367001500091
0.2% BACAnaerobic00720006000100
pH 6.5Both72000600010000
pH 6Both72000600010000
pH 5Both7020060009001
pH 4.6Both70200600010000
pH 4.3Aerobic14015731203205
pH 4.3Anaerobic6930031201100
pH 4.1 or 4.2Aerobic00720NDgND
pH 4.0Both0072000600091
M63 with supplemente
    GlucoseAerobicf6912050109010
    GlucoseAnaerobicf7002050109010
    GluconateBoth6912050109010
    GlucuronateAerobic6822050109010
    GlucuronateAnaerobic6912050109010
Open in a separate windowaEight LB controls were run, two for each set of LB experiments: SDS, CTAB, benzalkonium chloride (BAC), and pH stress.bGrowth was measured as either +++, +, or 0 (good, poor, and none, respectively), with +++ being the growth achieved on the LB control plates. “Variable” means that two or three replicates did not agree. All experiments were done at 37°C.c“Anaerobic” refers to use of an Oxoid anaerobic chamber. Aerobic and anaerobic growth data are presented together when the results were identical and separately when the results were not the same or the anaerobic set had not been done. LB plates were measured after 1 (aerobic) or 2 (anaerobic) days, and the M63 plates were measured after 2 or 3 days.dCTAB used at 0.05, 0.2%, and 0.4%.eM63 defined medium (3) was supplemented with glucose, gluconate, or glucuronate, all at 0.2%.fIdentical results were obtained with and without 0.0001% thiamine.gND, not determined.  相似文献   

3.
4.
Twelve cluster groups of Escherichia coli O26 isolates found in three cattle farms were monitored in space and time. Cluster analysis suggests that only some O26:H11 strains had the potential for long-term persistence in hosts and farms. As judged by their virulence markers, bovine enterohemorrhagic O26:H11 isolates may represent a considerable risk for human infection.Shiga toxin (Stx)-producing Escherichia coli (STEC) strains comprise a group of zoonotic enteric pathogens (42). In humans, infections with some STEC serotypes result in hemorrhagic or nonhemorrhagic diarrhea, which can be complicated by hemolytic-uremic syndrome (HUS) (49). These STEC strains are also designated “enterohemorrhagic E. coli” (EHEC). Consequently, EHEC strains represent a subgroup of STEC with a high pathogenic potential for humans. Strains of the E. coli serogroup O26 were originally classified as enteropathogenic E. coli due to their association with outbreaks of infantile diarrhea in the 1940s. In 1977, Konowalchuk et al. (37) recognized that these bacteria produced Stx, and 10 years later, the Stx-producing E. coli O26:H11/H− strains were classified as EHEC. EHEC O26 strains constitute the most common non-O157 EHEC group associated with diarrhea and HUS in Europe (12, 21, 23, 24, 26, 27, 55, 60). Reports on an association between EHEC O26 and HUS or diarrhea from North America including the United States (15, 30, 33), South America (51, 57), Australia (22), and Asia (31, 32) provide further evidence for the worldwide spread of these organisms. Studies in Germany and Austria (26, 27) on sporadic HUS cases between 1996 and 2003 found that EHEC O26 accounted for 14% of all EHEC strains and for ∼40% of non-O157 EHEC strains obtained from these patients. A proportion of 11% EHEC O26 strains was detected in a case-control study in Germany (59) between 2001 and 2003. In the age group <3 years, the number of EHEC O26 cases was nearly equal to that of EHEC O157 cases, although the incidence of EHEC O26-associated disease is probably underestimated because of diagnostic limitations in comparison to the diagnosis of O157:H7/H− (18, 34). Moreover, EHEC O26 has spread globally (35). Beutin (6) described EHEC O26:H11/H−, among O103:H2, O111:H, O145:H28/H−, and O157:H7/H−, as the well-known pathogenic “gang of five,” and Bettelheim (5) warned that we ignore the non-O157 STEC strains at our peril.EHEC O26 strains produce Stx1, Stx2, or both (15, 63). Moreover, these strains contain the intimin-encoding eae gene (11, 63), a characteristic feature of EHEC (44). In addition, EHEC strains possess other markers associated with virulence, such as a large plasmid that carries further potential virulence genes, e.g., genes coding for EHEC hemolysin (EHEC-hlyA), a catalase-peroxidase (katP), and an extracellular serine protease (espP) (17, 52). The efa1 (E. coli factor for adherence 1) gene was identified as an intestinal colonization factor in EHEC (43). EHEC O26 represents a highly dynamic group of organisms that rapidly generate new pathogenic clones (7, 8, 63).Ruminants, especially cattle, are considered the primary reservoir for human infections with EHEC. Therefore, the aim of this study was the molecular characterization of bovine E. coli field isolates of serogroup O26 using a panel of typical virulence markers. The epidemiological situation in the beef herds from which the isolates were obtained and the spatial and temporal behavior of the clonal distribution of E. coli serogroup O26 were analyzed during the observation period. The potential risk of the isolates inducing disease in humans was assessed.In our study, 56 bovine E. coli O26:H11 isolates and one bovine O26:H32 isolate were analyzed for EHEC virulence-associated factors. The isolates had been obtained from three different beef farms during a long-term study. They were detected in eight different cattle in farm A over a period of 15 months (detected on 10 sampling days), in 3 different animals in farm C over a period of 8 months (detected on 3 sampling days), and in one cow on one sampling day in farm D (Table (Table1)1) (28).

TABLE 1.

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

5.
Vertebrate genomic assemblies were analyzed for endogenous sequences related to any known viruses with single-stranded DNA genomes. Numerous high-confidence examples related to the Circoviridae and two genera in the family Parvoviridae, the parvoviruses and dependoviruses, were found and were broadly distributed among 31 of the 49 vertebrate species tested. Our analyses indicate that the ages of both virus families may exceed 40 to 50 million years. Shared features of the replication strategies of these viruses may explain the high incidence of the integrations.It has long been appreciated that retroviruses can contribute significantly to the genetic makeup of host organisms. Genes related to certain other viruses with single-stranded RNA genomes, formerly considered to be most unlikely candidates for such contribution, have recently been detected throughout the vertebrate phylogenetic tree (1, 6, 13). Here, we report that viruses with single-stranded DNA (ssDNA) genomes have also contributed to the genetic makeup of many organisms, stretching back as far as the Paleocene period and possibly the late Cretaceous period of evolution.Determining the evolutionary ages of viruses can be problematic, as their mutation rates may be high and their replication may be rapid but also sporadic. To establish a lower age limit for currently circulating ssDNA viruses, we analyzed 49 published vertebrate genomic assemblies for the presence of sequences derived from the NCBI RefSeq database of 2,382 proteins from known viruses in this category, representing a total of 23 classified genera from 7 virus families. Our survey uncovered numerous high-confidence examples of endogenous sequences related to the Circoviridae and to two genera in the family Parvoviridae: the parvoviruses and dependoviruses (Fig. (Fig.11).Open in a separate windowFIG. 1.Phylogenetic tree of vertebrate organisms and history of ssDNA virus integrations. Times of integration of ancestral dependoviruses (yellow icosahedrons), parvoviruses (blue icosahedrons), and circoviruses (triangles) are approximate.The Dependovirus and Parvovirus genomes are typically 4 to 6 kb in length, include 2 major open reading frames (encoding replicase proteins [Rep and NS1, respectively] and capsid proteins [Cap and VP1, respectively]), and have characteristic hairpin structures at both ends (Fig. (Fig.2).2). For replication, these viruses depend on host enzymes that are recruited by the viral replicase proteins to the hairpin regions, where self-primed viral DNA synthesis is initiated (2). Circovirus genomes are typically ∼2-kb circles. DNA of the type species, porcine circovirus 1 (PCV-1), contains a stem-loop structure within the origin of replication (Fig. (Fig.2),2), and the largest open reading frame includes sequences that are homologous to the Parvovirus replicase open reading frame (9, 11). The circoviruses also depend on host enzymes for replication, and DNA synthesis is self-primed from a 3′-OH end formed by endonucleolytic cleavage of the stem-loop structure (4). The frequency of Dependovirus infection is estimated to be as high as 90% within an individual''s lifetime. None of the dependoviruses have been associated with human disease, but related viruses in the family Parvoviridae (e.g., erythrovirus B19 and possibly human bocavirus) are pathogenic for humans, and members of both the Parvoviridae and the Circoviridae can cause a variety of animal diseases (2, 4).Open in a separate windowFIG. 2.Schematics illustrating the structure and organization of Parvoviridae and Circoviridae genomes and origins of several of the longest-integrated ancestral viral sequences found in vertebrates. Integrations were aligned to the Dependovirus adeno-associated virus 2 (AAV2), the Parvovirus minute virus of mice (MVM), and the Circovirus porcine circovirus 1 (PCV-1). The inverted terminal repeat (ITR) sequences in the Dependovirus and Parvovirus genomes are depicted on an expanded scale. A linear representation of the circular genome of PCV-1 is shown with the 10-bp stem-loop structure on an expanded scale. Horizontal lines beneath the maps indicate the lengths of similar sequences that could be identified by BLAST. The numbers indicate the locations of amino acids in the viral proteins where the sequence similarities in the endogenous insertions start and end. The actual ancestral virus-derived integrated sequences may extend beyond the indicated regions.With some ancestral endogenous sequences that we identified, phylogenetic comparisons can be used to estimate age. For example, as a Dependovirus-like sequence is present at the same location in the genomes of mice and rats, the ancestral virus must have existed before their divergence, more than 20 million years ago. Some Circovirus- and Dependovirus-related integrations also predate the split between dog and panda, about 42 million years ago. However, in most other cases, we rely on an indirect method for estimating age (1). As genomic sequences evolve, they accumulate new stop codons and insertion/deletion-induced frameshifts. The rates of these events can be tied directly to the rates of neutral sequence drift and, therefore, the time of evolution. To apply this method, we first performed a BLAST search of vertebrate genomes for all known ssDNA virus proteins (BLAST options, -p tblastn -M BLOSUM62 -e 1e−4). Candidate sequences were then recorded, along with 5 kb of flanking regions, and then again aligned against the database of ssDNA viruses to find the most complete alignment (BLAST options, -t blastx -F F -w 15 -t 1500 -Z 150 -G 13 -E 1 -e 1e−2). Detected alignments were then compared with a neutral model of genome evolution, as described in the supplemental material, and the numbers of stop codons and frameshifts were converted into the expected genomic drift undergone by the sequences. The age of integration was then estimated from the known phylogeny of vertebrates (7, 10). Using these methods, we discovered that as many as 110 ssDNA virus-related sequences have been integrated into the 49 vertebrate genomes considered, during a time period ranging from the present to over 40 to 60 million years ago (Table (Table1;1; see also Tables S1 to S3 in the supplemental material).

TABLE 1.

Selected endogenous sequences in vertebrate genomes related to single-stranded DNA viruses
Virus group and vertebrate speciesInitial genomic search using TBLASTN
Best sequence homology identified using BLASTX
Predicted nucleotide drift (%)Integration labelAge (million yr) or timing of integration based on sequence aging
Chromosomal or scaffold locationProteinBLAST E value/% sequence identityMost similar virusaProteinCoordinatesNo. of stop codons/frameshifts
Circoviruses
    CatScaffold_62068Rep6E−05/37Canary circovirusRep4-2833/7 in 268 aab14.2fcECLG-182
Scaffold_24038Rep6E−06/51Columbid circovirusRep44-3174/5 in 231 aac15.2fcECLG-287
    DogChr5dRep7E−16/46Raven circovirusRep16-2636/5 in 250 aa17.6cfECLG-198
Chr22Rep1E−14/43Beak and feather disease virusRep7-2642/1 in 261 aac4.5cfECLG-254
    OpossumChr3Rep4E−46/44Finch circovirusRep2-2910/2 in 282 aa2.3mdECLG12
Cap6-360/0 in 30 aa
Dependoviruses
    DogChrXRep6E−05/55AAV5Rep239-4453/4 in 200 aa14.0cfEDLG-178
    DolphinGeneScaffold1475Rep8E−39/39Avian AAV DA1Rep79-4863/4 in 379 aac6.6ttEDLG-255
Cap4E−61/47Cap1-7384/7 in 678 aac
    ElephantScaffold_4Rep0/55AAV5Rep3-5890/0 in 579 aa0.0laEDLGRecent
    HyraxGeneScaffold5020Cap3E−34/53AAV3Cap485-7350/5 in 256 aa7.0pcEDLG-129
Scaffold_19252Rep9E−72/47Bovine AAVRep2-3488/4 in 348 aa14.3pcEDLG-260
    MegabatScaffold_5601Rep2E−13/31AAV2Rep315-4791/5 in 175 aa13.1pvEDLG-376
    MicrobatGeneScaffold2026Rep1E−117/50AAV2Rep1-6172/5 in 612 aa5.8mlEDLG-127
Cap9E−33/51Cap1-7312/9 in 509 aac
Scaffold_146492Cap6E−32/42AAV2Cap479-7320/3 in 252 aa4.2mlEDLG-219
    MouseChr1Rep2E−06/34AAV2Rep4-2063/5 in 191 aa17.1mmEDLG-139
Chr3Rep2E−24/31AAV5Rep71-47812/7 in 389 aa16.5mmEDLG-237
Cap2E−22/45Cap22-72412/10 in 649aac
Chr8Rep1E−08/46AAV2Rep314-4733/3 in 147 aa13.8mmEDLG-331
Cap1-1371/2 in 114 aa
    PandaScaffold2359Rep2E−06/37Bovine AAVRep238-4262/3 in 186 aa10.4amEDLG-159
    PikaScaffold_9941Rep4E−14/28AAV5Rep126-4152/2 in 282 aa5.4opEDLG14
    PlatypusChr2Rep9E−10/35Bovine AAVRep297-4374/3 in 138 aa17.1oaEDLG-179
Cap272-4191/2 in 150 aac
Contig12430Rep2E−09/47Bovine AAVRep353-4503/1 in 123 aa12.0oaEDLG-255
Cap2E−05/32Cap253-3672/1 in 116 aa
    RabbitChr10Rep3E−97/39AAV2Rep1-6193/9 in 613 aa9.3ocEDLG43
Cap5E−50/45Cap1-72310/9 in 675 aa
    RatChr13Rep2E−09/33AAV2Rep4-1752/4 in 177 aa13.3rnEDLG-128
Chr2Rep4E−18/40AAV5Rep1-46112/12 in 454 aa22.7rnEDLG-251
Chr19Rep2E−07/33AAV5Rep329-4642/4 in 136 aa16.1rnEDLG-335
Cap31-1332/1 in 93 aa
    TarsierScaffold_178326Rep4E−14/23AAV5Rep96-4652/3 in 356 aa5.3tsEDLG23
Parvoviruses
    Guinea pigScaffold_188Rep3E−24/46Porcine parvovirusRep313-5675/3 in 250 aa12.3cpEPLG-140
Cap1E−16/36Cap10-68911/12 in 672 aa
Scaffold_27Rep1E−50/39Canine parvovirusRep11-6401/4 in 616 aa5.3cpEPLG-217
Cap1E−38/39Porcine parvovirusCap3-7192/14 in 700 aa
    TenrecScaffold_260946Rep2E−20/38LuIII virusRep406-5984/4 in 190 aa19.0etEPLG-260
Cap11-63916/15 in 595 aa
    RatChr5Rep6E−10/56Canine parvovirusRep1-2820/0 in 312 aa0.6rnEPLGRecent
Cap0/62Cap637-6670/2 in 760 aa
Rep0/631-751
    OpossumChr3Rep2E−39/33LuIII virusRep7-57011/3 in 502 aa10.9mdEPLG-256
Cap7E−8/33Cap11-72914/7 in 704 aa
Chr6Rep6E−58/44Porcine parvovirusRep16-5633/7 in 534 aac4.6mdEPLG-324
Cap6E−60/38Cap10-7152/5 in 707 aac
    WallabyScaffold_108040Rep4E−74/62Canine parvovirusRep341-6450/0 in 287 aa1.3meEPLG-37
Cap8E−37/32Cap35-7380/4 in 687 aa
Scaffold_72496Rep2E−61/42Porcine parvovirusRep23-5674/3 in 531 aa5.7meEPLG-630
Cap2E−31/38Cap10-5326/4 in 514 aa
Scaffold_88340Rep7E−37/55Mouse parvovirus 1Rep344-5660/3 in 223 aa6.7meEPLG-1636
Cap7E−22/33Cap11-7136/9 in 700 aa
Open in a separate windowaSome ambiguity in choosing the most similar virus is possible. We generally used the alignment with the lowest E value in the BLAST results. However, one or two points in the exponent of an E value were sometimes sacrificed to achieve a longer sequence alignment.baa, amino acids.cThese sequences have long insertions compared to the present-day viruses. In all cases tested, these insertions originated from short interspersed elements (SINEs). These insertions were excluded from the counts of stop codons and frameshifts and the estimation of integration age.dChr, chromosome.It is important to recognize that there is an intrinsic limit on how far back in time we can reach to identify ancient endogenous viral sequences. First, the sequences must be identified with confidence by BLAST or similar programs. This requirement places a lower limit on sequence identity at about 20 to 30% of amino acids, or about 75% of nucleotides (nucleotides evolve nearly 2.5 times slower than the amino acid sequence they encode). Second, the related, present-day virus must have evolved at a rate that is not much higher than that of the endogenous sequences. The viruses for which ancestral endogenous sequences were identified in this study exhibit sequence drift similar to that associated with mammalian genomes. Setting this rate at 0.14% per million years of evolution (8), we arrive at 90 million years as the theoretical limit for the oldest sequences that can be identified using our methods. This limit drops to less than 35 million years for endogenous viral sequences in rodents and even lower for sequences related to viruses that evolve faster than mammalian genomes.The most widespread integrations found in our survey are derived from the dependoviruses. These include nearly complete genomes related to adeno-associated virus (AAV) in microbat, wallaby, dolphin, rabbit, mouse, and baboon (Fig. (Fig.2).2). We did not detect inverted terminal repeats in several integrations tested, even though repeats are common in the present-day dependoviruses. This result could be explained by sequence decay or the absence of such structures in the ancestral viruses. However, we do see sequences that resemble degraded hairpin structures to which Dependovirus Rep proteins bind, with an example from microbat integration mlEDLG-1 shown in Fig. Fig.3.3. The second most widespread endogenous sequences are related to the parvoviruses. They are found in 6 of 49 vertebrate species considered, with nearly complete genomes in rat, opossum, wallaby, and guinea pig (Fig. (Fig.22).Open in a separate windowFIG. 3.Hairpin structure of the inverted terminal repeat of adeno-associated virus 2 (left) and a candidate degraded hairpin structure located close to the 5′ end of the mlEDLG-1 integration in microbats (right). Structures and mountain plots were generated using default parameters of the RNAfold program (5), with nucleotide coloring representing base-pairing probabilities: blue is below average, green is average, and red is above average. Mountain plots represent hairpin structures based on minimum free energy (mfe) calculations and partition function (pf) calculations, as well as the centroid structure (5). Height is expressed in numbers of nucleotides; position represents nucleotide.The Dependovirus AAV2 has strong bias for integration into human chromosome 19 during infection, driven by a host sequence that is recognized by the viral Rep protein(s). Rep mediates the formation of a synapse between viral and cellular sequences, and the cellular sequences are nicked to serve as an origin of viral replication (14). The related integrations in mice and rats, located in the same chromosomal locations, might be explained by such a mechanism. However, the extent of endogenous sequence decay and the frequency of stop codons indicate that these integrations occurred some 30 to 35 million years ago, implying that they are derived from a single event in a rodent ancestor rather than two independent integration events at the same location. Similarly, integrations EDLG-1 in dog and panda lie in chromosomal regions that can be readily aligned (based on University of California—Santa Cruz [UCSC] genome assemblies) and show sequence decay consistent with the age of the common ancestor, about 42 million years. Endogenous sequences related to the family Parvoviridae can thus be traced to over 40 million years back in time, and viral proteins related to this family have remained over 40% conserved.Sequences related to circoviruses were detected in five vertebrate species (Table (Table11 and Table S1 in the supplemental material). At least one of these sequences, the endogenous sequence in opossum, likely represents a recent integration. Several integrations in dog, cat, and panda, on the other hand, appear to date from at least 42 million years ago, which is the last time when pandas and dogs shared a common ancestor. We see evidence for this age in data from sequence degradation (Table (Table1),1), phylogenetic analyses of endogenous Circovirus-like genomes (see Fig. S2 in the supplemental material), and genomic synteny where integration ECLG-3 is surrounded by genes MTA3 and ARID5A in both dog and panda and integration ECLG-2 lies 35 to 43 kb downstream of gene UPF3A. In fact, Circovirus integrations may even precede the split between dogs and cats, about 55 million years ago, although the preliminary assembly and short genomic contigs for cats make synteny analysis impossible.The most common Circovirus-related sequences detected in vertebrate genomes are derived from the rep gene. We speculate that, like those of the Parvoviridae, the ancestral Circoviridae sequences might have been copied using a primer sequence in the host DNA that resembled the viral origin and was therefore recognized by the virus Rep protein. Higher incidence of rep gene identifications may represent higher conservation of this gene with time, or alternatively, possession of these sequences may impart some selective advantage to the host species. The largest Circovirus-related integration detected, in the opossum, comprises a short fragment of what may have been the cap gene immediately adjacent to and in the opposite orientation from the rep gene. This organization is similar to that of the present day Circovirus genome in which these genes share a promoter in the hairpin regions but are translated in opposite directions (Fig. (Fig.22).In summary, our results indicate that sequences derived from ancestral members of the families Parvoviridae and Circoviridae were integrated into their host''s genomes over the past 50 million years of evolution. Features of their replication strategies suggest mechanisms by which such integrations may have occurred. It is possible that some of the endogenous viral sequences could offer a selective advantage to the virus or the host. We note that rep open reading frame-derived proteins from some members of these families kill tumor cells selectively (3, 12). The genomic “fossils” we have discovered provide a unique glimpse into virus evolution but can give us only a lower estimate of the actual ages of these families. However, numerous recent integrations suggest that their germ line transfer has been continuing into present times.   相似文献   

6.
Specific therapy is not available for hantavirus cardiopulmonary syndrome caused by Andes virus (ANDV). Peptides capable of blocking ANDV infection in vitro were identified using antibodies against ANDV surface glycoproteins Gn and Gc to competitively elute a cyclic nonapeptide-bearing phage display library from purified ANDV particles. Phage was examined for ANDV infection inhibition in vitro, and nonapeptides were synthesized based on the most-potent phage sequences. Three peptides showed levels of viral inhibition which were significantly increased by combination treatment with anti-Gn- and anti-Gc-targeting peptides. These peptides will be valuable tools for further development of both peptide and nonpeptide therapeutic agents.Andes virus (ANDV), an NIAID category A agent linked to hantavirus cardiopulmonary syndrome (HCPS), belongs to the family Bunyaviridae and the genus Hantavirus and is carried by Oligoryzomys longicaudatus rodents (11). HCPS is characterized by pulmonary edema caused by capillary leak, with death often resulting from cardiogenic shock (9, 16). ANDV HCPS has a case fatality rate approaching 40%, and ANDV is the only hantavirus demonstrated to be capable of direct person-to-person transmission (15, 21). There is currently no specific therapy available for treatment of ANDV infection and HCPS.Peptide ligands that target a specific protein surface can have broad applications as therapeutics by blocking specific protein-protein interactions, such as preventing viral engagement of host cell receptors and thus preventing infection. Phage display libraries provide a powerful and inexpensive tool to identify such peptides. Here, we used selection of a cyclic nonapeptide-bearing phage library to identify peptides capable of binding the transmembrane surface glycoproteins of ANDV, Gn and Gc, and blocking infection in vitro.To identify peptide sequences capable of recognizing ANDV, we panned a cysteine-constrained cyclic nonapeptide-bearing phage display library (New England Biolabs) against density gradient-purified, UV-treated ANDV strain CHI-7913 (a gift from Hector Galeno, Santiago, Chile) (17, 18). To increase the specificity of the peptides identified, we eluted phage by using monoclonal antibodies (Austral Biologicals) prepared against recombinant fragments of ANDV Gn (residues 1 to 353) or Gc (residues 182 to 491) glycoproteins (antibodies 6B9/F5 and 6C5/D12, respectively). Peptide sequences were determined for phage from iterative rounds of panning, and the ability of phage to inhibit ANDV infection of Vero E6 cells was determined by immunofluorescent assay (IFA) (7). Primary IFA detection antibodies were rabbit polyclonal anti-Sin Nombre hantavirus (SNV) nucleoprotein (N) antibodies which exhibit potent cross-reactivity against other hantavirus N antigens (3). ReoPro, a commercially available Fab fragment which partially blocks infection of hantaviruses in vitro by binding the entry receptor integrin β3 (5), was used as a positive control (80 μg/ml) along with the original antibody used for phage elution (5 μg/ml). As the maximum effectiveness of ReoPro in inhibiting hantavirus entry approaches 80%, we set this as a threshold for maximal expected efficacy for normalization. The most-potent phage identified by elution with the anti-Gn antibody 6B9/F5 bore the peptide CPSNVNNIC and inhibited hantavirus entry by greater than 60% (61%) (Table (Table1).1). From phage eluted with the anti-Gc antibody 6C5/D12, those bearing peptides CPMSQNPTC and CPKLHPGGC also inhibited entry by greater than 60% (66% and 72%, respectively).

TABLE 1.

Peptide-bearing phage eluted from ANDV
Phage% Inhibition (SD)aP valueb
Phage bearing the following peptides eluted with anti-Gn antibody 6B9/F5
    Group 1 (<30% inhibition)
        CDQRTTRLC8.45 (15.34)0.0002
        CPHDPNHPC9.94 (7.72)0.333
        CQSQTRNHC11.76 (13.25)0.0001
        CLQDMRQFC13.26 (9.92)0.0014
        CLPTDPIQC15.70 (14.05)0.0005
        CPDHPFLRC16.65 (15.22)0.8523
        CSTRAENQC17.56 (16.50)0.0004
        CPSHLDAFC18.98 (20.06)0.0017
        CKTGHMRIC20.84 (7.47)0.0563
        CVRTPTHHC20.89 (27.07)0.1483
        CSGVINTTC21.57 (19.61)0.0643
        CPLASTRTC21.65 (5.98)0.004
        CSQFPPRLC22.19 (8.26)0.0004
        CLLNKQNAC22.34 (7.78)0.001
        CKFPLNAAC22.89 (6.15)0.0001
        CSLTPHRSC23.63 (16.74)0.0563
        CKPWPMYSC23.71 (6.68)0.0643
        CLQHDALNC24.01 (7.60)1
        CNANKPKMC24.67 (11.67)0.0004
        CPKHVLKVC25.30 (28.36)0.0003
        CTPDKKSFC26.91 (11.15)0.399
        CHGKAALAC27.22 (32.53)0.005
        CNLMGNPHC28.08 (21.35)0.0011
        CLKNWFQPC28.64 (18.49)0.0016
        CKEYGRQMC28.76 (29.33)0.0362
        CQPSDPHLC29.44 (31.22)0.0183
        CSHLPPNRC29.70 (17.37)0.0061
    Group 2 (30-59% inhibition)
        CSPLLRTVC33.05 (20.26)0.0023
        CHKGHTWNC34.17 (12.50)0.0795
        CINASHAHC35.62 (13.03)0.3193
        CWPPSSRTC36.75 (26.95)0.0006
        CPSSPFNHC37.78 (7.11)0.0001
        CEHLSHAAC38.47 (7.60)0.0115
        CQDRKTSQC38.74 (9.12)0.1802
        CTDVYRPTC38.90 (25.03)0.006
        CGEKSAQLC39.11 (27.52)0.0013
        CSAAERLNC40.13 (6.33)0.0033
        CFRTLEHLC42.07 (5.01)0.0608
        CEKLHTASC43.60 (27.92)0.1684
        CSLHSHKGC45.11 (49.81)0.0864
        CNSHSPVHC45.40 (28.80)0.0115
        CMQSAAAHC48.88 (44.40)0.5794
        CPAASHPRC51.84 (17.09)0.1935
        CKSLGSSQC53.90 (13.34)0.0145
    Group 3 (60-79% inhibition)
        CPSNVNNIC61.11 (25.41)0.1245
Negative control0 (6.15)
6B9/F5 (5 μg/ml)26.77 (5.33)
ReoPro (80 μg/ml)79.86 (4.88)
Phage bearing the following peptides eluted with anti-Gc antibody 6C5/D12
    Group 1 (<30% inhibition)
        CHPGSSSRC1.01 (7.03)0.0557
        CSLSPLGRC10.56 (13.62)0.7895
        CTARYTQHC12.86 (3.83)0.3193
        CHGVYALHC12.91 (7.32)0.0003
        CLQHNEREC16.79 (13.72)0.0958
        CHPSTHRYC17.23 (14.53)0.0011
        CPGNWWSTC19.34(9.91)0.1483
        CGMLNWNRC19.48 (19.42)0.0777
        CPHTQFWQC20.44 (13.65)0.0008
        CTPTMHNHC20.92 (11.68)0.0001
        CDQVAGYSC21.79 (23.60)0.0063
        CIPMMTEFC24.33 (9.28)0.2999
        CERPYSRLC24.38 (9.09)0.0041
        CPSLHTREC25.06 (22.78)0.1202
        CSPLQIPYC26.30 (34.29)0.4673
        CTTMTRMTC (×2)29.27 (8.65)0.0001
    Group 2 (30-59% inhibition)
        CNKPFSLPC30.09 (5.59)0.4384
        CHNLESGTC31.63 (26.67)0.751
        CNSVPPYQC31.96 (6.51)0.0903
        CSDSWLPRC32.95 (28.54)0.259
        CSAPFTKSC33.40 (10.64)0.0052
        CEGLPNIDC35.63 (19.90)0.0853
        CTSTHTKTC36.28 (13.42)0.132
        CLSIHSSVC36.40 (16.44)0.8981
        CPWSTQYAC36.81 (32.81)0.5725
        CTGSNLPIC36.83 (31.64)0.0307
        CSLAPANTC39.73 (4.03)0.1664
        CGLKTNPAC39.75 (16.98)0.2084
        CRDTTPWWC40.08 (18.52)0.0004
        CHTNASPHC40.26 (4.77)0.5904
        CTSMAYHHC41.89 (8.61)0.259
        CSLSSPRIC42.13 (29.75)0.2463
        CVSLEHQNC45.54 (6.55)0.5065
        CRVTQTHTC46.55 (8.45)0.3676
        CPTTKSNVC49.28 (14.00)0.3898
        CSPGPHRVC49.50 (42.60)0.0115
        CKSTSNVYC51.20 (4.60)0.0611
        CTVGPTRSC57.30 (11.31)0.0176
    Group 3 (60-79% inhibition)
        CPMSQNPTC65.60 (13.49)0.014
        CPKLHPGGC71.88 (27.11)0.0059
Negative control0.26 (4.53)
6C5/D12 (5 μg/ml)22.62 (8.40)
ReoPro (80 μg/ml)80.02 (76.64)
Open in a separate windowaStandard deviations of four experiments are shown in parentheses. Peptide-bearing phage were added at 109 phage/μl.bP values for the pairwise amino acid alignment score of each peptide versus that of integrin β3 were determined using an unpaired Student''s t test. P values considered statistically significant are shown in bold.To determine whether the peptide sequences of any of the identified inhibitory phage showed homology to integrin β3, a known entry receptor for pathogenic hantaviruses (6, 7), we used the Gap program to perform a pairwise amino acid alignment of each peptide versus the extracellular portion of integrin β3 and determined P values for the alignments. Of 45 phage eluted with the anti-Gn antibody, 6B9/F5, 27 of the peptide sequences showed homology to integrin β3 (P < 0.05), and 9 were highly significant (P ≤ 0.0005) (Fig. (Fig.1A).1A). Of the latter, CKFPLNAAC and CSQFPPRLC map to the hybrid domain (Fig. (Fig.1B),1B), which is proximal to the plexin-semaphorin-integrin domain (PSI) containing residue D39, shown to be critical for viral entry in vitro (19). Five sequences (CPSSPFNH, CPKHVLKVC, CNANKPKMC, CQSQTRNHC, and CDQRTTRLC) map to the I-like (or βA) domain near the binding site of ReoPro (2). Finally, CLPTDPIQC maps to the epidermal growth factor 4 (EGF-4) domain, and CSTRAENQC aligns to a portion of β3 untraceable in the crystal structure, specifically the linker region between the hybrid domain and EGF-1. Although this represents a disordered portion of the protein (22), the location of this loop proximal to the PSI domain is worth noting, due to the role of the PSI domain in facilitating viral entry (19). Therefore, 60% of phage eluted with the anti-Gn antibody showed some homology to integrin β3, and those with highly significant P values predominantly mapped to or proximal to regions of known interest in viral entry.Open in a separate windowFIG. 1.Inhibitory peptides identified through phage panning against ANDV show homology to integrin β3. (A) Alignment of phage peptide sequences with P values for integrin β3 pairwise alignment of less than 0.05. Residues comprising the signal peptide, transmembrane, and cytoplasmic domains, which were not included during pairwise alignment, are underlined. Residues 461 to 548, which are missing in the crystal structure, are italicized. Residues involved in the ReoPro binding site are highlighted in green (2). Residue D39 of the PSI domain is highlighted in yellow (19). Peptides are shown above the sequence of integrin β3, with antibody 6C5/D12-eluted sequences shown in blue text and sequences eluted with antibody 6B9/F5 shown in red. Peptide sequences with alignment P values of ≤0.0005 are highlighted in yellow. Percent inhibition of the peptide-bearing phage is shown in parentheses. (B) View of integrin αvβ3 (PDB ID 1U8C [23]). αv is shown in blue ribbon diagram, and β3 is shown in salmon-colored surface representation, with specific domains circled. Residues corresponding to the ReoPro binding site are shown in green, as in panel A, and D39 is shown in yellow. Regions corresponding to 6C5/D12-eluted peptides with P values of ≤0.0005 for alignment with integrin β3 (highlighted in panel A) are shown in blue, and those corresponding to 6B9/F5-eluted peptides with P values of ≤0.0005 for alignment with integrin β3 are shown in red. Alignment of peptide PLASTRT (P value of 0.0040) adjacent to D39 of the PSI domain is shown in magenta. Graphics were prepared using Pymol (DeLano Scientific LLC, San Carlos, CA).Of the 41 peptide-bearing phage eluted with the anti-Gc antibody 6C5/D12, 14 showed sequence homology to integrin β3 (P < 0.05), 4 of which had P values of ≤0.0005 (Fig. (Fig.1A).1A). Of the latter, sequence CTTMTRMTC mapped to the base of the I-like domain (Fig. (Fig.1B),1B), while CHGVYALHC and CRDTTPWWC mapped to the EGF-3 domain. Finally, sequence CTPTMHNHC mapped to the linker region untraceable in the crystal structure. Therefore, in contrast to peptide sequences identified by competition with the anti-Gn antibody, sequences identified by competition with the anti-Gc antibody 6C5/D12 appear to be mostly unrelated to integrin β3.As a low level of pathogenic hantavirus infection can be seen in cells lacking integrin β3, such as CHO cells (19), we asked if any of the identified peptide sequences could represent a previously unidentified receptor. We used the Basic Local Alignment Search Tool to search a current database of human protein sequences for potential alternate receptors represented by these peptides. However, none of the alignments identified proteins that are expressed at the cell surface, eliminating them as potential candidates for alternate viral entry receptors. This suggests that the majority of the peptides identified here likely represent novel sequences for binding ANDV surface glycoproteins.To determine whether synthetic peptides would also block infection, we synthesized cyclic peptides based on the 10 most-potent peptide-bearing phage. These peptides, in the context of phage presentation, showed levels of inhibition ranging from 44 to 72% (Table (Table2).2). When tested by IFA at 1 mM, four of the synthetic peptides showed inhibition levels significantly lower than those of the same peptide presented in the context of phage. This is not surprising, as steric factors due to the size of the phage and the multivalent presentation of peptide in the context of phage may both contribute to infection inhibition (8). However, there was no significant difference in inhibition by synthetic peptide versus peptide-bearing phage for six of the sequences, implying that inhibition in the context of phage was due solely to the nature of the peptide itself and not to steric factors or valency considerations contributed by the phage, which contrasts with our previous results, determined by using phage directed against αvβ3 integrin (10).

TABLE 2.

Synthetic cyclic peptides inhibit ANDV infection
TargetSample% Inhibition bya:
Peptide-bearing phageSynthetic peptide
GnCMQSAAAHC48.88 (44.40)59.66 (11.17)
GcCTVGPTRSC57.30 (11.31)46.47 (7.61)
GnCPSNVNNIC61.11 (25.41)44.14 (10.74)
GnCEKLHTASC43.60 (27.92)34.87 (9.26)
GcCPKLHPGGC71.88 (27.11)30.95 (7.73)b
GnCSLHSHKGC45.11 (49.81)29.79 (9.34)
GcCPMSQNPTC65.60 (13.49)18.19 (8.55)b
GnCKSLGSSQC53.90 (13.34)18.10 (7.55)b
GnCNSHSPVHC45.40 (28.80)15.52 (10.48)
GnCPAASHPRC51.84 (17.09)0 (10.72)b
Integrin β3ReoPro80.10 (7.72)
Gn6B9/F5 antibody42.72 (6.75)
Gc6C5/D12 antibody31.04 (7.81)
Open in a separate windowaStandard deviations of the results of at least four experiments are shown in parentheses.bMean percent inhibition between phage and synthetic peptide differs significantly (P < 0.05).The three most-potent synthetic peptides were examined for their ability to inhibit ANDV entry in a dose-dependent manner. The concentration of each peptide that produces 50% of its maximum potential inhibitory effect was determined. As shown in Fig. Fig.2A,2A, the 50% inhibitory concentration for each of the peptides was in the range of 10 μM, which from our experience is a reasonable potency for a lead compound to take forward for optimization.Open in a separate windowFIG. 2.Activities of synthetic peptides in inhibition of ANDV infection in vitro. (A) Peptides were examined for their ability to block ANDV infection of Vero E6 cells in a dose-dependent manner by IFA. (B) Peptides were tested in parallel for the ability to block infection of Vero E6 cells by ANDV, SNV, HTNV, and PHV. (C) Peptides were tested, singly or in combination, for the ability to block ANDV infection of Vero E6 cells. For all experiments, controls included media, ReoPro at 80 μg/ml, and monoclonal antibodies 6C5/D12 and 6B9/F5 at 5 μg/ml. All peptides were used at 1 mM. Data points represent n = 2 to 6, with error bars showing the standard errors of the means. Statistical analyses were performed on replicate samples using an unpaired Student''s t test.In order to determine the specificity of the three most-potent synthetic cyclic peptides in blocking ANDV, we examined them for inhibition of ANDV infection versus two other pathogenic hantaviruses, SNV and Hantaan virus (HTNV), or the nonpathogenic hantavirus Prospect Hill virus (PHV). As shown in Fig. Fig.2B,2B, ReoPro, which binds integrin β3, showed inhibition of infection by each of the pathogenic hantavirus strains, known to enter cells via β3, but not the nonpathogenic PHV, which enters via integrin β1 (6, 7). In contrast, peptides selected for the ability to bind ANDV were highly specific inhibitors of ANDV versus SNV, HTNV, or PHV. The specificities of peptides eluted by the anti-Gn monoclonal antibody are not surprising, as they are likely due to global differences in the Gn amino acid sequence. Specifically, sequence homologies between ANDV and SNV, HTNV, and PHV are 61%, 36%, and 51%, respectively, for the region corresponding to the immunogen for antibody 6B9/F5. Although homology between the immunogen for antibody 6C5/D12 and the corresponding Gc region of these viruses is somewhat higher (82% with SNV, 63% with HTNV, and 71% with PHV), the possibility that the monoclonal antibody used here recognizes a three-dimensional epitope lends itself to the high specificity of the peptides.The current model for cellular infection by hantaviruses (14) is as follows. Viral binding of the host cell surface target integrin is followed by receptor-mediated endocytosis and endosome acidification. Lowered pH induces conformational changes in Gn and/or Gc, which facilitate membrane fusion and viral release into the cytosol. As there is currently little information available about whether one glycoprotein is dominant in mediating infection, and as neutralizing epitopes have been found on both Gn and Gc glycoproteins (1, 4, 12, 13, 20), we examined whether combining anti-Gn- and anti-Gc-targeted synthetic peptides would lead to an increased infection blockade compared to those for single treatments. As shown in Fig. Fig.2C,2C, the combination of anti-Gn and anti-Gc peptides CMQSAAAHC and CTVGPTRSC resulted in a significant increase in infection inhibition (P = 0.0207 for CMQSAAAHC, and P = 0.0308 for CTVGPTRSC) compared to that resulting from single treatments. Although the high specificity of the peptides for ANDV makes it unlikely that this combination treatment will lead to more cross-reactivity with other pathogenic hantaviruses, this can be determined only by additional testing. Regardless, these data suggest a unique role for each of these viral proteins in the infection process as well as the benefits of targeting multiple viral epitopes for preventing infection.To our knowledge, the peptides reported here are the first identified that directly target ANDV, and this work further illustrates the power of coupling phage display and selective elution techniques in the identification of novel peptide sequences capable of specific protein-protein interactions from a large, random pool of peptide sequences. These novel peptide inhibitors (R. S. Larson, P. R. Hall, H. Njus, and B. Hjelle, U.S. patent application 61/205,211) provide leads for the development of more-potent peptide or nonpeptide organics for therapeutic use against HCPS.  相似文献   

7.
Basal cell carcinoma (BCC) is a very common malignant skin tumor that rarely metastatizes, but is often locally aggressive. Several factors, like large size (more than 3 cm), exposure to ultraviolet rays, histological variants, level of infiltration and perineural or perivascular invasion, are associated with a more aggressive clinical course. These morphological features seem to be more determinant in mideface localized BCC, which frequently show a significantly higher recurrence rate. An immunohistochemical profile, characterized by reactivity of tumor cells for p53, Ki67 and alpha-SMA has been associated with a more aggressive behaviour in large BCCs. The aim of this study was to verify if also little (<3 cm) basal cell carcinomas can express immunohistochemical markers typical for an aggressive behaviour.Basal cell carcinoma (BCC) is a very common malignant skin tumor that rarely metastatizes, even If Is often locally aggressive. Several factors, like large size (more than 3 cm), face localization, exposure to ultraviolet rays, histological variants, infiltration level and perineural or perivascular invasion, are associated with a more aggressive clinical course. In particular, the incidence of metastasis and/or death correlates with tumors greater than 3 cm in diameter in which setting patients are said to have 1–2 % risk of metastases that increases to 20–25% in lesions greater than 5 cm and to 50% in lesions greater than 10 cm in diameter (Snow et al., 1994). Histologically morpheiform, keratotic types and infiltrative growth of BCC are also considered features of the most aggressive course (Crowson, 2006). This can be explained by the fact that both the superficial and nodular variants of BCC are surrounded by a continuous basement membrane zone comprising collagens type IV and V admixed with laminin, while the aggressive growth variants (i.e. morpheiform, metatypical, and infiltrative growth subtypes) manifest the absence of basement membrane (Barsky et al., 1987).The molecular markers which characterize aggressive BCC include: increased expression of stromolysin (MMP-3) and collagenase-1 (MMP-1) (Cribier et al., 2001), decreased expression of syndecan-1 proteoglycan (Bayer-Garner et al., 2000) and of anti-apoptotic protein bcl-2 (Ramdial et al., 2000; Staibano et al., 2001).C-ras , c-fos (Urabe et al., 1994; Van der Schroeff et al., 1990) and p53 tumor supressor gene mutations (Auepemikiate et al., 2002) are indicative of an aggressive course.Focusing upon bcl-2 and p53 expression in BCC, there have been numerous studies documenting the utility of bcl-2 as a marker of favourable clinical behaviour while p53 expression may be a feature of a more aggressive outcome (Ramdial et al., 2000; Staibano et al., 2001; Bozdogan et al., 2002).An increased expression of cytoskeletal microfilaments like α–smooth muscle actin, frequently found in invasive BCC subtypes (Jones JCR et al., 1989), may explain an enhanced tumor mobility and deep tissue invasion through the stroma. (Cristian et al., 2001; Law et al., 2003). The aim of this preliminary study was to verify if also little (<3 cm) basal cell carcinomas may express aggressive immunohistochemical markers like p53, Ki67 and alpha-SMA. We used 31 excisional BCCs with tumor size less than 2 cm (ranging from 2 up to 20 mm) and with different skin localization (19 in the face, 6 in the trunk and 6 in the body extremities). All cases were immunostained for p53, BCL2, Ki67 and alpha-smooth muscle actin (α-SMA) (
AgeSexLocationHystotypeMax.DimDepthUlcEssInfp53Bcl-2Ki67AML
161MExtrKeratotic10×81No+++URD+++++-
261MFaceAdenoid10×94No+URD+++---
364MExtrSup mult11×130.8No+DRD+---
473MFaceNodular10×82Yes+DRD+++++++++
584MFaceNodular9×122Yes+DRD----
684MFaceAdenoid50.8No+URD+++---
784MExtrNodular13×103No+DRD+++++-
852FFaceNodular40.8No+URD+++-
976FFaceAdenoid10×44No+DRD+++-++-
1077FFaceMorph8×61Yes+++DRD+++---
1186MFaceMorph81Yes+DRD+++-++
1263FFaceAdenoid41No+URD+++++
1376FFaceNodular71.5No+DRD++++++-
1484MFaceNodular114Yes+++DRD+--+
1563FFaceKeratotic10×61.8No++DRD-+++-
1668FTrunkSup mult10×60.7No++URD++--
1767MFaceSup mult12×60.4No+URD+-+-
1867MExtrSup mult4×30.3No+URD+++++-
1932FExtrSup mult1×30.4No+URD+++-
2045MTrunkNodular7×52Yes+++URD+++-
2162MTrunkSup mult11×70.9No++URD-++-++
2265MTrunkAdenoid7×61.5No+URD+++++-
2372MTrunkNodular12×61No+URD+++-++
2486FFaceKeratotic20×113.1No++DRD+++-
2585MFaceNodular0.51.3No++DRD++++-
2674FExtrNodular4×40.9No+URD--+-
2771MFaceNodular6×121.7No+DRD--+-
2864FTrunkSup mult1.3×1.50.4No++URD+++---
2978FFaceNodular4×31.5No++DRD+++-+++
3080MFaceKeratotic4×41.6Yes+DRD--++++
Open in a separate window Our data show that p53 (75%), Bcl2 (50%) and Ki67 (63%) positivity was generally diffuse in the majority of cases. On the contrary, cytoplasmatic α-SMA expression was present only in 8 out of 31 cases (25,8%). All these 8 α-SMA positive BCCs, prevalently found in the mideface (6 out of 8), were characterized by an initial invasion beyond the dermis. Among these 6 face-localized α-SMA positive BCCs, 1 showed a sclerosing aggressive histotype, 1 a keratotic type and 4 a nodular histotype.These 8 little α-SMA-positive BCCs, compared to the others 23 α-SMA negative samples, all showed a major aggressiveness features: facial location, ulceration, morpheiform histotype and deeper infiltration into the dermis (Location
Histotype
Local aggressiveness
Immunohistochemistry
FaceKeratoticMorpheiformDepht of invasion Mean value(mm)UlcerationInfiltration of the dermisP53Bcl-2Ki678 α-SMA Positive cases75%12%12%1.650%63%75%50%63%23 α-SMA Negative cases56%13%4%1.413%48%78%43%65%
Open in a separate windowGiven the absence of a specific difference between α-SMA positive cases and α-SMA negative cases in the expression of aggressive immunohistochemical markers, except for a light reduction of bcl-2 in the α-SMA positive group (and2).2). By the analysis of the data, we selected the combination that could better define an aggressive behaviour even for little BCC: α-SMA, p53, Ki67 positivity and bcl-2 negativity. We considered p53 and ki67 markers of proliferation and cell-cycle alteration, combined with a loss of apoptotic activity expressed by Bcl-2 negativity, quite characteristic of aggressiveness; moreover α-SMA positivity probably reflects invasive potential and acquired mobility by neoplastic cells.This immunohistochemical profile (α-SMA, p53, Ki67 positivity and bcl-2 negativity) in our cases of BCC is present in two of them; one is a morpheiform BCC, that is an aggressive variant, while the other one is a nodular subtype (less aggressive).Therefore, our preliminary data suggest that only α-SMA positivity should be considered as an early diagnostic marker of potential aggressiveness in little BCC: all α-SMA positive little BCC in fact showed clinical and histological features of aggressiveness. Invasive potential is probably acquired by some BCCs not only when they reach large size, but it is probably present also when they have still little size, and can be revealed by α-SMA positivity in the neoplastic cells. Open in a separate windowFigure 1BCC, nodular type, HE, 10×. Open in a separate windowFigure 2BCC, nodular type, α-SMA positivity, 10×.  相似文献   

8.
Spontaneous Quinolone Resistance in the Zoonotic Serovar of Vibrio vulnificus     
Francisco J. Roig  A. Llorens  B. Fouz  C. Amaro 《Applied and environmental microbiology》2009,75(8):2577-2580
This work demonstrates that Vibrio vulnificus biotype 2, serovar E, an eel pathogen able to infect humans, can become resistant to quinolone by specific mutations in gyrA (substitution of isoleucine for serine at position 83) and to some fluoroquinolones by additional mutations in parC (substitution of lysine for serine at position 85). Thus, to avoid the selection of resistant strains that are potentially pathogenic for humans, antibiotics other than quinolones must be used to treat vibriosis on farms.Vibrio vulnificus is an aquatic bacterium from warm and tropical ecosystems that causes vibriosis in humans and fish (http://www.cdc.gov/nczved/dfbmd/disease_listing/vibriov_gi.html) (33). The species is heterogeneous and has been subdivided into three biotypes and more than eight serovars (6, 15, 33; our unpublished results). While biotypes 1 and 3 are innocuous for fish, biotype 2 can infect nonimmune fish, mainly eels, by colonizing the gills, invading the bloodstream, and causing death by septicemia (23). The disease is rapidly transmitted through water and can result in significant economic losses to fish farmers. Surviving eels are immune to the disease and can act as carriers, transmitting vibriosis between farms. Interestingly, biotype 2 isolates belonging to serovar E have been isolated from human infections, suggesting that serovar E is zoonotic (2). This serovar is also the most virulent for fish and has been responsible for the closure of several farms due to massive losses of fish. A vaccine, named Vulnivaccine, has been developed from serovar E isolates and has been successfully tested in the field (14). Although the vaccine provides fish with long-term protection from vibriosis, at present its use is restricted to Spain. For this reason, in many fish farms around the world, vibriosis is treated with antibiotics, which are usually added to the food or water.Quinolones are considered the most effective antibiotics against human and fish vibriosis (19, 21, 31). These antibiotics can persist for a long time in the environment (20), which could favor the emergence of resistant strains under selective pressure. In fact, spontaneous resistances to quinolones by chromosomal mutations have been described for some gram-negative bacteria (10, 11, 17, 24, 25, 26). Therefore, improper antibiotic treatment of eel vibriosis or inadequate residue elimination at farms could favor the emergence of human-pathogenic serovar E strains resistant to quinolones by spontaneous mutations. Thus, the main objective of the present work was to find out if the zoonotic serovar of biotype 2 can become quinolone resistant under selective pressure and determine the molecular basis of this resistance.Very few reports on resistance to antibiotics in V. vulnificus have been published; most of them have been performed with biotype 1 isolates. For this reason, the first task of this study was to determine the antibiotic resistance patterns in a wide collection of V. vulnificus strains belonging to the three biotypes that had been isolated worldwide from different sources (see Table S1 in the supplemental material). Isolates were screened for antimicrobial susceptibility to the antibiotics listed in Table S1 in the supplemental material by the agar diffusion disk procedure of Bauer et al. (5), according to the standard guideline (9). The resistance pattern found for each isolate is shown in Table S1 in the supplemental material. Less than 14% of isolates were sensitive to all the antibiotics tested, and more than 65% were resistant to more than one antibiotic, irrespective of their biotypes or serovars. The most frequent resistances were to ampicillin-sulbactam (SAM; 65.6% of the strains) and nitrofurantoin (F; 60.8% of the strains), and the least frequent were to tetracycline (12%) and oxytetracycline (8%). In addition, 15% of the strains were resistant to nalidixic acid (NAL) and oxolinic acid (OA), and 75% of these strains came from fish farms (see Table S1 in the supplemental material). Thus, high percentages of strains of the three biotypes were shown to be resistant to one or more antibiotics, with percentages similar to those found in nonbiotyped environmental V. vulnificus isolates from Asia and North America (4, 27, 34). In those studies, resistance to antibiotics could not be related to human contamination. However, the percentage of quinolone-resistant strains found in our study is higher than that reported in other ones, probably due to the inclusion of fish farm isolates, where the majority of quinolone-resistant strains were concentrated. This fact suggests that quinolone resistance could be related to human contamination due to the improper use of these drugs in therapy against fish diseases, as has been previously suggested (18, 20). Although no specific resistance pattern was associated with particular biotypes or serovars, we found certain differences in resistance distribution, as shown in Table Table1.1. In this respect, biotype 3 displayed the narrowest spectrum of resistances and biotype 1 the widest. The latter biotype encompassed the highest number of strains with multiresistance (see Table S1 in the supplemental material). Within biotype 2, there were differences among serovars, with quinolone resistance being restricted to the zoonotic serovar (Table (Table11).

TABLE 1.

Percentage of resistant strains distributed by biotypes and serovars
V. vulnificusNo. of isolatesResistance distribution (%) for indicated antibiotica
SAMCTXENALFOTOASXT-TMPTE
Biotype 14975.524.514.330.683.78.230.628.68.2
Biotype 2 (whole)7258.313.912.54.247.29.74.24.213.9
Biotype 2
    Serovar E3630.312.139.127.315.29.1321.2
    Serovar A231009.118.2077.3009.14.6
    Nontypeable82914.325057.114.30014.3
    Serovar I5100202002020000
Biotype 3510002008000020
Open in a separate windowaCTX, cefotaxime; E, erythromycin; OT, oxytetracycline; SXT-TMP, sulfamethoxazole-trimethoprim; TE, tetracycline.The origin of resistance to quinolones in the zoonotic serovar was further investigated. To this end, spontaneous mutants of sensitive strains were selected from colonies growing within the inhibition halo around OA or NAL disks. Two strains (strain CG100 of biotype 1 and strain CECT 4604 of biotype 2, serovar E) developed isolated colonies within the inhibition zone. These colonies were purified, and maintenance of resistance was confirmed by serial incubations on medium without antibiotics. Using the disk diffusion method, CG100 was shown to be resistant to SAM and F and CECT 4604 to F (see Table S1 in the supplemental material). The MICs for OA, NAL, flumequine (UB), and ciprofloxacin (CIP) were determined by using the microplate assay according to the recommendations of the Clinical and Laboratory Standards Institute and the European Committee for Antimicrobial Susceptibility Testing of the European Society of Clinical Microbiology and Infectious Diseases (8, 12) and interpreted according to the European Committee for Antimicrobial Susceptibility Testing of the European Society of Clinical Microbiology and Infectious Diseases (13). The MICs for OA and NAL and for the fluoroquinolones UB and CIP exhibited by the mutants and their counterparts are shown in Table Table2.2. The inhibition zone diameters correlated well with MICs (data not shown). Mutants FR1, FR2, FR3, and FR4 were resistant to NAL and sensitive to the remaining quinolones, although they showed higher resistances than their parental strains (Table (Table2).2). Thus, these four mutants showed increases of 32- to 128-fold for NAL MICs, 4- to 8-fold for UB MICs, and 16-fold for CIP MICs (Table (Table2).2). The fifth mutant, FR5, was resistant to the two tested quinolones and to UB, a narrow-spectrum fluoroquinolone. This mutant, although sensitive to CIP, multiplied its MIC for this drug by 128 with respect to the parental strain (Table (Table22).

TABLE 2.

MICs for quinolones and fluoroquinolones and mutations in gyrA, gyrB, and parC detected in naturally and artificially induced resistant strains
Strain(s)MIC (μg ml−1) for indicated antibioticb
Gene mutationa
gyrA
gyrB
parC
Position
Codon changeaa changePosition
Codon changeaa changePosition
Codon changeaa change
NALOAUBCIPntaantaantaa
CG1000.5 (S)0.125 (S)0.0625 (S)0.0078 (S)
FR116 (R)1 (S)0.25 (S)0.125 (S)24883AGT→ATTS→INCNCNCNCNCNCNCNC
FR216 (R)1 (S)0.25 (S)0.125 (S)24883AGT→ATTS→INCNCNCNCNCNCNCNC
CECT 46040.25 (S)0.0625 (S)0.0625 (S)0.0078 (S)
FR332 (R)2 (S)0.5 (S)0.125 (S)24883AGT→ATTS→INCNCNCNCNCNCNCNC
FR432 (R)2 (S)0.5 (S)0.125 (S)24883AGT→ATTS→INCNCNCNCNCNCNCNC
FR5256 (R)16 (R)16 (R)1 (S)24883AGT→ATTS→I1156386GCA→ACAA→T25485TCA→TTAS→L
1236412CAG→CACQ→H
CECT 4602128 (R)8 (R)64 (R)1 (S)24883AGT→ATTS→INCNCNCNC25485TCA→TTAS→L
CECT 4603, CECT 4606, CECT 4608, PD-5, PD-12, JE32 (R)2 (S)<1 (S)<1 (S)24883AGT→ATTS→INCNCNCNCNCNCNCNC
CECT 486264 (R)2 (S)2 (S)<1 (S)24983AGT→AGAS→RNCNCNCNCNCNCNCNC
A2, A4, A5, A6, A7, PD-1, PD-364-128 (R)2 (S)4 (S)<1 (S)24883AGT→ATTS→INCNCNCNC338113GCA→GTAA→V
V1128 (R)4 (S)4 (S)<1 (S)24883AGT→ATTS→I1274425GAG→GGGE→GNCNCNCNC
1314438AAC→AAAN→K
Open in a separate windowaMutations in a nucleotide (nt) that gave rise to a codon change and to a change in amino acids (aa) are indicated. NC, no change detected.bThe resistance (R) or sensitivity (S) against the antibiotic determined according to the Clinical and Laboratory Standards Institute and the European Committee for Antimicrobial Susceptibility Testing of the European Society of Clinical Microbiology and Infectious Diseases (9, 13) is indicated in parentheses.For other gram-negative pathogens, quinolone resistance relies on spontaneous mutations in the gyrA, gyrB, parC, and parE genes that occur in a specific region of the protein known as the quinolone resistance-determining region (QRDR) (1, 11, 17, 24, 25, 26, 28). To test the hypothesis that mutations in these genes could also produce quinolone resistance in V. vulnificus, the QRDRs of these genes were sequenced in the naturally resistant strains and in the two sensitive strains that had developed resistances by selective pressure in vitro. The genomic DNA was extracted (3), and the QRDRs of gyrA, gyrB, parE, and parC were amplified using the primers shown in Table Table3,3, which were designed from the published genomes of biotype 1 strains YJ016 and CMCP6 (7, 22). PCR products of the predicted size were sequenced in an ABI 3730 sequencer (Applied Biosystems). Analysis of the QRDR sequences for gyrA, gyrB, parC, and parE of the mutants and the naturally resistant strains revealed that all naturally resistant strains, except one, shared a specific mutation at nucleotide position 248 with the laboratory-induced mutants (Table (Table2).2). This mutation gave rise to a change from serine to isoleucine at amino acid position 83. The exception was a mutation in the adjacent nucleotide that gave rise to a substitution of arginine for serine at the same amino acid position (Table (Table2).2). All the isolates that were resistant to the quinolone NAL had a unique mutation in the gyrA gene, irrespective of whether resistance was acquired naturally or in the laboratory (Table (Table2).2). This result strongly suggests that a point mutation in gyrA that gives rise to a change in nucleotide position 83 can confer resistance to NAL in V. vulnificus biotypes 1 and 2 and that this mutation could be produced by selective pressure under natural conditions. gyrA mutations consisting of a change from serine 83 to isoleucine have also been described in isolates of Aeromonas from water (17) and in diseased fish isolates of Vibrio anguillarum (26). Similarly, replacement of serine by arginine at amino acid position 83 in diseased fish isolates of Yersinia ruckeri (16) suggests that this mechanism of quinolone resistance is widespread among gram-negative pathogens. In all cases, these single mutations were also related to increased resistance to other quinolones (OA) and fluoroquinolones (UB and CIP) (Table (Table2),2), although the mutants remained sensitive according to the standards of the Clinical and Laboratory Standards Institute and the European Committee for Antimicrobial Susceptibility Testing of the European Society of Clinical Microbiology and Infectious Diseases (9, 13). A total of 50% of the naturally resistant strains, all of them of biotype 1, showed additional mutations that affected parC (a change in amino acid position 113) or gyrB (changes in amino acids at positions 425 and 438) (Table (Table2).2). These strains exhibited higher MICs for OA and fluoroquinolones (Table (Table2),2), although they were still sensitive to these drugs (9, 13). Finally, one isolate of biotype 2, serovar E, which was naturally resistant to quinolones and UB, showed a mutation in parC that gave rise to a substitution of leucine for serine at amino acid position 85 (Table (Table2).2). This mutation was shared only with the laboratory-induced mutant, also a biotype 2, serovar E mutant, which was resistant to the fluoroquinolone UB. The same mutation in parC had been previously described in diseased fish isolates of V. anguillarum that were highly resistant to quinolones (28), but this had not been related to fluoroquinolone resistance in Vibrio spp. nor in other gram-negative bacteria. These results strongly suggest that resistance to fluoroquinolones in V. vulnificus is related to specific mutations in gyrA and parC and that mutations in different positions for parC or in gyrB could contribute to increased resistance to quinolones and fluoroquinolones. Our results also agree with previous studies confirming that the acquisition of higher quinolone resistance is more probable when arising from a gyrA parC double mutation than from a gyrA gyrB double mutation (29).

TABLE 3.

Oligonucleotides used in this study
PrimerSequenceAnnealing temp (°C)Size (bp)
GyrAFGGCAACGACTGGAATAAACC55.8416
GyrARCAGCCATCAATCACTTCCGTC
ParCFCGCAAGTTCACCGAAGATGC56.6411
ParCRGGCATCCGCAACTTCACG
GyrBFCGACTTCTGGTGACGATGCG57.4642
GyrBRGACCGATACCACAACCTAGTG
ParEFGCCAGGTAAGTTGACCGATTG56.8512
ParERCACCCAGACCTTTGAATCGTTG
Open in a separate windowFinally, the evolutionary history for each protein was inferred from previously published DNA sequences of the whole genes from different Vibrio species after multiple sequence alignment with MEGA4 software (32) by applying the neighbor-joining method (30) with the Poisson correction (35). The distance tree for each whole protein showed a topology similar to the phylogenetic tree based on 16S rRNA analysis, with the two isolates of V. vulnificus forming a single group, closely related to Vibrio parahaemolyticus, Vibrio cholerae, V. anguillarum, and Vibrio harveyi (see Fig. S1A in the supplemental material). A second analysis was performed with the QRDR sequences of the different mutants and isolates of V. vulnificus (GenBank accession numbers FJ379836 to FJ379927) to infer the intraspecies relationships (see Fig. S1B in the supplemental material). This analysis showed that QRDRs of gyrA, gyrB, parC, and parE were highly homogeneous within V. vulnificus.In summary, the zoonotic serovar of V. vulnificus can mutate spontaneously to gain quinolone resistance, under selective pressure in vitro, due to specific mutations in gyrA that involve a substitution of isoleucine for serine at amino acid position 83. This mutation appears in biotype 2, serovar E diseased-fish isolates and biotype 1 strains, mostly recovered from fish farms. An additional mutation in parC, resulting in a substitution of lysine for serine at amino acid position 85, seems to endow partial fluoroquinolone resistance on biotype 2, serovar E strains. This kind of double mutation is present in diseased-fish isolates of the zoonotic serovar but not in resistant biotype 1 isolates, which show different mutations in gyrB or in parC that increase their resistance levels but do not make the strains resistant to fluoroquinolones. Thus, antibiotics other than quinolones should be used at fish farms to prevent the emergence and spread of quinolone resistances, especially to CIP, a drug widely recommended for human vibriosis treatment.  相似文献   

9.
Dominant Bacteria and Biomass in the Kuytun 51 Glacier     
Shu-Rong Xiang  Tian-Cui Shang  Yong Chen  Ze-Fan Jing  Tandong Yao 《Applied and environmental microbiology》2009,75(22):7287-7290
  相似文献   

10.
Identification of a Polyketide Synthase Coding Sequence Specific for Anatoxin-a-Producing Oscillatoria Cyanobacteria     
Sabrina Cadel-Six  Isabelle Iteman  Caroline Peyraud-Thomas  Stéphane Mann  Olivier Ploux  Annick Méjean 《Applied and environmental microbiology》2009,75(14):4909-4912
  相似文献   

11.
Intrathecal Humoral Responses Are Inversely Associated with the Frequency of Simian Immunodeficiency Virus Macrophage-Tropic Variants in the Central Nervous System     
Elena Ryzhova  Pyone Aye  Tom Harvey  Wei Cao  Andrew Lackner  Francisco González-Scarano 《Journal of virology》2009,83(16):8282-8288
  相似文献   

12.
Impact of Viral Dose and Major Histocompatibility Complex Class IB Haplotype on Viral Outcome in Mauritian Cynomolgus Monkeys Vaccinated with Tat upon Challenge with Simian/Human Immunodeficiency Virus SHIV89.6P     
Aurelio Cafaro  Stefania Bellino  Fausto Titti  Maria Teresa Maggiorella  Leonardo Sernicola  Roger W. Wiseman  David Venzon  Julie A. Karl  David O'Connor  Paolo Monini  Marjorie Robert-Guroff  Barbara Ensoli 《Journal of virology》2010,84(17):8953-8958
The effects of the challenge dose and major histocompatibility complex (MHC) class IB alleles were analyzed in 112 Mauritian cynomolgus monkeys vaccinated (n = 67) or not vaccinated (n = 45) with Tat and challenged with simian/human immunodeficiency virus (SHIV) 89.6Pcy243. In the controls, the challenge dose (10 to 20 50% monkey infectious doses [MID50]) or MHC did not affect susceptibility to infection, peak viral load, or acute CD4 T-cell loss, whereas in the chronic phase of infection, the H1 haplotype correlated with a high viral load (P = 0.0280) and CD4 loss (P = 0.0343). Vaccination reduced the rate of infection acquisition at 10 MID50 (P < 0.0001), and contained acute CD4 loss at 15 MID50 (P = 0.0099). Haplotypes H2 and H6 were correlated with increased susceptibility (P = 0.0199) and resistance (P = 0.0087) to infection, respectively. Vaccination also contained CD4 depletion (P = 0.0391) during chronic infection, independently of the challenge dose or haplotype.Advances in typing of the major histocompatibility complex (MHC) of Mauritian cynomolgus macaques (14, 20, 26) have provided the opportunity to address the influence of host factors on vaccine studies (13). Retrospective analysis of 22 macaques vaccinated with Tat or a Tat-expressing adenoviral vector revealed that monkeys with the H6 or H3 MHC class IB haplotype were overrepresented among aviremic or controller animals, whereas macaques with the H2 or H5 haplotype clustered in the noncontrollers (12). More recently, the H6 haplotype was reported to correlate with control of chronic infection with simian immunodeficiency virus (SIV) mac251, regardless of vaccination (18).Here, we performed a retrospective analysis of 112 Mauritian cynomolgus macaques, which included the 22 animals studied previously (12), to evaluate the impact of the challenge dose and class IB haplotype on the acquisition and severity of simian/human immunodeficiency virus (SHIV) 89.6Pcy243 infection in 45 control monkeys and 67 monkeys vaccinated with Tat from different protocols (Table (Table11).

TABLE 1.

Summary of treatment, challenge dose, and outcome of infection in cynomolgus monkeys
Protocol codeNo. of monkeysImmunogen (dose)aAdjuvantbSchedule of immunization (wk)RoutecChallenged (MID50)Virological outcomee
Reference(s) or source
ACV
ISS-ST6Tat (10)Alum or RIBI0, 2, 6, 12, 15, 21, 28, 32, 36s.c., i.m.104114, 17
ISS-ST1Tat (6)None0, 5, 12, 17, 22, 27, 32, 38, 42, 48i.d.101004, 17
ISS-PCV3pCV-tat (1 mg)Bupivacaine + methylparaben0, 2, 6, 11, 15, 21, 28, 32, 36i.m.103006
ISS-ID3Tat (6)none0, 4, 8, 12, 16, 20, 24, 28, 39, 43, 60i.d.10111B. Ensoli, unpublished data
ISS-TR6Tat (10)Alum-Iscom0, 2, 6, 11, 16, 21, 28, 32, 36s.c., i.d., i.m.10420Ensoli, unpublished
ISS-TGf3Tat (10)Alum0, 4, 12, 22s.c.1503Ensoli, unpublished
ISS-TG3Tatcys22 (10)Alum1503Ensoli, unpublished
ISS-TG4Tatcys22 (10) + Gag (60)Alum1504Ensoli, unpublished
ISS-TG4Tat (10) + Gag (60)Alum1504Ensoli, unpublished
ISS-MP3Tat (10)H1D-Alum0, 4, 12, 18, 21, 38s.c., i.m.15021Ensoli, unpublished
ISS-MP3Tat (10)Alums.c.15003Ensoli, unpublished
ISS-GS6Tat (10)H1D-Alum0, 4, 12, 18, 21, 36s.c., i.m.15132Ensoli, unpublished
NCI-Ad-tat/Tat7Ad-tat (5 × 108 PFU), Tat (10)Alum0, 12, 24, 36i.n., i.t., s.c.15232Ensoli, unpublished
NCI-Tat9Tat (6 and 10)Alum/Iscom0, 2, 6, 11, 15, 21, 28, 32, 36s.c., i.d., i.m.1524312
ISS-NPT3pCV-tat (1 mg)Bupivacaine + methylparaben-Iscom0, 2, 8, 13, 17, 22, 28, 46, 71i.m.20003Ensoli, unpublished
ISS-NPT3pCV-tatcys22 (1 mg)Bupivacaine + methylparaben-Iscom0, 2, 8, 13, 17, 22, 28, 46, 71i.m.20111
    Total vaccinated67191731
        Naive11NoneNoneNAgNA10 or 15137
        Control34None, Ad, or pCV-0Alum, RIBI, H1D, Iscom or bupivacaine + methylparaben-Iscoms.c., i.d., i.n., i.t., i.m.10, 15, or 2051316
    Total controls4561623
    Total112253354
Open in a separate windowaAll animals were inoculated with the indicated dose of Tat plasmid DNA (pCV-tat [8], adenovirus-tat [Ad-tat] [27]) or protein, Gag protein, or empty vectors (pCV-0, adenovirus [Ad]) by the indicated route. Doses are in micrograms unless indicated otherwise.bAlum, aluminum phosphate (4); RIBI oil-in-water emulsions containing squalene, bacterial monophosphoryl lipid A, and refined mycobacterial products (4); Iscom, immune-stimulating complex (4); H1D are biocompatible anionic polymeric microparticles used for vaccine delivery (10, 12, 25a).cs.c., subcutaneous; i.m., intramuscular; i.d., intradermal; i.n., intranasal; i.t., intratracheal.dAll animals were inoculated intravenously with the indicated dose of the same SHIV89.6.Pcy243 stock.eAccording to the virological outcome upon challenge, monkeys were grouped as aviremic (A), controllers (C), or viremic (V).fBecause of the short follow-up, controller status could not be determined and all infected monkeys of the ISS-TG protocol were therefore considered viremic.gNA, not applicable.  相似文献   

13.
Increased Crystalline Cellulose Activity via Combinations of Amino Acid Changes in the Family 9 Catalytic Domain and Family 3c Cellulose Binding Module of Thermobifida fusca Cel9A     
Yongchao Li  Diana C. Irwin  David B. Wilson 《Applied and environmental microbiology》2010,76(8):2582-2588
Amino acid modifications of the Thermobifida fusca Cel9A-68 catalytic domain or carbohydrate binding module 3c (CBM3c) were combined to create enzymes with changed amino acids in both domains. Bacterial crystalline cellulose (BC) and swollen cellulose (SWC) assays of the expressed and purified enzymes showed that three combinations resulted in 150% and 200% increased activity, respectively, and also increased synergistic activity with other cellulases. Several other combinations resulted in drastically lowered activity, giving insight into the need for a balance between the binding in the catalytic cleft on either side of the cleavage site, as well as coordination between binding affinity for the catalytic domain and CBM3c. The same combinations of amino acid variants in the whole enzyme, Cel9A-90, did not increase BC or SWC activity but did have higher filter paper (FP) activity at 12% digestion.Cellulases catalyze the breakdown of cellulose into simple sugars that can be fermented to ethanol. The large amount of natural cellulose available is an exciting potential source of fuels and chemicals. However, the detailed molecular mechanisms of crystalline cellulose degradation by glycoside hydrolases are still not well understood and their low efficiency is a major barrier to cellulosic ethanol production.Thermobifida fusca is a filamentous soil bacterium that grows at 50°C in defined medium and can utilize cellulose as its sole carbon source. It is a major degrader of plant cell walls in heated organic materials such as compost piles and rotting hay and produces a set of enzymes that includes six different cellulases, three xylanases, a xyloglucanase, and two CBM33 binding proteins (12). Among them are three endocellulases, Cel9B, Cel6A, and Cel5A (7, 8), two exocellulases, Cel48A and Cel6B (6, 19), and a processive endocellulase, Cel9A (5, 7).T. fusca Cel9A-90 (Uniprot P26221 and YP_290232) is a multidomain enzyme consisting of a family 9 catalytic domain (CD) rigidly attached by a short linker to a family 3c cellulose binding module (CBM3c), followed by a fibronectin III-like domain and a family 2 CBM (CBM2). Cel9A-68 consists of the family 9 CD and CBM3c. The crystal structure of this species (Fig. (Fig.1)1) was determined by X-ray crystallography at 1.9 Å resolution (Protein Data Bank [PDB] code 4tf4) (15). Previous work has shown that E424 is the catalytic acid and D58 is the catalytic base (11, 20). H125 and Y206 were shown to play an important role in activity by forming a hydrogen bonding network with D58, an important supporting residue, D55, and Glc(−1)O1. Several enzymes with amino acid changes in subsites Glc(−1) to Glc(−4) had less than 20% activity on bacterial cellulose (BC) and markedly reduced processivity. It was proposed that these modifications disturb the coordination between product release and the subsequent binding of a cellulose chain into subsites Glc(−1) to Glc(−4) (11). Another variant enzyme with a deletion of a group of amino acids forming a block at the end of the catalytic cleft, Cel9A-68 Δ(T245-L251)R252K (DEL), showed slightly improved filter paper (FP) activity and binding to BC (20).Open in a separate windowFIG. 1.Crystal structure of Cel9A-68 (PDB code 4tf4) showing the locations of the variant residues, catalytic acid E424, catalytic base D58, hydrogen bonding network residues D55, H125, and Y206, and six glucose residues, Glc(−4) to Glc(+2). Part of the linker is visible in dark blue.The CBM3c domain is critical for hydrolysis and processivity. Cel9A-51, an enzyme with the family 9 CD and the linker but without CBM3c, had low activity on carboxymethyl cellulose (CMC), BC, and swollen cellulose (SWC) and showed no processivity (4). The role of CBM3c was investigated by mutagenesis, and one modified enzyme, R557A/E559A, had impaired activity on all of these substrates but normal binding and processivity (11). Variants with changes at five other CBM3c residues were found to slightly lower the activity of the modified enzymes, while Cel9A-68 enzymes containing either F476A, D513A, or I514H were found to have slightly increased binding and processivity (11) (see Table Table1).1). In the present work, CBM3c has been investigated more extensively to identify residues involved in substrate binding and processivity, understand the role of CBM3c more clearly, and study the coordination between the CD and CBM3c. An additional goal was to combine amino acid variants showing increased crystalline cellulose activity to see if this further increased activity. Finally, we have investigated whether the changes that improved the activity of Cel9A-68 also enhanced the activity of intact Cel9A-90.

TABLE 1.

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

14.
Quantitative Real-Time PCR Assays for Sensitive Detection of Canada Goose-Specific Fecal Pollution in Water Sources     
B. Fremaux  T. Boa  C. K. Yost 《Applied and environmental microbiology》2010,76(14):4886-4889
Canada geese (Branta canadensis) are prevalent in North America and may contribute to fecal pollution of water systems where they congregate. This work provides two novel real-time PCR assays (CGOF1-Bac and CGOF2-Bac) allowing for the specific and sensitive detection of Bacteroides 16S rRNA gene markers present within Canada goose feces.The Canada goose (Branta canadensis) is a prevalent waterfowl species in North America. The population density of Canada geese has doubled during the past 15 years, and the population was estimated to be close to 3 million in 2007 (4). Canada geese often congregate within urban settings, likely due to available water sources, predator-free grasslands, and readily available food supplied by humans (6). They are suspected to contribute to pollution of aquatic environments due to the large amounts of fecal matter that can be transported into the water. This can create a public health threat if the fecal droppings contain pathogenic microorganisms (6, 7, 9, 10, 12, 13, 19). Therefore, tracking transient fecal pollution of water due to fecal inputs from waterfowl, such as Canada geese, is of importance for protecting public health.PCR detection of host-specific 16S rRNA gene sequences from Bacteroidales of fecal origin has been described as a promising microbial source-tracking (MST) approach due to its rapidity and high specificity (2, 3). Recently, Lu et al. (15) characterized the fecal microbial community from Canada geese by constructing a 16S rRNA gene sequence database using primers designed to amplify all bacterial 16S rRNA gene sequences. The authors reported that the majority of the 16S rRNA gene sequences obtained were related to Clostridia or Bacilli and to a lesser degree Bacteroidetes, which represent possible targets for host-specific source-tracking assays.The main objective of this study was to identify novel Bacteroidales 16S rRNA gene sequences that are specific to Canada goose feces and design primers and TaqMan fluorescent probes for sensitive and specific quantification of Canada goose fecal contamination in water sources.Primers 32F and 708R from Bernhard and Field (2) were used to construct a Bacteroidales-specific 16S rRNA gene clone library from Canada goose fecal samples (n = 15) collected from grass lawns surrounding Wascana Lake (Regina, SK, Canada) in May 2009 (for a detailed protocol, see File S1 in the supplemental material). Two hundred eighty-eight clones were randomly selected and subjected to DNA sequencing (at the Plant Biotechnology Institute DNA Technologies Unit, Saskatoon, SK, Canada). Representative sequences of each operational taxonomic unit (OTU) were recovered using an approach similar to that described by Mieszkin et al. (16). Sequences that were less than 93% similar to 16S rRNA gene sequences from nontarget host species in GenBank were used in multiple alignments to identify regions of DNA sequence that were putatively goose specific. Subsequently, two TaqMan fluorescent probe sets (targeting markers designated CGOF1-Bac and CGOF2-Bac) were designed using the RealTimeDesign software provided by Biosearch Technologies (http://www.biosearchtech.com/). The newly designed primer and probe set for the CGOF1-Bac assay included CG1F (5′-GTAGGCCGTGTTTTAAGTCAGC-3′) and CG1R (5′-AGTTCCGCCTGCCTTGTCTA-3′) and a TaqMan probe (5′-6-carboxyfluorescein [FAM]-CCGTGCCGTTATACTGAGACACTTGAG-Black Hole Quencher 1 [BHQ-1]-3′), and the CGOF2-Bac assay had primers CG2F (5′-ACTCAGGGATAGCCTTTCGA-3′) and CG2R (5′-ACCGATGAATCTTTCTTTGTCTCC-3′) and a TaqMan probe (5′-FAM-AATACCTGATGCCTTTGTTTCCCTGCA-BHQ-1-3′). Oligonucleotide specificities for the Canada goose-associated Bacteroides 16S rRNA primers were verified through in silico analysis using BLASTN (1) and the probe match program of the Ribosomal Database Project (release 10) (5). Host specificity was further confirmed using DNA extracts from 6 raw human sewage samples from various geographical locations in Saskatchewan and 386 fecal samples originating from 17 different animal species in Saskatchewan, including samples from Canada geese (n = 101) (Table (Table1).1). An existing nested PCR assay for detecting Canada goose feces (15) (targeting genetic marker CG-Prev f5) (see Table S1 in the supplemental material) was also tested for specificity using the individual fecal and raw sewage samples (Table (Table1).1). All fecal DNA extracts were obtained from 0.25 g of fecal material by using the PowerSoil DNA extraction kit (Mo Bio Inc., Carlsbad, CA) (File S1 in the supplemental material provides details on the sample collection).

TABLE 1.

Specificities of the CGOF1-Bac, CGOF2-Bac, and CG-Prev f5 PCR assays for different species present in Saskatchewan, Canada
Host group or sample typeNo. of samplesNo. positive for Bacteroidales marker:
CGOF1-BacCGOF2-BacCG-Prev f5All-Bac
Individual human feces2500125
Raw human sewage60006
Cows4100041
Pigs4800148
Chickens3400834
Geese10158515995a
Gulls1600614
Pigeons2510222
Ducks1000010
Swans10001
Moose1000010
Deer
    White tailed1000010
    Mule1000010
    Fallow1000010
Caribou1000010
Bison1000010
Goats1000010
Horses1500015
Total392595177381
Open in a separate windowaThe 6 goose samples that tested negative for the All-Bac marker also tested negative for the three goose markers.The majority of the Canada goose feces analyzed in this study (94%; 95 of 101) carried the Bacteroidales order-specific genetic marker designated All-Bac, with a relatively high median concentration of 8.2 log10 copies g1 wet feces (Table (Table11 and Fig. Fig.1).1). The high prevalence and abundance of Bacteroidales in Canada goose feces suggested that detecting members of this order could be useful in identifying fecal contamination associated with Canada goose populations.Open in a separate windowFIG. 1.Concentrations of the Bacteroidales (All-Bac, CGOF1-Bac, and CGOF2-Bac) genetic markers in feces from various individual Canada geese.The composition of the Bacteroidales community in Canada goose feces (n = 15) was found to be relatively diverse since 52 OTUs (with a cutoff of 98% similarity) were identified among 211 nonchimeric 16S rRNA gene sequences. Phylogenetic analysis of the 52 OTUs (labeled CGOF1 to CGOF52) revealed that 43 (representing 84% of the 16S rRNA gene sequences) were Bacteroides like and that 9 (representing 16% of the 16S rRNA gene sequences) were likely to be members of the Prevotella-specific cluster (see Fig. S2 in the supplemental material). Similarly, Jeter et al. (11) reported that 75.7% of the Bacteroidales 16S rRNA clone library sequences generated from goose fecal samples were Bacteroides like. The majority of the Bacteroides- and Prevotella-like OTUs were dispersed among a wide range of previously characterized sequences from various hosts and did not occur in distinct clusters suitable for the design of Canada goose-associated real-time quantitative PCR (qPCR) assays (see Fig. S2 in the supplemental material). However, two single Bacteroides-like OTU sequences (CGOF1 and CGOF2) contained putative goose-specific DNA regions that were identified by in silico analysis (using BLASTN, the probe match program of the Ribosomal Database Project, and multiple alignment). The primers and probe for the CGOF1-Bac and CGOF2-Bac assays were designed with no mismatches to the clones CGOF1 and CGOF2, respectively.The CGOF2-Bac assay demonstrated no cross-amplification with fecal DNA from other host groups, while cross-amplification for the CGOF1-Bac assay was limited to one pigeon fecal sample (1 of 25, i.e., 4% of the samples) (Table (Table1).1). Since the abundance in the pigeon sample was low (3.3 log10 marker copies g1 feces) and detection occurred late in the qPCR (with a threshold cycle [CT] value of 37.1), it is unlikely that this false amplification would negatively impact the use of the assay as a tool for detection of Canada goose-specific fecal pollution in environmental samples. In comparison, the nested PCR CG-Prev f5 assay described by Lu and colleagues (15) demonstrated non-host-specific DNA amplification with fecal DNA samples from several animals, including samples from humans, pigeons, gulls, and agriculturally relevant pigs and chickens (Table (Table11).Both CGOF1-Bac and CGOF2-Bac assays showed limits of quantification (less than 10 copies of target DNA per reaction) similar to those of other host-specific Bacteroidales real-time qPCR assays (14, 16, 18). The sensitivities of the CGOF1-Bac and CGOF2-Bac assays were 57% (with 58 of 101 samples testing positive) and 50% (with 51 of 101 samples testing positive) for Canada goose feces, respectively (Table (Table1).1). A similar sensitivity of 58% (with 59 of 101 samples testing positive) was obtained using the CG-Prev f5 PCR assay. The combined use of the three assays increased the detection level to 72% (73 of 101) (Fig. (Fig.2).2). Importantly, all markers were detected within groups of Canada goose feces collected each month from May to September, indicating relative temporal stability of the markers. The CG-Prev f5 PCR assay is an end point assay, and therefore the abundance of the gene marker in Canada goose fecal samples could not be determined. However, development of the CGOF1-Bac and CGOF2-Bac qPCR approach allowed for the quantification of the host-specific CGOF1-Bac and CGOF2-Bac markers. In the feces of some individual Canada geese, the concentrations of CGOF1-Bac and CGOF2-Bac were high, reaching levels up to 8.8 and 7.9 log10 copies g1, respectively (Fig. (Fig.11).Open in a separate windowFIG. 2.Venn diagram for Canada goose fecal samples testing positive with the CGOF1-Bac, CGOF2-Bac, and/or CG-Prev f5 PCR assay. The number outside the circles indicates the number of Canada goose fecal samples for which none of the markers were detected.The potential of the Canada goose-specific Bacteroides qPCR assays to detect Canada goose fecal pollution in an environmental context was tested using water samples collected weekly during September to November 2009 from 8 shoreline sampling sites at Wascana Lake (see File S1 and Fig. S1 in the supplemental material). Wascana Lake is an urban lake, located in the center of Regina, that is routinely frequented by Canada geese. In brief, a single water sample of approximately 1 liter was taken from the surface water at each sampling site. Each water sample was analyzed for Escherichia coli enumeration using the Colilert-18/Quanti-Tray detection system (IDEXX Laboratories, Westbrook, ME) (8) and subjected to DNA extraction (with a PowerSoil DNA extraction kit [Mo Bio Inc., Carlsbad, CA]) for the detection of Bacteroidales 16S rRNA genetic markers using the Bacteroidales order-specific (All-Bac) qPCR assay (14), the two Canada goose-specific (CGOF1-Bac and CGOF2-Bac) qPCR assays developed in this study, and the human-specific (BacH) qPCR assay (17). All real-time and conventional PCR procedures as well as subsequent data analysis are described in the supplemental material and methods. The E. coli and All-Bac quantification data demonstrated that Wascana Lake was regularly subjected to some form of fecal pollution (Table (Table2).2). The All-Bac genetic marker was consistently detected in high concentrations (6 to 7 log10 copies 100 ml1) in all the water samples, while E. coli concentrations fluctuated according to the sampling dates and sites, ranging from 0 to a most probable number (MPN) of more than 2,000 100 ml1. High concentrations of E. coli were consistently observed when near-shore water experienced strong wave action under windy conditions or when dense communities of birds were present at a given site and time point.

TABLE 2.

Levels of E. coli and incidences of the Canada goose-specific (CGOF1-Bac and CGOF2-Bac), human-specific (BacH), and generic (All-Bac) Bacteroidales 16S rRNA markers at the different Wascana Lake sites sampled weeklya
SiteE. coli
All-Bac
CGOF1-Bac
CGOF2-Bac
BacH
No. of positive water samples/total no. of samples analyzed (%)Min level-max level (MPN 100 ml−1)Mean level (MPN 100 ml−1)No. of positive water samples/total no. of samples analyzed (%)Min level-max level (log copies 100 ml−1)Mean level (log copies 100 ml−1)No. of positive water samples/total no. of samples analyzed (%)Min level-max level (log copies 100 ml−1)Mean level (log copies 100 ml−1)No. of positive water samples/total no. of samples analyzed (%)Min level-max level (log copies 100 ml−1)Mean level (log copies 100 ml−1)No. of positive water samples/total no. of samples analyzedMin level-max level (log copies 100 ml−1)Mean level (log copies 100 ml−1)
W18/8 (100)6-19671.18/8 (100)6.2-8.16.96/8 (75)0-4.72.44/8 (50)0-41.72/80-3.71.7
W29/10 (90)0-1,12019410/10 (100)5.8-6.86.49/10 (90)0-3.72.68/10 (80)0-3.32.20/1000
W310/10 (100)6-1,55053410/10 (100)6-7.8710/10 (100)2.9-4.83.810/10 (100)2-4.53.40/1000
W410/10 (100)16-1,73252910/10 (100)6.4-7.6710/10 (100)3.2-4.63.910/10 (100)2.8-4.33.40/1000
W510/10 (100)2-2,42068710/10 (100)5.5-6.96.37/10 (70)0-3.21.75/10 (50)0-3.11.20/1000
W610/10 (100)3-1,99038910/10 (100)5.5-76.39/10 (90)0-4.32.86/10 (60)0-5.121/100-3.41.3
W77/7 (100)5-2,4204457/7 (100)5.7-7.876/7 (86)0-3.82.65/7 (71)0-4.42.42/70-5.12.8
W810/10 (100)17-98016010/10 (100)6.3-8.67.18/10 (80)0-4.62.87/10 (70)0-4.42.30/1000
Open in a separate windowaMin, minimum; max, maximum.The frequent detection of the genetic markers CGOF1-Bac (in 65 of 75 water samples [87%]), CGOF2-Bac (in 55 of 75 samples [73%]), and CG-Prev f5 (in 60 of 75 samples [79%]) and the infrequent detection of the human-specific Bacteroidales 16S rRNA gene marker BacH (17) (in 5 of 75 water samples [7%[) confirmed that Canada geese significantly contributed to the fecal pollution in Wascana Lake during the sampling period. Highest mean concentrations of both CGOF1-Bac and CGOF2-Bac markers were obtained at the sampling sites W3 (3.8 and 3.9 log10 copies 100 ml1) and W4 (3.4 log10 copies 100 ml1 for both), which are heavily frequented by Canada geese (Table (Table2),2), further confirming their significant contribution to fecal pollution at these particular sites. It is worth noting that concentrations of the CGOF1-Bac and CGOF2-Bac markers in water samples displayed a significant positive relationship with each other (correlation coefficient = 0.87; P < 0.0001), supporting the accuracy of both assays for identifying Canada goose-associated fecal pollution in freshwater.In conclusion, the CGOF1-Bac and CGOF2-Bac qPCR assays developed in this study are efficient tools for estimating freshwater fecal inputs from Canada goose populations. Preliminary results obtained during the course of the present study also confirmed that Canada geese can serve as reservoirs of Salmonella and Campylobacter species (see Fig. S3 in the supplemental material). Therefore, future work will investigate the cooccurence of these enteric pathogens with the Canada goose fecal markers in the environment.  相似文献   

15.
Cross-Subtype Neutralization Sensitivity despite Monoclonal Antibody Resistance among Early Subtype A,C, and D Envelope Variants of Human Immunodeficiency Virus Type 1     
Catherine A. Blish  Zahra Jalalian-Lechak  Stephanie Rainwater  Minh-An Nguyen  Ozge C. Dogan  Julie Overbaugh 《Journal of virology》2009,83(15):7783-7788
The human immunodeficiency virus type 1 (HIV-1) variants that are transmitted to newly infected individuals are the primary targets of interventions, such as vaccines and microbicides, aimed at preventing new infections. Newly acquired subtype A, B, and C variants have been the focus of neutralization studies, although many of these viruses, particularly of subtypes A and B, represent viruses circulating more than a decade ago. In order to better represent the global diversity of transmitted HIV-1 variants, an additional 31 sexually transmitted Kenyan HIV-1 env genes, representing several recent infections with subtype A, as well as subtypes A/D, C, and D, were cloned, and their neutralization profiles were characterized. Most env variants were resistant to neutralization by the monoclonal antibodies (MAbs) b12, 4E10, 2F5, and 2G12, suggesting that targeting the epitopes of these MAbs may not be effective against variants that are spreading in areas of endemicity. However, significant cross-subtype neutralization by plasma was observed, indicating that there may be other epitopes, not yet defined by the limited available MAbs, which could be recognized more broadly.Most effective viral vaccines are thought to provide protection primarily by stimulating neutralizing antibodies (NAbs) to clear cell-free virus (25, 27). Because protection by NAbs requires recognition of common viral epitopes, the extreme genetic diversity of human immunodeficiency virus type 1 (HIV-1) presents a particular challenge to NAb-based vaccine approaches. Therefore, a critical starting point for studies of immune-mediated protection against HIV-1 is a collection of newly transmitted HIV-1 variants, particularly from areas of endemicity, such as sub-Saharan Africa, in order to determine whether vaccines are appropriately targeted to common epitopes from these relevant transmitted strains.During HIV-1 transmission, a bottleneck allows only one or a few variants to be transmitted to a newly infected individual (6, 9, 16, 29, 34, 37, 39), and the sensitivity of these early transmitted strains to antibody-mediated neutralization is therefore of particular interest. Newly transmitted HIV-1 variants have demonstrated significant heterogeneity in their neutralization phenotypes both within and between subtypes (2, 3, 6-8, 11, 13-15, 22, 30, 32, 36). Panels of sexually transmitted HIV-1 envelope variants (based on the envelope gene, env) have been characterized, including subtype B variants from North America, Trinidad, and Europe, subtype C variants from South Africa and Zambia, and subtype A variants from Kenya collected between 1994 and 1996 (2, 14, 15). Here, we characterize an additional 31 envelope variants from 14 subjects with sexually transmitted HIV-1 who were infected in Kenya, where subtypes A, C, and D circulate, between 1993 and 2005 (24, 31).The env genes were cloned from samples drawn 14 to 391 (median, 65) days postinfection from individuals enrolled in a prospective cohort of high-risk women in Mombasa, Kenya (19-21). Demographic characteristics of the subjects are summarized in Table Table1;1; the timing of first infection was determined by both HIV-1 serology and HIV RNA testing as described previously (12). All of the subjects were presumably infected by male-to-female transmission and displayed a range of plasma viral loads at the time of env gene cloning (Table (Table1).1). For most individuals, full-length env genes were cloned from uncultured peripheral blood mononuclear cell (PBMC) DNA, though for two individuals, clones were obtained from DNA following short-term coculture with donor PBMCs (Table (Table1).1). env genes were cloned by single-copy nested PCR with primers and PCR conditions as described previously (4, 17). We tested env genes for their ability to mediate infection by transfecting env plasmid DNA into 293T cells along with an env-deficient HIV-1 subtype A proviral plasmid, Q23Δenv, to make pseudoviral particles (17). More than 80 env clones were obtained from 16 subjects; less than one-half were functional on the basis of the infectivity of pseudoviral particles in a single-round infection of TZM-bl cells (AIDS Research and Reference Reagent Program, National Institutes of Health), as observed previously for env genes cloned from proviral sequences (17); a lower fraction of functional env genes have been reported from plasma (18). We focused on the proviral sequences here because they presumably best represent the sequence closest to that of the transmitted strains. The 31 functional env variants are described in Table Table11.

TABLE 1.

Demographic characteristics, diversities, gp120 variable-region lengths, numbers of PNGS, and accession numbers of cloned env variants
SubjectVirus subtypeSample date (mo/day/yr)dpiaPlasma VLbSourcecIndividual env clonePairwise difference (%)dVariable-loop length (aa)
No. of PNGS
GenBank accession no.
V1/V2V3V4V5gp120gp41gp41 ecto
QB726A04/16/967061,940ucPBMCQB726.70M.ENV.B30.16633536102244FJ866111
QB726.70M.ENV.C4633536102244FJ866112
QF495A05/16/0623217,050ucPBMCQF495.23M.ENV.A10.121073537113044FJ866113
QF495.23M.ENV.A31073537113044FJ866114
QF495.23M.ENV.B21133537113144FJ866115
QF495.23M.ENV.D11133537113144FJ866116
QG984A07/12/042130,300ucPBMCQG984.21M.ENV.A3NA693436112433FJ866117
QH209A10/13/051428,600ucPBMCQH209.14M.ENV.A2NA723529112444FJ866118
QH343A09/08/052140,750,000ucPBMCQH343.21M.ENV.A100.19773532152644FJ866119
QH343.21M.ENV.B5773532152644FJ866120
QH359A10/05/052132,120ucPBMCQH359.21M.ENV.C11.4843536102944FJ866121
QH359.21M.ENV.D1733535102644FJ866122
QH359.21M.ENV.E2723540132844FJ866123
QA790eA/D06/10/9620448,100ccPBMCQA790.204I.ENV.A40.36773533112544FJ866124
QA790.204I.ENV.C1773533112644FJ866125
QA790.204I.ENV.C8773533112444FJ866126
QA790.204I.ENV.E2773533112544FJ866127
QG393A2/D06/23/046017,360ucPBMCQG393.60M.ENV.A10.7603431102455FJ866128
QG393.60M.ENV.B7573431102455FJ866129
QG393.60M.ENV.B8573431102455FJ866130
QB099eC02/10/9539127,280ucPBMCQB099.391M.ENV.B10.43653529102544FJ866131
QB099.391M.ENV.C8653529102544FJ866132
QC406C07/08/9770692,320ucPBMCQC406.70M.ENV.F3NA643520112254FJ866133
QA013D10/11/95701,527,700ccPBMCQA013.70I.ENV.H10.16603429122544FJ866134
QA013.70I.ENV.M12603429122544FJ866135
QA465D08/19/935937,750ucPBMCQA465.59M.ENV.A10.24653530112844FJ866136
QA465.59M.ENV.D1653530112744FJ866137
QB857D10/16/9711014,640ucPBMCQB857.23I.ENV.B3NA683432112654FJ866138
QD435D04/06/9910017,470ucPBMCQD435.100M.ENV.A40.88693429122654FJ866139
QD435.100M.ENV.B5673429112454FJ866140
QD435.100M.ENV.E1693429122654FJ866141
Open in a separate windowadpi, days postinfection as defined by RNA testing (12).bVL, viral load on the sample date in which env genes were cloned.cucPBMC, uncultured PBMCs; ccPBMC, cocultured PBMCs.dAverage pairwise distance between the full-length env variants from a given subject. NA, not applicable because there was only one variant available from the subject.eenv variants from these two subjects were cloned from >6 months postinfection, as noted, and should not be considered true early env variants.The full-length, functional env genes were sequenced and aligned to generate a maximum likelihood phylogenetic tree with reference sequences from the Los Alamos National Laboratory HIV database, as described previously (26). Viral env clones from the same subject clustered together, and a wide spectrum of genetic diversity was observed overall (Fig. (Fig.1).1). Some women, such as subject QF495, were infected with a relatively homogeneous viral population, with average pairwise differences of only 0.12% between env variants (Table (Table11 and Fig. Fig.1).1). However, as observed previously in this cohort (16, 28, 29, 33-35), other individuals, such as subjects QH359 and QD435, were infected with more heterogeneous viral populations with average pairwise differences of 1.4% and 0.88% between variants, respectively (Table (Table11 and Fig. Fig.1).1). env genes from subtypes A (13 variants), C (3 variants), and D (8 variants), as well as A/D recombinants (4 variants) and A2/D recombinants (3 variants), were represented (Fig. (Fig.1).1). The viral subtypes were confirmed using the NCBI genotyping database (http://www.ncbi.nlm.nih.gov/).Open in a separate windowFIG. 1.Maximum likelihood phylogenetic tree of full-length sequences from early subtype A, C, D, and A/D recombinant env variants in Kenya. The 31 novel env clones from Kenyan early infections and reference sequences for subtypes A, B, C, D, and K from the Los Alamos HIV database (http://www.hiv.lanl.gov/content/index) are displayed. The phylogenetic tree was rooted with subtype K env sequences. Values at nodes indicate the percentage of bootstraps in which the cluster the right was found; only values of 70% or greater are shown.The deduced amino acid sequences revealed that all functional variants had an uninterrupted open reading frame in env except for variant QB099.391I.ENV.C8, which had a frameshift mutation within the cytoplasmic tail of gp41. There was significant heterogeneity in the length of the protein variable loops, particularly V1/V2, which ranged from 57 amino acids (aa) to 113 aa (Table (Table1).1). The V3, V4, and V5 loops also varied in length, though less dramatically (Table (Table1).1). Variants from the same subject were generally similar in their variable-loop lengths. Moderate variation was also observed in the number and position of potential N-linked glycosylation sites (PNGS) (Table (Table11).Previous analyses indicated that early subtype C env proteins had shorter variable loops than did early subtype B env proteins (13), suggesting that there are different env protein features between subtypes. Thus, to compare variable-loop lengths and the numbers of PNGS between subtypes using this expanded group of early env variants, we evaluated the 31 newly cloned variants plus an additional 15 subtype A variants (2), 19 subtype B variants (14), and 18 subtype C variants (15) from other early virus panels. In order to avoid bias, when more than one env variant was available from a subject, the average loop length or PNGS number for that subject''s env proteins was used. We did not observe significant differences in V1/V2 length, V5 length, or the numbers of PNGS between subtypes by the Kruskal-Wallis equality-of-populations rank test (Table (Table2)2) . However, there were significant differences between the V3 and V4 loop lengths of the subtypes after adjusting for multiple comparisons (Table (Table2).2). The differences in V3 length appeared to be a result of shorter V3 loops in subtype D env proteins than in early subtype B (P = 0.006) or C (P < 0.001) env proteins (Table (Table2).2). The differences in V4 length were caused by shorter V4 loops in subtype C env proteins in comparison to both subtype A and B env proteins (P < 0.001; Table Table22).

TABLE 2.

Summary of variable-loop lengths and the numbers of PNGS in gp120 and gp41 within early HIV-1 env variantsa
ParameterMedian value (25th percentile, 75th percentile) for subtype:
Kruskal- Wallis P valuebWilcoxon rank sum P values for individual comparisonsc
A (n = 11)B (n = 19)C (n = 20)D (n = 4)A vs. BA vs. CA vs. DB vs. CB vs. DC vs. D
Length
    V1/V270.3 (62, 76)70 (66, 70)65 (62, 76)66.5 (62, 69)0.210.7300.2820.2150.0510.1130.846
    V335 (34, 35)35 (35, 35)35 (34, 35)34 (34, 35)0.0010.2400.0160.1070.1410.006<0.001
    V432 (30, 36)33 (31, 34)26.5 (22, 29)29.5 (29, 31)0.00010.880<0.0010.148<0.0010.0230.056
    V511 (11, 11)10 (9, 11)10 (9, 11)11.5 (11, 12)0.0300.0960.0150.1840.6770.0990.021
No. of PNGS in:
    gp12024 (23, 28)25 (24, 26)24 (23, 25)26 (26, 27)0.200.6800.6920.2650.1460.1860.042
    gp414 (4, 5)5 (4, 5)5 (4, 5)4.5 (4, 5)0.200.0300.1790.4700.4100.4080.799
    gp41ecto4 (4, 4)4 (4, 4)4 (4, 5)4 (4, 4)0.0440.1070.0250.5500.0880.5070.201
Open in a separate windowaVariable-loop lengths and the numbers of PNGS in gp120 and gp41 within early HIV-1 env variants from subtypes A, B, C, and D characterized here and previously (2, 14, 15). n, number of samples.bKruskal-Wallis equality-of-populations rank test (based on multiple comparisons; P values of <0.007 were considered significant; significant values are presented in boldface).cWilcoxon rank sum test (based on multiple comparisons; P values of <0.008 were considered significant; significant values are presented in boldface).We then assessed the neutralization sensitivity of the pseudoviruses to antibodies in plasma from HIV-1-infected individuals and to HIV-1-specific MAbs by using the TZM-bl neutralization assay as described previously (2, 23, 38). Median inhibitory concentrations (IC50s) were defined as the reciprocal dilution of plasma or concentration of MAb that resulted in 50% inhibition of infection (2, 38). The Kenya pool was derived by pooling plasma collected between 1998 and 2000 from 30 HIV-1-infected individuals in Mombasa, Kenya, and the other three pools were derived by pooling plasma collected between 1993 and 1997 from 10 individuals from Nairobi, Kenya, and with an infection with a known subtype (A, C, or D) of HIV-1 as described previously (2).The env variants demonstrated a range of neutralization sensitivities to plasma samples, from neutralization resistant (defined as <50% neutralization with a 1:50 dilution of plasma) to neutralization sensitive with an IC50 of 333 (Fig. (Fig.2).2). Some clones, such as QF495.23M.ENV.A1, were relatively sensitive to all the plasma pools, with IC50s from 100 to 333, whereas other clones, such as QH343.21M.ENV.A10, were relatively resistant to these plasma pools, with IC50s from <50 to 85 (Fig. (Fig.2).2). The plasma pools did differ in their neutralization potencies. The Kenya pool, with a median IC50 of <50 across all viruses tested, was significantly less likely to neutralize these transmitted variants than were the subtype A, C, and D plasma pools, which had median IC50s of 110, 105, and 123, respectively (P values of <0.0001, 0.0001, and 0.001, respectively, by paired t test on log-transformed IC50s). The basis for these differences in neutralizing activity is not clear, although the location, timing, and level of immunodeficiency at the time of sample collection could have contributed to the differences in NAb levels between the pools.Open in a separate windowFIG. 2.Neutralization sensitivity of early subtype A, C, D, and A/D recombinant env variants to plasma samples and MAbs in relation to the sequences of the MAb binding sites. The env used to generate the pseudovirus tested is shown at the left, and the plasma pool or MAb tested is indicated at the top. The IC50s of each plasma sample or MAb against each viral pseudotype is shown, with darker shading indicating more potent neutralization, as defined at the bottom of the figure. Gray boxes indicate that <50% neutralization was observed at the highest dilution of plasma or concentration of MAb tested. Each IC50 shown is an average of the results from two independent neutralization assays, using pseudovirus generated in independent transfection experiments. The median IC50s from the 31 variants are shown at the bottom. Neutralization of the pseudovirus derived from the subtype B variant SF162 is shown as a control, and neutralizations of murine leukemia virus (MLV) and simian immunodeficiency virus clone 8 (SIV) are shown as negative controls. In the panels on the right, the sequences for the MAbs 2G12, 2F5, and 4E10 are displayed. For 2G12, the amino acid numbers for the five PNGS that are important for 2G12 binding are shown for each virus tested. A plus sign indicates that the PNGS at that site in the envelope sequence was preserved, and a minus sign indicates that the PNGS was deleted. A shift in the PNGS position is indicated by the amino acid position to which the PNGS shifted. All sequences were numbered relative to the HXB2 sequence. The two rightmost panels show data for the canonical 2F5 and 4E10 epitopes, with a period indicating that the amino acid is preserved.The env variants were significantly more susceptible to their subtype-matched plasma pool, with a higher mean IC50 for subtype-matched plasma samples than for unmatched plasma samples (138 versus 108, P = 0.0081, paired t test). However, a significant amount of cross-subtype neutralization was observed, as every env variant that was susceptible to the subtype-matched plasma pool was also susceptible to at least one of the other plasma pools (Fig. (Fig.2).2). Thus, although potency was enhanced when the plasma antibodies were produced in response to infection with the same subtype of HIV-1, there were shared neutralization determinants between subtypes, as has been observed previously (reviewed in reference 3).To identify potential correlates of neutralization sensitivity to the antibodies within these plasma pools, we included these 31 env variants and an additional 15 subtype A env variants we previously characterized from the same cohort with the same plasma pools (2). We did not observe a change in neutralization sensitivity during the evolution of the HIV-1 epidemic in Kenya, as no correlation was observed between neutralization sensitivity and the calendar date from which the env variants were isolated. In addition, no correlation was observed between the neutralization sensitivity of a variant to the plasma pools and the duration of estimated infection within that individual. Finally, there was no significant correlation between the neutralization sensitivity and variable-loop length or the number of PNGS. Thus, although changes in the variable-loop length or number of PNGS may alter the exposure of epitopes within the HIV-1 env protein, these changes do not appear to be the primary determinant of neutralization sensitivity.Despite relatively universal sensitivity to at least one of the pooled plasma samples, these transmitted Kenyan env variants were generally resistant to the MAbs 2G12 (provided by Hermann Katinger, Polymun Scientific) and b12 (provided by Dennis Burton, The Scripps Research Institute), as well as 2F5 and 4E10 (obtained from the AIDS Research and Reference Reagent Program, National Institutes of Health) (Fig. (Fig.2),2), though these MAbs neutralized the subtype B env variant SF162, with IC50s similar to those reported previously (1). Subtype D strains were the most susceptible to MAbs, with 4/8 variants neutralized with <20 μg/ml of 2F5 and 2/8 neutralized with <20 μg/ml of the other MAbs. This could reflect the fact that subtype D variants are more closely related to subtype B strains (Fig. (Fig.1)1) (see reference 10), and these MAbs were all derived from subtype B-infected individuals.Among all 31 variants, 2F5 was the most broadly neutralizing, with 15/31 variants from 8/14 subjects neutralized with <20 μg/ml of this MAb. Some 2F5-resistant env variants, such as QH209.14M.ENV.A2 and QB857.110I.ENV.B3, had mutations in the canonical 2F5 binding epitopes, though other 2F5-resistant env variants such as QF495.23M.ENV.A3 and QA790.204I.ENV.A4 maintained the canonical 2F5 epitope. The results with the MAb 4E10 were similar; 4E10 neutralized only seven variants from 4 of the 14 subjects, and the presence of mutations in the 4E10 epitope, which were common, did not predict neutralization sensitivity (Fig. (Fig.2).2). For instance, the env variants QH343.21M.ENV.A10 and QH343.21M.ENV.B5 contained identical N671S and D674S mutations and QH343.21M.ENV.B5 was highly sensitive to 4E10, while QH343.21M.ENV.A10 was resistant (Fig. (Fig.2).2). Thus, for the 2F5 and 4E10 epitopes, the presumed epitopes appear to be shielded in a subset of these early non-subtype B env variants, as has been previously observed (Fig. (Fig.2)2) (1, 2, 5, 14).The MAb b12 neutralized only two variants from two subtype D-infected individuals, with no neutralization of the subtype A, C, and A/D recombinant pseudoviruses. Only four variants from two subjects were neutralized by 2G12 at <20 μg/ml, and these were the only variants that maintained all five of the PNGS within the 2G12 epitope (Fig. (Fig.2).2). Overall, the median IC50 of all the MAbs against these transmitted variants was >20 μg/ml. None of the variants was susceptible to all four MAbs (Fig. (Fig.2),2), unlike many of the early subtype B env variants characterized previously (14).In summary, these newly characterized HIV-1 env clones represent a range of neutralization sensitivities and can be used to supplement existing panels of transmitted variants, in particular, adding the first subtype D and A/D recombinant variants. Some differences between subtypes in env structure following transmission were noted, though these differences did not correlate with neutralization sensitivity. Although the significant levels of cross-subtype neutralization sensitivity observed with plasma samples indicate that some neutralization determinants were shared across subtypes, the epitopes for the MAbs b12, 2G12, 2F5, and 4E10 did not appear to be among the shared determinants. Thus, despite the fact that significant attention has focused on using vaccination to develop antibodies that resemble these MAbs in their specificity, such antibodies may not neutralize the transmitted strains that are causing most new infections worldwide. These data therefore stress the importance of evaluating transmitted variants in endemic areas when designing immunogens and evaluating vaccine and microbicide strategies.  相似文献   

16.
Environmental Isolates of Burkholderia pseudomallei in Ceará State,Northeastern Brazil     
Dione B. Rolim  Marcos F. G. Rocha  Raimunda S. N. Brilhante  Rossana A. Cordeiro  Natanael P. Leit?o-Junior  Timothy J. J. Inglis  José J. C. Sidrim 《Applied and environmental microbiology》2009,75(4):1215-1218
Melioidosis has been considered an emerging disease in Brazil since the first cases were reported to occur in the northeast region. This study investigated two municipalities in Ceará state where melioidosis cases have been confirmed to occur. Burkholderia pseudomallei was isolated in 26 (4.3%) of 600 samples in the dry and rainy seasons.Melioidosis is an endemic disease in Southeast Asia and northern Australia (2, 4) and also occurs sporadically in other parts of the world (3, 7). Human melioidosis was reported to occur in Brazil only in 2003, when a family outbreak afflicted four sisters in the rural part of the municipality of Tejuçuoca, Ceará state (14). After this episode, there was one reported case of melioidosis in 2004 in the rural area of Banabuiú, Ceará (14). And in 2005, a case of melioidosis associated with near drowning after a car accident was confirmed to occur in Aracoiaba, Ceará (11).The goal of this study was to investigate the Tejuçuoca and Banabuiú municipalities, where human cases of melioidosis have been confirmed to occur, and to gain a better understanding of the ecology of Burkholderia pseudomallei in this region.We chose as central points of the study the residences and surrounding areas of the melioidosis patients in the rural areas of Banabuiú (5°18′35″S, 38°55′14″W) and Tejuçuoca (03°59′20″S, 39°34′50′W) (Fig. (Fig.1).1). There are two well-defined seasons in each of these locations: one rainy (running from January to May) and one dry (from June to December). A total of 600 samples were collected at five sites in Tejuçuoca (T1, T2, T3, T4, and T5) and five in Banabuiú (B1, B2, B3, B4, and B5), distributed as follows (Fig. (Fig.2):2): backyards (B1 and T1), places shaded by trees (B2 and T2), water courses (B3 and T3), wet places (B4 and T4), and stock breeding areas (B5 and T5).Open in a separate windowFIG. 1.Municipalities of Banabuiú (5°18′35″S, 38°55′14″W) and Tejuçuoca (03°59′20″S, 39°34′50″W).Open in a separate windowFIG. 2.Soil sampling sites in Banabuiú and Tejuçuoca.Once a month for 12 months (a complete dry/rainy cycle), five samples were gathered at five different depths: at the surface and at 10, 20, 30 and 40 cm (Table (Table1).1). The samples were gathered according to the method used by Inglis et al. (9). Additionally, the sample processing and B. pseudomallei identification were carried out as previously reported (1, 8, 9).

TABLE 1.

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

17.
Influence of Culture Conditions and Medium Composition on the Production of Cellulose by Shiga Toxin-Producing Escherichia coli Cells     
Byong Kwon Yoo  Jinru Chen 《Applied and environmental microbiology》2009,75(13):4630-4632
  相似文献   

18.
Antimicrobial Activity of Simulated Solar Disinfection against Bacterial,Fungal, and Protozoan Pathogens and Its Enhancement by Riboflavin     
Wayne Heaselgrave  Simon Kilvington 《Applied and environmental microbiology》2010,76(17):6010-6012
Riboflavin significantly enhanced the efficacy of simulated solar disinfection (SODIS) at 150 watts per square meter (W m−2) against a variety of microorganisms, including Escherichia coli, Fusarium solani, Candida albicans, and Acanthamoeba polyphaga trophozoites (>3 to 4 log10 after 2 to 6 h; P < 0.001). With A. polyphaga cysts, the kill (3.5 log10 after 6 h) was obtained only in the presence of riboflavin and 250 W m−2 irradiance.Solar disinfection (SODIS) is an established and proven technique for the generation of safer drinking water (11). Water is collected into transparent plastic polyethylene terephthalate (PET) bottles and placed in direct sunlight for 6 to 8 h prior to consumption (14). The application of SODIS has been shown to be a simple and cost-effective method for reducing the incidence of gastrointestinal infection in communities where potable water is not available (2-4). Under laboratory conditions using simulated sunlight, SODIS has been shown to inactivate pathogenic bacteria, fungi, viruses, and protozoa (6, 12, 15). Although SODIS is not fully understood, it is believed to achieve microbial killing through a combination of DNA-damaging effects of ultraviolet (UV) radiation and thermal inactivation from solar heating (21).The combination of UVA radiation and riboflavin (vitamin B2) has recently been reported to have therapeutic application in the treatment of bacterial and fungal ocular pathogens (13, 17) and has also been proposed as a method for decontaminating donor blood products prior to transfusion (1). In the present study, we report that the addition of riboflavin significantly enhances the disinfectant efficacy of simulated SODIS against bacterial, fungal, and protozoan pathogens.Chemicals and media were obtained from Sigma (Dorset, United Kingdom), Oxoid (Basingstoke, United Kingdom), and BD (Oxford, United Kingdom). Pseudomonas aeruginosa (ATCC 9027), Staphylococcus aureus (ATCC 6538), Bacillus subtilis (ATCC 6633), Candida albicans (ATCC 10231), and Fusarium solani (ATCC 36031) were obtained from ATCC (through LGC Standards, United Kingdom). Escherichia coli (JM101) was obtained in house, and the Legionella pneumophila strain used was a recent environmental isolate.B. subtilis spores were produced from culture on a previously published defined sporulation medium (19). L. pneumophila was grown on buffered charcoal-yeast extract agar (5). All other bacteria were cultured on tryptone soy agar, and C. albicans was cultured on Sabouraud dextrose agar as described previously (9). Fusarium solani was cultured on potato dextrose agar, and conidia were prepared as reported previously (7). Acanthamoeba polyphaga (Ros) was isolated from an unpublished keratitis case at Moorfields Eye Hospital, London, United Kingdom, in 1991. Trophozoites were maintained and cysts prepared as described previously (8, 18).Assays were conducted in transparent 12-well tissue culture microtiter plates with UV-transparent lids (Helena Biosciences, United Kingdom). Test organisms (1 × 106/ml) were suspended in 3 ml of one-quarter-strength Ringer''s solution or natural freshwater (as pretreated water from a reservoir in United Kingdom) with or without riboflavin (250 μM). The plates were exposed to simulated sunlight at an optical output irradiance of 150 watts per square meter (W m−2) delivered from an HPR125 W quartz mercury arc lamp (Philips, Guildford, United Kingdom). Optical irradiances were measured using a calibrated broadband optical power meter (Melles Griot, Netherlands). Test plates were maintained at 30°C by partial submersion in a water bath.At timed intervals for bacteria and fungi, the aliquots were plated out by using a WASP spiral plater and colonies subsequently counted by using a ProtoCOL automated colony counter (Don Whitley, West Yorkshire, United Kingdom). Acanthamoeba trophozoite and cyst viabilities were determined as described previously (6). Statistical analysis was performed using a one-way analysis of variance (ANOVA) of data from triplicate experiments via the InStat statistical software package (GraphPad, La Jolla, CA).The efficacies of simulated sunlight at an optical output irradiance of 150 W m−2 alone (SODIS) and in the presence of 250 μM riboflavin (SODIS-R) against the test organisms are shown in Table Table1.1. With the exception of B. subtilis spores and A. polyphaga cysts, SODIS-R resulted in a significant increase in microbial killing compared to SODIS alone (P < 0.001). In most instances, SODIS-R achieved total inactivation by 2 h, compared to 6 h for SODIS alone (Table (Table1).1). For F. solani, C. albicans, ands A. polyphaga trophozoites, only SODIS-R achieved a complete organism kill after 4 to 6 h (P < 0.001). All control experiments in which the experiments were protected from the light source showed no reduction in organism viability over the time course (results not shown).

TABLE 1.

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

19.
Mosaic-Like Sequences Containing Transposon,Phage, and Plasmid Elements among Listeria monocytogenes Plasmids     
Carlos Canchaya  Vanessa Giubellini  Marco Ventura  Clara G. de los Reyes-Gavilán  Abelardo Margolles 《Applied and environmental microbiology》2010,76(14):4851-4857
  相似文献   

20.
Distribution of Shiga-Toxigenic Escherichia coli O157 in the Gastrointestinal Tract of Naturally O157-Shedding Cattle at Necropsy     
James E. Keen  William W. Laegreid  Carol G. Chitko-McKown  Lisa M. Durso  James L. Bono 《Applied and environmental microbiology》2010,76(15):5278-5281
  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

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