Skip to main content
Erschienen in: Gut Pathogens 1/2017

Open Access 01.12.2017 | Research

Cytotoxic Escherichia coli strains encoding colibactin and cytotoxic necrotizing factor (CNF) colonize laboratory macaques

verfasst von: Yan Feng, Anthony Mannion, Carolyn M. Madden, Alton G. Swennes, Catherine Townes, Charles Byrd, Robert P. Marini, James G. Fox

Erschienen in: Gut Pathogens | Ausgabe 1/2017

Abstract

Background

Many Escherichia coli strains are considered to be a component of the normal flora found in the human and animal intestinal tracts. While most E. coli strains are commensal, some strains encode virulence factors that enable the bacteria to cause intestinal and extra-intestinal clinically-relevant infections. Colibactin, encoded by a genomic island (pks island), and cytotoxic necrotizing factor (CNF), encoded by the cnf gene, are genotoxic and can modulate cellular differentiation, apoptosis and proliferation. Some commensal and pathogenic pks+ and cnf+ E. coli strains have been associated with inflammation and cancer in humans and animals.

Results

In the present study, E. coli strains encoding colibactin and CNF were identified in macaque samples. We performed bacterial cultures utilizing rectal swabs and extra-intestinal samples from clinically normal macaques. A total of 239 E. coli strains were isolated from 266 macaques. The strains were identified biochemically and selected isolates were serotyped as O88:H4, O25:H4, O7:H7, OM:H14, and OM:H16. Specific PCR for pks and cnf1 gene amplification, and phylogenetic group identification were performed on all E. coli strains. Among the 239 isolates, 41 (17.2%) were pks+/cnf1−, 19 (7.9%) were pks−/cnf1+, and 31 (13.0%) were pks+/cnf1+. One hundred forty-eight (61.9%) E. coli isolates were negative for both genes (pks−/cnf1−). In total, 72 (30.1%) were positive for pks genes, and 50 (20.9%) were positive for cnf1. No cnf2+ isolates were detected. Both pks+ and cnf1+ E. coli strains belonged mainly to phylogenetic group B2, including B21. Colibactin and CNF cytotoxic activities were observed using a HeLa cell cytotoxicity assay in representative isolates. Whole genome sequencing of 10 representative E. coli strains confirmed the presence of virulence factors and antibiotic resistance genes in rhesus macaque E. coli isolates.

Conclusions

Our findings indicate that colibactin- and CNF-encoding E. coli colonize laboratory macaques and can potentially cause clinical and subclinical diseases that impact macaque models.

Background

Escherichia coli is the predominant aero-anaerobic Gram-negative species of the normal microflora colonizing the gastrointestinal tract of humans and animals [1]. Most E. coli strains are commensal and rarely cause clinically-relevant disease. However, some strains carry virulence genes that enable selected E. coli strains to cause intestinal and extra-intestinal infections [2, 3]. Based on a phylogenetic assay, E. coli can be classified into four main phylogenetic groups (A, B1, B2 and D) [47]. Pathogenic strains which often encode virulence factors belong to group B2 and D, while most fecal E. coli strains belonging to group A and B1 lack virulence factors [8, 9]. A group of cytotoxins, including colibactin, cytotoxic necrotizing factors (CNFs), cytolethal distending toxin (CDT) and cycle inhibiting factor (CIF), are classified as cyclomodulins and are genotoxic and/or modulate cellular differentiation, apoptosis and proliferation [1012].
Colibactin is a cytotoxic hybrid polyketide/nonribosomal peptide produced by several species of Enterobacteriaceae. It was first identified in 2006 in an extra-intestinal pathogenic E. coli (ExPEC) strain isolated from a case of neonatal meningitis [12]. This secondary metabolite, colibactin, is produced by the clbA-S genes present in the 54-kb pathogenicity pks island, a genetic island encoding a non-ribosomal peptide synthetase-polyketide synthase (NRPS-PKS) assembly line [12, 13]. In vitro studies have shown that pks+ E. coli strains induce enlargement of cells and nuclei without mitosis (megalocytosis), cause G2 cell cycle arrest, and DNA double strand breaks [12]. In animal model experiments, a pks+ E. coli strain (NC101), isolated from specific pathogen free wild-type mice induced inflammation in the cecum in interleukin 10 knockout (IL10−/−) mice after a 3 week monoassociation period [14]. Studies also demonstrated that monoassociation with NC101 promotes invasive carcinoma in IL10−/− mice treated with azoxymethane (AOM). The promotion effect was dependent on expression of the pks island [15]. In a previous study from our laboratory, 88% of E. coli isolates from laboratory mice encoded pks genes and belonged to phylogenetic group B2 [16]. Our findings indicated that colibactin-encoding E. coli commonly colonize laboratory mice and may induce clinical and subclinical disease that may impact in vivo experimental results [16].
Escherichia coli strains that produce CNF belong to the pathotype necrotoxigenic E. coli (NTEC) and are associated with intestinal and extra-intestinal infection in both humans and animals [2]. The majority of CNFs include chromosomally encoded cnf1 [17] and plasmid-encoded cnf2 [18]. CNF1 is a 115 kDa protein toxin which activates Rho GTPases, leading to cytoskeletal and cell cycle alterations with subsequent macropinocytosis and formation of megalocytic, multinucleated cells. CNF1-producing and β-hemolytic E. coli strains most notably cause urinary tract and meningeal infections in humans [19]. These strains are also isolated from healthy and diseased animals. In our laboratory, cnf1+ E. coli strains were isolated from ferrets with diarrhea and extra-intestinal infections [20] and from healthy macaques [21]. cnf1-encoding E. coli strains have also been isolated from cats [22], dogs [23, 24], pigs [25], and birds [26].
The prevalence of pks+ E. coli in rhesus macaques is not known, nor is there published evidence that E. coli strains encoding both pks and cnf genes colonize macaques. The purpose of the present study was to characterize rectal E. coli isolates, as well as extra-intestinal isolates, from macaques for the presence of specific virulence genes (pks and cnf) and demonstrate their in vitro toxin activities.

Methods

Animals

Macaques (Macaca mulatta and M. fascicularis), originally received from three US-based vendors, received physical examinations and routine diagnostic evaluations during quarantine and at quarterly intervals in 2012, 2014 and 2016. The colony contained 84 animals in 2012 (4 cynomolgus macaques and 80 rhesus macaques, of which 23 were female), 85 animals in 2014 (4 cynomolgus macaques and 81 rhesus macaques, of which 24 were female) and 97 animals in 2016 (2 cynomolgus macaques and 95 rhesus macaques, of which 25 were female). They ranged in age from 4 to 20 years. Animals were routinely pair-housed and maintained in an animal facility accredited by the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC) International. They were fed specified amounts of primate chow (Purina Lab Diet 5038) twice a day and supplemented daily with treats, seasonal fruits and vegetables. Water was provided ad libitum when animals were not on studies requiring water regulation. Housing conditions were maintained at 20–22 °C, 30–70  % humidity, 10–15 non-recirculated air changes per hour and a light cycle of 12 h light:12 h dark.

Microbiological analysis

Culture and isolation

Two hundred and sixty-nine samples (265 rectal swab, one gingival, two from cephalic recording chambers and one from margin skin) were collected from a cohort of 266 clinically normal macaques (those received in quarantine as well as the established cohort of macaques actively being used in neurobiological research) during quarterly colony health examinations in 2012 (n = 84), 2014 (n = 85) and 2016 (n = 97). Rectal swabs were placed in tubes with sterile Gram-negative broth (BD) and incubated aerobically overnight at 37 °C. The next day, a sterile swab was placed in the Gram-negative culture tube and then streaked onto MacConkey lactose agar plates (BD/BBL, Sparks, MD). For extraintestinal samples, the swab were placed in sterile trypticase soy broth (TSB) (BBL) and incubated aerobically overnight at 37 °C. The next day, a sterile swab was placed in the TSB culture tube and streaked onto Chocolate agar (BBL) and a split plate of blood agar with sheep blood and MacConkey agar (BBL). Lactose-positive colonies were selected and plated onto a sheep blood agar plate (Remel, Lenexa, KS), incubated aerobically, and β-hemolysis, if present, was noted. Suspect E. coli isolates were then characterized by analytic profile index (API) 20 E (Biomérieux, Cambridge, MA). API 20 E is a panel of biochemical tests used for the identification and differentiation of enteric Gram-negative rods. A profile number, determined by the sequence of positive and negative test reactions, is referenced in the API codebook database to determine the bacterial species. Swabs from cranial implants were placed in sterile TSB (BBL), used as an enrichment broth. The swabs were then plated onto chocolate agar (BBL) and a split plate of blood agar with sheep blood and MacConkey agar (BBL). Colonies that were lactose-fermenting positive and Gram-negative on Gram’s stain were subcultured onto sheep blood agar and identification was confirmed with the API 20 E (Biomérieux).

Clinical correlation

Medical records were evaluated for temporal correlations of culture and clinical signs from animals which had pks+ or pks/cnf1 double-positive E. coli isolates. Four records (3 for 2012 and 1 for 2014) were unavailable.

DNA extraction, PCR amplification, and sequencing

Escherichia coli colonies grown on blood agar plates were collected in sterile phosphate buffered saline (PBS) in microfuge tubes, boiled for 10 min, followed by centrifugation at 12,000g for 10 min. The supernatant was used for PCR analysis. The primers and annealing temperatures used are shown in Additional file 1: Table S1. To detect the colibactin genes, three sets of primers were used to amplify clbA, clbB and clbQ genes. Multiplex cnf primers were used to screen for the cnf gene followed by PCR with cnf1 and cnf2 specific primers to further distinguish cnf subgroups. Selected strains were assayed for cdt and cif using primers listed in Additional file 1: Table S1. To determine the phylogenetic groups of isolates, five sets of primers for the genes yjaA, TspE4.C2, chuA, svg and uidA were used in a multiplex PCR [4, 7]. The phylogenetic groups were determined based on the PCR gel pattern. Sequencing of 16S rRNA and clbA and clbQ genes was performed at QuintaraBio (Allston, MA) using primers 9F and F16 (16s rRNA), IHAPJPN42 and IHAPJPN46 (clbA), IHAPJPN55 and IHAPJPN56 (clbQ) for selected isolates.

Serotyping

Eleven E. coli isolates chosen from different cohorts and representing pks+/cnf1−, pks−/cnf1+, pks+/cnf1+, and pks−/cnf1− genotypes were submitted to the E. coli Reference Center at Penn State University for serotype testing, which included: O and H typing and PCR analyses for heat-labile enterotoxin (elt), heat-stabile enterotoxin (estA and estB), Shiga-type toxin 1 and 2 (Stx1 and Stx2), intimin gamma (eae), cnf1, and cnf2.

Cytotoxicity assay

Escherichia coli strains screened in the cytotoxicity assay included K12 (negative control), V27 (a pks and cdt positive control acquired from the E. coli Reference Center), NC101 and NC101∆pks (a pks positive control and pks mutant strain, respectively, both gifts from Dr. Christian Jobin), and 12 selected E. coli isolates representing pks+/cnf1−, pks−/cnf1+, pks+/cnf1+, and pks−/cnf1− genotypes (three for each genotype). Selected tests were also performed on the E. coli strains used in the cytotoxicity assays and included: API 20 E for biochemical characterization and PCR for phylogenetic groups as well as for clbA, clbQ, cnf1, cif and cdt genes.

Cell culture assay for colibactin cytotoxicity

The cytotoxicity assay was performed as described previously with modifications [12, 16]. HeLa S3 cells (ATCC CCL2.2) were grown and maintained in Eagle’s Minimum Essential Medium (EMEM, ATCC) containing 10% Fetal Calf Serum (FCS, Sigma) and 1% antibiotic–antimycotic (Gibco) at 37 °C with 5% CO2. 5 × 103 cells were seeded onto 96-well cell culture plates and incubated at 37 °C with 5% CO2 for 24 h. Overnight liquid cultures of E. coli strains were grown for 2 h at 37 °C and then adjusted to O.D. 600 nm in 1% FCS EMEM media to concentrations corresponding to a multiplicity of infection (MOI; the number of bacteria per cell at the onset of infection) of 1, 5, 25 and 100, respectively. Following inoculation, plates were centrifuged at 200g for 10 min to facilitate bacterial interaction and then incubated at 37 °C with 5% CO2 for 4 h. Cells were then washed with EMEM and replaced with EMEM containing 10% FCS and 200 µg/mL gentamicin (Gibco). Following 72 h incubation, plates were stained with Diff-quick stain (Thermo Scientific). Cells were then inspected under a microscope for confluence and morphological changes. Images were captured with a Zeiss Axiovert-10 microscope using Image Pro-Plus software version 7.0 at 20× magnification.

Cell culture assay for sonicate cytotoxicity

Overnight cultures of E. coli strains were pelleted by centrifugation at 12,000 rpm for 5 min. Supernatant was removed, filtered through a 0.2 μm filter, and stored at − 80 °C for later use. The pellets were washed in 1 mL of PBS and pelleted again by centrifugation at 12,000 rpm for 5 min. Pellets were re-suspended in 2 mL of PBS and then sonicated on ice using the following program: amplitude: 35; power: 7 W; 30 s intervals for a total of 5 min with 1 min breaks between intervals. Sonicate samples were centrifuged at 12,000 rpm for 10 min at 4 °C to remove large debris and filtered through a 0.2 µM filter. Total protein was quantified using the BCA assay (Thermo Fisher Scientific). HeLa cells, 5 × 103 were seeded onto 96-well, cell culture plates and incubated at 37 °C with 5% CO2 for 24 h. Cells were treated with crude bacterial sonicate (40 μg/mL total protein) or supernatant (25 μL) for 72 h. Cells were stained and microscopically analyzed for confluence and morphological changes, as described above.

Draft genome sequencing and comparative analysis

The draft genomes of ten representative rhesus macaque E. coli isolates were sequenced. Genomic DNA was isolated using the MasterPure Complete DNA and RNA Purification Kit (Epicentre, Madison, WI) following the manufacturer’s protocol for bacterial cell samples. DNA libraries were prepared by the Sequencing Core at the Forsyth Institute (Cambridge, MA) using NextraXT for sequencing of 2 × 150 paired-end reads by Illumina MiSeq. Raw sequenced reads were decontaminated of adapter sequences and quality trimmed to a Phred quality score (Q) ≥ 10 using BBDuk from the BBMap package version 37.17 (http://​sourceforge.​net/​projects/​bbmap/​). Decontaminated reads were then assembled into contigs with SPAdes followed by genome annotation with RAST, both services hosted by PATRIC [27]. Sequences encoding putative virulence factor and antibiotic-resistance genes were identified using VirulenceFinder 1.5 [28] and ResFinder 2.1 [29], both using the 90% identity and 60% minimum length threshold parameters. Syntenic relationships of pks genes and the hemolysin-cnf1 operon between genomes were determined with SimpleSynteny [30].

Results

Clinical correlates

Evaluation of medical records did not establish an unequivocal association between E. coli—culture status and clinical signs. There were several animals who had experienced soft feces in the months before rectal culture, but the presence of other agents (e.g. Balantidium coli or Trichuris trichuria) or clinical conditions (e.g. gastric ulcer and Helicobacter suis gastritis) preclude definitive statements. E. coli was isolated from feces in the vast majority of cases, but E. coli isolates were also obtained from gingiva and surgical implants. In one E. coli positive animal, a severe eosinophilic colitis was identified via colonoscopy and histopathological evaluation in an appropriate time frame to suspect E. coli-associated colitis, but this animal also had immune-mediated thrombocytopenia and harbored Balantidium coli [31].

Microbiological characterization of E. coli strains isolated from macaques

A total of 239 E. coli isolates were cultured from 269 rectal swabs and extraintestinal sites obtained from 266 macaques. The yearly prevalence of E. coli in macaques was 60.7% (51 out of 84), 85.7% (72 out of 84) and 75.3% (73 out of 97) in years 2012, 2014, and 2016, respectively (Table 1). In some macaques, there were more than one isolates as determined by different colony morphology characteristics, hemolysis and API code. Forty-three (18.0%) of 239 E. coli isolates were positive for β-hemolysis on sheep blood agar plates (Table 1). The most common API code recorded in this study was 5144572 observed in 44.8% (107 out of 239) of E. coli isolates. Other API codes assigned to the E. coli isolates were 1144572 (11.0%), 5044552 (7.5%), 5144552 (6.9%), 5044542 (6.4%) and 5144512 (4.0%). Fewer than 1% of isolates belonged to other API codes. These API codes confirm the isolates cultured were E. coli.
Table 1
Prevalence of E. coli strains isolated from macaques
 
2012
2014
2016
Total
E. coli positive animals
51/84 (60.7%)
72/85 (85.7%)
73/97 (75.3%)
196/266 (73.7%)
β-hemolytic E. coli isolates
4/66 (6.1%)
16/84 (19.0%)
23/89 (25.8%)
43/239 (18.0%)

Escherichia coli isolates from macaques encoding colibactin and CNF1

To detect whether cytotoxic virulence factors were present in isolated E. coli, PCRs were performed with primers for the clbA, clbB and clbQ genes of the pks island, and the cnf1and cnf2 genes. Among the 239 E. coli isolates, 41 (17.2%) were pks+/cnf1−, 19 (7.9%) were pks−/cnf1+, and 31 (13.0%) were pks+/cnf1+. One hundred forty-eight (61.9%) E. coli isolates were pks−/cnf1− (Table 2, Fig. 1). No cnf2+ isolates were detected.
Table 2
Prevalence of pks and cnf1 genes in E. coli isolates from macaques
 
2012
2014
2016
Total
pks+/cnf1− (A)
11/66 (16.7%)
22/84 (26.2%)
8/89 (9.0%)
41/239 (17.2%)
pks−/cnf1+ (B)
0/66 (0.0%)
5/84 (6.0%)
14/89 (15.7%)
19/239 (7.9%)
pks+/cnf1+ (C)
7/66 (10.6%)
14/84 (16.7%)
10/89 (11.2%)
31/239 (13.0%)
Total pks+ (A + C)
18/66 (27.2%)
36/84 (42.9%)
18/89 (20.2%)
72/239 (30.1%)
Total cnf1+ (B + C)
7/66 (10.6%)
19/84 (22.6%)
24/89 (27.0%)
50/239 (20.9%)
Interestingly, pks+ E. coli strains persistently colonized 10 individual monkeys during the 5-year survey, while persistent colonization by cnf1+ E. coli was observed in seven macaques. There were two E. coli strains isolated from two rhesus; one from a cephalic recording chamber and one from the implant-margin skin site. These two β-hemolytic isolates belonged to the B2 group, were pks+/cnf1+, and were identified as API code 5144572.
Close correlation was observed between the PCR results and microbiological characteristics (β-hemolysis and API code). Almost all (94.0%) β-hemolytic isolates harbored the cnf1 gene. API code 1144572 was related to pks−/cnf1+ isolates, whereas pks+/cnf1− and pks+/cnf1+ isolates were associated with API codes 5144572 and 5144552.

Phylogenetic distribution of E. coli strains isolated from macaques

Based on the PCR amplification pattern of multiplex PCR amplifying five genes (yjaA, TspE4.C2, chuA, svg, and uidA), the E. coli phylogenetic groups were defined as A, B1, B2, B21, and D groups (Fig. 2). The frequency of these groups in E. coli isolates from this study (2014 and 2016) is shown in Table 3 and Fig. 3. The frequency distribution of the phylogenetic groups among the E. coli isolates was comparable between 2014 and 2016. Based on the total from 2014 to 2016 combined, the most common phylogenetic group was the B2 group (37.6%) followed by B1 (26.0%), B21 (22.5%), A (13.3%), and D (0.6%).
Table 3
Phylogenetic distribution of E. coli isolates from macaques
Phylogenetic group
2014
2016
Total
A
5/84 (6.0%)
18/89 (20.2%)
23/173 (13.3%)
B1
23/84 (27.4%)
22/89 (24.7%)
45/173 (26.0%)
B2
37/84 (44.0%)
28/89 (31.5%)
65/173 (37.6%)
B21
18/84 (21.4%)
21/89 (23.6%)
39/173 (22.5%)
D
1/84 (1.2%)
0/89 (0.0%)
1/173 (0.6%)
The distribution of pks, cnf1, and β-hemolysis in E. coli strains according to phylogenetic groups is illustrated in Table 4 and Fig. 4. All β-hemolytic E. coli and all cnf1+ E. coli (except for one isolate) belonged to the B2 phylogenetic group. E. coli strains encoding pks genes belonged predominately to the B2 and B21 groups.
Table 4
Distribution of pks, cnf1 genes and β-hemolysis in E. coli isolates according to phylogenetic group
Phylogenetic group
pks+
cnf1+
β-hemolytic
2014
2016
Total
2014
2016
Total
2014
2016
Total
A
0/36 (0.0%)
1/18 (5.6%)
1/54 (1.9%)
0/19 (0.0%)
0/24 (0.0%)
0/43 (0.0%)
0/16 (0.0%)
0/23 (0.0%)
0/39 (0.0%)
B1
1/36 (2.8%)
1/18 (5.6%)
2/54 (3.7%)
0/19 (0.0%)
1/24 (4.2%)
1/43 (2.3%)
0/16 (0.0%)
0/23 (0.0%)
0/39 (0.0%)
B2
33/36 (91.7%)
11/18 (61.1%)
44/54 (81.5%)
19/19 (100%)
23/24 (95.8%)
42/43 (97.7%)
16/16 (100%)
23/23 (100%)
39/39 (100%)
B21
2/36 (5.6%)
5/18 (27.8%)
7/54 (13.0%)
0/19 (0.0%)
0/24 (0.0%)
0/43 (0.0%)
0/16 (0.0%)
0/23 (0.0%)
0/39 (0.0%)
D
0/36 (0.0%)
0/18 (0.0%)
0/54 (0.0%)
0/19 (0.0%)
0/24 (0.0%)
0/43 (0.0%)
0/16 (0.0%)
0/23 (0.0%)
0/39 (0.0%)

Serotyping

The serotype for two pks+/cnf1+ isolates (S1 and S2) was O88:H4, for five pks−/cnf1+ isolates (S3, S6, S9, S10, and S11) was O25:H4, and for two pks+/cnf1− isolates (S4 and S5) was O7:H7 (Table 5). The two pks−/cnf1− isolates (S7 and S8) had two different serotypes: OM:H14 and OM:H16 (Table 5). All five pks−/cnf1+ isolates (S3, S6, S9, S10, and S11) and both pks+/cnf1+ isolates (S1 and S2) were confirmed positive for the cnf1 gene, and none of the isolates serotyped were positive for elt, estA, estB, stx1, stx2, eae and cnf2 genes (Table 5).
Table 5
Serotype and virulence factors testing results of E. coli isolates from macaques
Sample #
O type
H type
elt
estA
estB
stx1
stx2
eae
cnf1
cnf2
S1 (pks+/cnf1+)
88
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S2 (pks+/cnf1+)
88
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S3 (pks−/cnf1+)
25
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S6 (pks−/cnf1+)
25
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S9 (pks−/cnf1+)
25
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S10 (pks−/cnf1+)
25
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S11 (pks−/cnf1+)
25
4
NEG
NEG
NEG
NEG
NEG
NEG
POS
NEG
S4 (pks+/cnf1−)
7
7
NEG
NEG
NEG
NEG
NEG
NEG
NEG
NEG
S5 (pks+/cnf1−)
7
7
NEG
NEG
NEG
NEG
NEG
NEG
NEG
NEG
S7 (pks−/cnf1−)
M
14
NEG
NEG
NEG
NEG
NEG
NEG
NEG
NEG
S8 (pks−/cnf1−)
M
16
NEG
NEG
NEG
NEG
NEG
NEG
NEG
NEG

In vitro cytotoxicity of E. coli isolates from macaques

Previous studies have found that the cyclomodulins colibactin, CNF, CDT, and CIF cause megalocytic-like cytotoxicity to cell lines in vitro; however this effect by colibactin and CIF is dependent on contact with live bacteria, while for CNF and CDT is observable only with bacterial sonicate or supernatant treatment [11]. Therefore, we tested representative E. coli isolates using cell culture assays to confirm colibactin and CNF1 cytotoxic activity predicted by the PCR results.
Transient infection with pks+/cnf1E. coli isolates (S4, S5, and S13) at MOI 100 caused contact-dependent megalocytosis (Fig. 5a); however, sonicate treatment with these isolates did not cause observable cytotoxicity (Fig. 5b). Infection with pks+/cnf1+ E. coli isolates (S1, S2, and S14) killed all HeLa cells at MOI ≥ 5. Surviving HeLa cells at MOI 1 appeared megalocytic (Fig. 5a). Sonicate treatment by these isolates also caused HeLa cell body distention and multi-nucleation (Fig. 5b). E. coli isolates that were pks−/cnf1+ (S3, S9, and S10) did not cause megalocytosis to HeLa cells after infection, but instead caused a cytotoxic effect observed as cellular elongation (Fig. 5a). Sonicate treatment with these isolates caused HeLa cells to become distended with multiple nuclei (Fig. 5b). HeLa cells treated with live bacteria or sonicate from pks−/cnf1− (S7, S8, and S12) appeared indistinguishable from those treated with media, K12, and NC101 Δpks negative controls (Fig. 5). Megalocytic CNF1 cytotoxicity was also observed only when HeLa cells were treated with culture supernatant from the cnf1+ isolates (S3 and S14) (Additional file 2: Figure S1). All isolates were found by PCR to be negative for cdt and cif genes, therefore excluding the influence of these cyclomodulins. Together, these results demonstrate that the novel E. coli isolates exhibit colibactin and CNF1 cytotoxicity, as predicted by their genotype.

Draft genome sequencing and comparative analysis

The draft genomes of ten representative rhesus macaque E. coli isolates were sequenced for identification of virulence factor and antimicrobial resistance genes, as well as for comparative analysis. Genome sizes, GC content, and the number of annotated protein coding and RNA genes were comparable between the different rhesus macaque E. coli isolates and representative pathogenic and non-pathogenic E. coli strains (Table 6).
Table 6
Genome characteristics
Strain/isolate
Source
Genome length (bp)
Contigs
N50
GC content (%)
Protein genes (CDS)
tRNA genes
rRNA genes
Virulence factor genes
Antibiotic resistance genes
GenBank accession
S1 (pks+/cnf1+)
Research rhesus macaque rectal swab
5,070,329
92
268,704
50.49
5022
78
10
astA, celb, cnf1, gad, hlyCABD, iroN, iss, mchB, mchC, mchF, mcmA, pks, vat
NHZD00000000
S2 (pks+/cnf1+)
Research rhesus macaque rectal swab
5,070,134
98
268,677
50.49
5017
78
10
astA, celb, cnf1, gad, hlyCABD, iroN, iss, mchB, mchC, mchF, mcmA, pks, vat
NHZC00000000
S4 (pks+/cnf1−)
Research rhesus macaque rectal swab
4,843,133
160
65,852
50.72
4721
83
10
celb, gad, iroN, iss, mchB, mchC, mchF, mcmA, pic, pks, vat
NHZA00000000
S5 (pks+/cnf1−)
Research rhesus macaque rectal swab
4,899,189
116
127,782
50.54
4791
81
10
celb, gad, iroN, iss, mchB, mchC, mchF, mcmA, pic, pks, vat
NHYZ00000000
S3 (pks−/cnf1+)
Research rhesus macaque rectal swab
5,239,168
227
105,395
50.66
5403
80
8
cnf1, hlyCABD, gad, iha, iss, sat
aac(6)Ib-cr, blaCTX-M-15, blaOXA-1, catB3-like, tet(A)
NHZB00000000
S6 (pks−/cnf1+)
Research rhesus macaque rectal swab
5,345,115
176
178,747
50.53
5502
79
9
cnf1, hlyCABD, gad, iha, iss, sat
aac(6)Ib-cr, blaCTX-M-15, blaOXA-1, catB3-like, tet(A)
NHYY00000000
S9 (pks−/cnf1+)
Research rhesus macaque rectal swab
5,536,267
173
170,757
50.32
5765
81
9
cnf1, hlyCABD, gad, iha, iss, sat
aac(6)Ib-cr, blaCTX-M-15, blaOXA-1, catB3-like, tet(A)
NHYV00000000
S10 (pks−/cnf1+)
Research rhesus macaque rectal swab
5,254,454
178
113,006
50.70
5393
80
9
cnf1, hlyCABD, gad, iha, iss, sat
aac(6)Ib-cr, blaCTX-M-15, blaOXA-1, catB3-like, tet(A)
NHYU00000000
S7 (pks−/cnf1−)
Research rhesus macaque rectal swab
4,840,594
136
147,699
50.76
4807
84
10
gad, iss, lpfA
NHYX00000000
S8 (pks−/cnf1−)
Research rhesus macaque rectal swab
4,770,474
119
168,371
50.68
4730
81
11
gad, iss, lpfA
NHYW00000000
IHE3034
Human neonatal meningitis
5,108,383
1 (complete genome)
50.70
5045
97
22
gad, iroN, iss, pks, sfaS, vat
CP001969.1
NC101
Research mouse intestinal commensal, pro-carcinogenic
5,021,144
27
511,891
50.57
4917
72
4
gad, iroN, iss, pks, sfaS, vat
AEFA00000000.1
UTI89
Human uropathogenic strain
5,179,971
1 (complete genome)
50.60
5040
89
14
cnf1, gad, gad, iroN, iss, senB, sfaS, vat
CP000243.1
K-12 substr. DH10B
Human non-pathogenic intestinal commensal
4,686,137
1 (complete genome)
50.80
4606
87
14
gad, iss
AP012306.1
Escherichia coli isolates that were PCR-positive for pks and/or cnf1 genes also had the full-length gene sequences detected in their annotated genomes. Additionally, sequences for the cyclomodulins cnf2, cdt, and cif, as well as the virulence factor genes elt, estA, estB, stx1, stx2, and eae were not detected in any of the genomes, which agrees with previous PCR and serotyping results.
Complete pks gene islands were identified in isolates S1, S2, S4, and S5. All pks island genes showed ≥ 98% sequence homology and identical syntenic relationships compared to prototype pks+ strains IHE3034 and NC101 (Fig. 6a). Likewise, all isolates positive for cnf1 had the β-hemolysin genes hlyCABD immediately upstream, consistent with the hemolysin-cnf1 operon found in uropathogenic E. coli (UPEC) such as UTI89 (Fig. 6b).
Other putative virulence factor genes were also detected in the E. coli isolates (Table 6). These virulence factor genes represent toxins (astA, pic, vat, sat), survival and immune evasion factors (gad, iss), iron acquisition (iroN), adherence (iha, lpfA) and bacteriocin synthesis (celb, mchB, mchC, mchF, mcmA). These virulence genes are associated with host colonization and pathogenicity in intestinal and extra-intestinal diseases (Additional file 1: Table S2).

Discussion

Escherichia coli is a normal inhabitant of the gastrointestinal tract of macaques and several different serotypes of bacteria have been isolated from asymptomatic rhesus macaques [32]. To our knowledge this is the first reported isolation of colibactin-encoding E. coli strains from macaques. Colibactin was first identified in several E. coli strains by Oswald and co-workers in 2006 [12]. Colibactin, encoded by a 54 kb gene cluster (the pks island), is a genotoxin which causes DNA double strand breaks and activation of the DNA damage checkpoint pathway, leading to cell cycle arrest and eventually cell death. The role of colibactin-encoding E. coli has been explored in human colorectal cancer and investigated in different types of mouse models including IL10−/− mice treated with azoxymethane (AOM), C57BL/6J-ApcMin/J mice treated with AOM and dextran sodium sulfate (DSS), and nude mice with xenografts [11, 15, 33, 34]. These studies have established a role of pks-encoding E. coli in inflammation and cancer. The increase in E. coli growth and colonization may be due to inflammation-generated nitrate, as E. coli, a facultative anaerobe, can produce energy through the use of nitrate, S-oxides and N-oxides as terminal electron acceptors for anaerobic respiration and thus outcompete obligate anaerobes, the major colonizers of the lower bowel [35].
Among the E. coli isolates from macaques, 30.1% of isolates were pks+ strains and colibactin activities were confirmed in selected isolates. The prevalence of pks+ E. coli colonization in macaques was similar to the 25% prevalence of pks+ B2 E. coli colonization in humans [36]. Several publications have revealed the higher prevalence of pks+ E. coli strains in biopsies from colorectal cancer (CRC) patients (66.7% in CRC patients, 40% in IBD patients and 20.8% in no IBD/no CRC controls) [11, 15, 37].
The 20.9% prevalence of cnf1+ E. coli in this study was consistent with our previous findings [21]. Close correlation was observed between the cnf1 gene and β-hemolysis in E. coli isolates. This association was reported in our previous studies [20, 21] and by other authors [19, 38]. In the E. coli J96 strain and other strains, the cnf1 gene is located downstream of hemolysin (hlyCABD) in pathogenicity island II (PAI II), and the expression of cnf1 is regulated by the hemolysin promoter [3941]. Likewise, our draft genome sequences of macaque E. coli strains indicate that hemolysin (hlyCABD) is located directly upstream of the cnf1 gene. These two toxins could therefore be associated with enhanced virulence. In our in vitro cytotoxicity assays, infection with the pks−/cnf1+ isolates (S3, S9, S10) caused ~ 50% of HeLa cells to die, and the surviving cells exhibited an elongated morphology; CNF1 cytotoxicity by these isolates was only observed with sonicate and supernatant treatment. The draft genome sequence data showed that only pks−/cnf1+ isolates (S3, S6, S9, and S10) also contained an annotated secreted autotransporter toxin (sat), which is expressed by some UPEC strains and reported to cause cell elongation in vitro [42]. We speculate that expression of sat by these isolates may have caused HeLa cells to adopt an elongated morphology. Interestingly, we also identified E. coli strains co-harboring the pks and cnf1 genes. Thirty-one (13.0%) E. coli isolates were positive for both pks and cnf1 (pks+/cnf1+). The E. coli strains co-harboring these two toxins have also been reported in human samples [10, 11]. The authors noted that 15% of E. coli isolates from urosepsis patients and from the feces of healthy individuals were double-positive for pks and cnf1 (pks+/cnf1+). In our in vitro cytotoxicity assays, the isolates pks+/cnf1+ (S1, S2, and S14) exhibited severe toxicity to HeLa cells, given all cells were dead when treated with these live isolates at MOI 5, 25 and 100 (Fig. 5).
Yasuda and co-workers characterized biogeographic relationships in the rhesus macaque intestinal microbiome and found that stool microbiota was highly representative of the colonic lumen and mucosa, which were respectively enriched in obligate and facultative anaerobes [43]. In our study the E. coli strains were isolated from rectal swab samples and may, by analogy, be representative of the E. coli strains colonized in the macaque gastrointestinal tract. Moreover, among E. coli isolates in the present study, two isolates which were β-hemolytic and pks+/cnf1+ were isolated from a cephalic recording chamber and implant-margin skin of two macaques. This raises the concern that these E. coli isolates may cause meningitis in macaques used in neurobiological research based on the previous reports that pks+ E. coli were isolated and related to meningitis in humans and animals [44].
For the E. coli isolates from macaques in this study, the predominant phylogenetic group was B2 group including B21 subgroup (60.1%) followed by B1 (26.0%), A (13.3%), and D group (0.6%). The distribution of phylogenetic groups of macaque E. coli strains were similar to distribution of E. coli isolates from healthy humans [10, 11]. The distribution of cyclomodulin-encoding genes (pks, cnf, cdt and cif) in relation to the phylogenetic background in E. coli isolates from urosepsis patients and healthy individuals indicated that strains pks+ and/or cnf 1+ strongly associated with the B2 group [10]. In another study, the prevalence of E. coli producing cyclomodulins and genotoxins in colon cancer had a higher prevalence of the B2 phylogenetic group E. coli harboring the pks gene (55.0%) and cnf1 gene (39.5%) in biopsies of patients with colorectal cancer than that in patients with diverticulosis (19.3% pks+ and 12.9% cnf1+) [11]. In both studies, the percentage of E. coli strains harboring cdt and cif genes were much lower (1–6%). Representative isolates in our study were negative for cdt and cif according to PCR, in vitro cytotoxicity assay, and genome analysis results. We found that pks+, cnf1+, and β-hemolytic E. coli strains belonged to group B2. 49.0% (51 out of 104) of the isolates belonging to B2 (including the B21 subgroup) were pks+, 40.4% (42 out of 104) were cnf1+, and 37.5% (39 out of 104) were β-hemolytic. Seven pks+ E. coli isolates belonged to the B21 subgroup, which is a highly virulent phylogenetic subgroup among extra-intestinal pathogenic E. coli B2 strains [7, 44].
The serotype data in the present study revealed that the serotype of selected E. coli strains corresponded to their toxin-harboring content. Of the isolates serotyped, those pks+/cnf1+ (S1 and S2) were the O88:H4 serotype. The pks−/cnf1+ isolates (S3, S6, S9, S10, S11) were O25:H4, the pks+/cnf1− isolates (S4 and S5) were O7:H7, the pks−/cnf1− isolates (S7 and S8) were OM:H14 or OM:H16. The O7:H7:K1 serotype belonging to phylogenetic group B2 was cultured from IL10−/− and wild-type mice [45]. In these experiments, cecal and colonic inflammation observed in IL10−/− mice was accompanied by diminished intestinal microbial diversity and a higher number of E. coli organisms compared to wild-type mice [45]. Serotype O7:H7 E. coli strains were also found among Shiga-toxin-producing E. coli strain isolated from calves in Brazil [46]. UPEC O25:H4 strains were reported in patients with urinary tract infection [47] and patients with cystitis or prostatitis [48]. These strains belonging to group B2 had multiple antibiotic drug resistance. By analyzing the draft genome sequences of our macaque E. coli isolates, putative multi-antibiotic resistance genes were identified exclusively in cnf1+ strains (serotype O25:H4). These included resistance genes to the tetracycline (tet(A)), phenicol (catB3-like), fluoroquinolone and aminoglycoside (aac(6′6′)Ib-cr), and β-lactam (blaCTX-M-15, blaOXA-1) classes of antibiotics, consistent with our previous antibiotic resistance findings of cnf1+ E. coli strains in macaques [21]. Other studies have noted that antibiotic-susceptibility is inversely related to the number of virulence factor genes present in extra-intestinal E. coli strains [49]. Similarly, in this study isolates that were pks−/cnf1+ had the second fewest number of virulence factor genes and also were the only isolates with putative antibiotic resistance genes detected.
Different virulence factor gene profiles appear to be present depending on whether the isolates were pks+/cnf1+, pks+/cnf1−, pks−/cnf1+, or pks−/cnf1−. The number and prolife of these virulence genes agrees with in vitro cytotoxicity to HeLa cells in that the isolates showing pronounced cytotoxicity (pks+ and/or cnf1+ strains) had substantially more virulence factor genes present in their genomes compared to the less cytotoxic pks−/cnf1− isolates. Except for lpfA, the pks−/cnf1E. coli isolates had the same virulence factor genes as K12, suggesting that along with the in vitro cytotoxicity results, these isolates likely have attenuated pathogenic potential. Interestingly, pks+ strains harboring the bacteriocin synthesis genes for colicin E2 and microcin H47 also had the most virulence genes. This agrees with other studies reporting that E. coli strains expressing bacteriocins are statistically more likely to co-associate with more virulence factor genes in their genomes compared to strains that lack bacteriocin potential [4951]. Likewise, bacteriocin genes are found more frequently in E. coli strains belonging to the pathogenic B2 or D phylogroup, such as our monkey isolates [49, 50, 52]. In particular, microcin H47 is predominantly found in the UPEC strains [53]. It is hypothesized that bacteriocin activity by pathogenic E. coli may provide a competitive survival and colonization advantage against commensal organisms, especially when availability of essential nutrients, like iron, is scarce [51].
In summary, E. coli strains encoding colibactin, CNF1, or both were identified in macaques. The pks+ and/or cnf1+ isolates belonged to phylogenetic group B2 and induced cytotoxic effects to HeLa cells in vitro. The genomic data supports the presence of virulence factor and antibiotic resistance genes in these isolates and suggests that they may have the pathogenic potential to influence clinical and subclinical disease. The impact of these strains on the health of macaques is unclear as analysis of medical records did not allow an association of clinical events and isolation of E. coli. Given colibactin and CNF1-encoding E. coli has been isolated from human and animals populations, there is a concern about potential zoonotic spread. The presence of colibactin and CNF1-producing E. coli strains in primates used in neurobiology emphasizes the importance of appropriate personnel protection and hygiene practices when handling these primates.

Conclusions

The prevalence of pks+ E. coli in rhesus macaques is not known, nor is there published evidence that E. coli strains encoding both pks and cnf1 genes colonize macaques. In the present study, E. coli strains encoding colibactin and CNF1 were identified in the rectal swabs and extra-intestinal samples of macaques sampled over a 3-year period. Among the 239 isolates, 72 (30.1%) were positive for pks genes and 50 (20.9%) were cnf1+. Our findings indicate that colibactin and CNF1-encoding E. coli colonizing laboratory macaques can potentially cause clinical and subclinical diseases that impact studies conducted in macaque models.

Authors’ contributions

YF performed PCR, cell cultures, assisted in data analysis, and was a major contributor in writing the manuscript. AM performed genome sequencing and comparative analysis, cell culture assays, assisted in data analysis, and was a major contributor in writing the manuscript. CMM performed bacterial culture, isolation, and identification, as well as data management. AGS collected samples, assisted in bacterial culture, DNA extraction, and PCR, as well as data interpretation. CT assisted with statistical analyses by evaluating data from an NHP survey of pks+ E. coli samples collected from the colonies at MIT, assisted in bacterial culture, DNA extraction, and PCR. CB assisted in bacterial culture, DNA extraction, and PCR. RPM analyzed medical records for survey, assisted in project oversight, and contributed to writing the manuscript. JGF was responsible for project conception and design, provided project oversight, data interpretation and analysis, and was a major contributor in writing the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We would like to thank Alyssa Pappa for her excellent assistance preparing the manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.

Data availability

Please contact corresponding author (James G. Fox) for data requests.
Work involving animals was approved by the Massachusetts Institute of Technology’s Committee on Animal Care and Use Office.

Funding

Research reported in this publication was supported by The National Institutes of Health under Award Numbers T32OD010978, R01CA093405, P01CA028842-23 and National Institute of Environmental Health Sciences under Award P30ES0022109. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Leser TD, Molbak L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ Microbiol. 2009;11(9):2194–206.CrossRefPubMed Leser TD, Molbak L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ Microbiol. 2009;11(9):2194–206.CrossRefPubMed
2.
Zurück zum Zitat Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol. 2004;2(2):123–40.CrossRefPubMed Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol. 2004;2(2):123–40.CrossRefPubMed
3.
Zurück zum Zitat Croxen MA, Finlay BB. Molecular mechanisms of Escherichia coli pathogenicity. Nat Rev Microbiol. 2010;8(1):26–38.PubMed Croxen MA, Finlay BB. Molecular mechanisms of Escherichia coli pathogenicity. Nat Rev Microbiol. 2010;8(1):26–38.PubMed
4.
Zurück zum Zitat Clermont O, Bonacorsi S, Bingen E. Rapid and simple determination of the Escherichia coli phylogenetic group. Appl Environ Microbiol. 2000;66(10):4555–8.CrossRefPubMedPubMedCentral Clermont O, Bonacorsi S, Bingen E. Rapid and simple determination of the Escherichia coli phylogenetic group. Appl Environ Microbiol. 2000;66(10):4555–8.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Herzer PJ, et al. Phylogenetic distribution of branched RNA-linked multicopy single-stranded DNA among natural isolates of Escherichia coli. J Bacteriol. 1990;172(11):6175–81.CrossRefPubMedPubMedCentral Herzer PJ, et al. Phylogenetic distribution of branched RNA-linked multicopy single-stranded DNA among natural isolates of Escherichia coli. J Bacteriol. 1990;172(11):6175–81.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Carlos C, et al. Escherichia coli phylogenetic group determination and its application in the identification of the major animal source of fecal contamination. BMC Microbiol. 2010;10:161.CrossRefPubMedPubMedCentral Carlos C, et al. Escherichia coli phylogenetic group determination and its application in the identification of the major animal source of fecal contamination. BMC Microbiol. 2010;10:161.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Bidet P, et al. Detection and identification by PCR of a highly virulent phylogenetic subgroup among extraintestinal pathogenic Escherichia coli B2 strains. Appl Environ Microbiol. 2007;73(7):2373–7.CrossRefPubMedPubMedCentral Bidet P, et al. Detection and identification by PCR of a highly virulent phylogenetic subgroup among extraintestinal pathogenic Escherichia coli B2 strains. Appl Environ Microbiol. 2007;73(7):2373–7.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Picard B, et al. The link between phylogeny and virulence in Escherichia coli extraintestinal infection. Infect Immun. 1999;67(2):546–53.PubMedPubMedCentral Picard B, et al. The link between phylogeny and virulence in Escherichia coli extraintestinal infection. Infect Immun. 1999;67(2):546–53.PubMedPubMedCentral
9.
Zurück zum Zitat Escobar-Paramo P, et al. Identification of forces shaping the commensal Escherichia coli genetic structure by comparing animal and human isolates. Environ Microbiol. 2006;8(11):1975–84.CrossRefPubMed Escobar-Paramo P, et al. Identification of forces shaping the commensal Escherichia coli genetic structure by comparing animal and human isolates. Environ Microbiol. 2006;8(11):1975–84.CrossRefPubMed
11.
12.
Zurück zum Zitat Nougayrede JP, et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 2006;313(5788):848–51.CrossRefPubMed Nougayrede JP, et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 2006;313(5788):848–51.CrossRefPubMed
13.
Zurück zum Zitat Putze J, et al. Genetic structure and distribution of the colibactin genomic island among members of the family Enterobacteriaceae. Infect Immun. 2009;77(11):4696–703.CrossRefPubMedPubMedCentral Putze J, et al. Genetic structure and distribution of the colibactin genomic island among members of the family Enterobacteriaceae. Infect Immun. 2009;77(11):4696–703.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Kim SC, et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology. 2005;128(4):891–906.CrossRefPubMed Kim SC, et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology. 2005;128(4):891–906.CrossRefPubMed
16.
17.
Zurück zum Zitat Falbo V, et al. Isolation and nucleotide sequence of the gene encoding cytotoxic necrotizing factor 1 of Escherichia coli. Infect Immun. 1993;61(11):4909–14.PubMedPubMedCentral Falbo V, et al. Isolation and nucleotide sequence of the gene encoding cytotoxic necrotizing factor 1 of Escherichia coli. Infect Immun. 1993;61(11):4909–14.PubMedPubMedCentral
18.
Zurück zum Zitat Oswald E, et al. Cytotoxic effect of multinucleation in HeLa cell cultures associated with the presence of Vir plasmid in Escherichia coli strains. FEMS Microbiol Lett. 1989;49(1):95–9.CrossRefPubMed Oswald E, et al. Cytotoxic effect of multinucleation in HeLa cell cultures associated with the presence of Vir plasmid in Escherichia coli strains. FEMS Microbiol Lett. 1989;49(1):95–9.CrossRefPubMed
19.
Zurück zum Zitat Landraud L, et al. Frequency of Escherichia coli strains producing the cytotoxic necrotizing factor (CNF1) in nosocomial urinary tract infections. Lett Appl Microbiol. 2000;30(3):213–6.CrossRefPubMed Landraud L, et al. Frequency of Escherichia coli strains producing the cytotoxic necrotizing factor (CNF1) in nosocomial urinary tract infections. Lett Appl Microbiol. 2000;30(3):213–6.CrossRefPubMed
20.
Zurück zum Zitat Marini RP, et al. Characterization of hemolytic Escherichia coli strains in ferrets: recognition of candidate virulence factor CNF1. J Clin Microbiol. 2004;42(12):5904–8.CrossRefPubMedPubMedCentral Marini RP, et al. Characterization of hemolytic Escherichia coli strains in ferrets: recognition of candidate virulence factor CNF1. J Clin Microbiol. 2004;42(12):5904–8.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Martin HR, et al. Characterization of cytotoxic necrotizing factor 1-producing Escherichia coli strains from faeces of healthy macaques. J Med Microbiol. 2009;58(Pt 10):1354–8.CrossRefPubMedPubMedCentral Martin HR, et al. Characterization of cytotoxic necrotizing factor 1-producing Escherichia coli strains from faeces of healthy macaques. J Med Microbiol. 2009;58(Pt 10):1354–8.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Feria C, et al. Virulence genes and P fimbriae PapA subunit diversity in canine and feline uropathogenic Escherichia coli. Vet Microbiol. 2001;82(1):81–9.CrossRefPubMed Feria C, et al. Virulence genes and P fimbriae PapA subunit diversity in canine and feline uropathogenic Escherichia coli. Vet Microbiol. 2001;82(1):81–9.CrossRefPubMed
23.
Zurück zum Zitat Johnson JR, et al. Identification of urovirulence traits in Escherichia coli by comparison of urinary and rectal E. coli isolates from dogs with urinary tract infection. J Clin Microbiol. 2003;41(1):337–45.CrossRefPubMedPubMedCentral Johnson JR, et al. Identification of urovirulence traits in Escherichia coli by comparison of urinary and rectal E. coli isolates from dogs with urinary tract infection. J Clin Microbiol. 2003;41(1):337–45.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Siqueira AK, et al. Virulence factors in Escherichia coli strains isolated from urinary tract infection and pyometra cases and from feces of healthy dogs. Res Vet Sci. 2009;86(2):206–10.CrossRefPubMed Siqueira AK, et al. Virulence factors in Escherichia coli strains isolated from urinary tract infection and pyometra cases and from feces of healthy dogs. Res Vet Sci. 2009;86(2):206–10.CrossRefPubMed
25.
Zurück zum Zitat Toth I, et al. Characterization of intestinal cnf1+ Escherichia coli from weaned pigs. Int J Med Microbiol. 2000;290(6):539–42.CrossRefPubMed Toth I, et al. Characterization of intestinal cnf1+ Escherichia coli from weaned pigs. Int J Med Microbiol. 2000;290(6):539–42.CrossRefPubMed
26.
Zurück zum Zitat Rodriguez-Siek KE, et al. Comparison of Escherichia coli isolates implicated in human urinary tract infection and avian colibacillosis. Microbiology. 2005;151(Pt 6):2097–110.CrossRefPubMed Rodriguez-Siek KE, et al. Comparison of Escherichia coli isolates implicated in human urinary tract infection and avian colibacillosis. Microbiology. 2005;151(Pt 6):2097–110.CrossRefPubMed
27.
Zurück zum Zitat Wattam AR, et al. Improvements to PATRIC, the all-bacterial bioinformatics database and analysis resource center. Nucleic Acids Res. 2017;45(D1):D535–42.CrossRefPubMed Wattam AR, et al. Improvements to PATRIC, the all-bacterial bioinformatics database and analysis resource center. Nucleic Acids Res. 2017;45(D1):D535–42.CrossRefPubMed
28.
Zurück zum Zitat Joensen KG, et al. Real-time whole-genome sequencing for routine typing, surveillance, and outbreak detection of verotoxigenic Escherichia coli. J Clin Microbiol. 2014;52(5):1501–10.CrossRefPubMedPubMedCentral Joensen KG, et al. Real-time whole-genome sequencing for routine typing, surveillance, and outbreak detection of verotoxigenic Escherichia coli. J Clin Microbiol. 2014;52(5):1501–10.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Veltri D, Wight MM, Crouch JA. SimpleSynteny: a web-based tool for visualization of microsynteny across multiple species. Nucleic Acids Res. 2016;44(W1):W41–5.CrossRefPubMedPubMedCentral Veltri D, Wight MM, Crouch JA. SimpleSynteny: a web-based tool for visualization of microsynteny across multiple species. Nucleic Acids Res. 2016;44(W1):W41–5.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Frydman GH, et al. Local and systemic changes associated with long-term, percutaneous, static implantation of titanium alloys in rhesus macaques (Macaca mulatta). Comp Med. 2017;67(2):165–75.PubMedPubMedCentral Frydman GH, et al. Local and systemic changes associated with long-term, percutaneous, static implantation of titanium alloys in rhesus macaques (Macaca mulatta). Comp Med. 2017;67(2):165–75.PubMedPubMedCentral
32.
Zurück zum Zitat Schiff LJ, et al. Enteropathogenic Escherichia coli infections: increasing awareness of a problem in laboratory animals. Lab Anim Sci. 1972;22(5):705–8.PubMed Schiff LJ, et al. Enteropathogenic Escherichia coli infections: increasing awareness of a problem in laboratory animals. Lab Anim Sci. 1972;22(5):705–8.PubMed
33.
Zurück zum Zitat Cougnoux A, et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut. 2014;63(12):1932–42.CrossRefPubMed Cougnoux A, et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut. 2014;63(12):1932–42.CrossRefPubMed
34.
Zurück zum Zitat Bonnet M, et al. Colonization of the human gut by E. coli and colorectal cancer risk. Clin Cancer Res. 2014;20(4):859–67.CrossRefPubMed Bonnet M, et al. Colonization of the human gut by E. coli and colorectal cancer risk. Clin Cancer Res. 2014;20(4):859–67.CrossRefPubMed
36.
Zurück zum Zitat Secher T, Brehin C, Oswald E. Early settlers: which E. coli strains do you not want at birth? Am J Physiol Gastrointest Liver Physiol. 2016;311(1):G123–9.CrossRefPubMed Secher T, Brehin C, Oswald E. Early settlers: which E. coli strains do you not want at birth? Am J Physiol Gastrointest Liver Physiol. 2016;311(1):G123–9.CrossRefPubMed
37.
Zurück zum Zitat Raisch J, et al. Colon cancer-associated B2 Escherichia coli colonize gut mucosa and promote cell proliferation. World J Gastroenterol. 2014;20(21):6560–72.CrossRefPubMedPubMedCentral Raisch J, et al. Colon cancer-associated B2 Escherichia coli colonize gut mucosa and promote cell proliferation. World J Gastroenterol. 2014;20(21):6560–72.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Blanco M, et al. Polymerase chain reaction for detection of Escherichia coli strains producing cytotoxic necrotizing factor type 1 and type 2 (CNF1 and CNF2). J Microbiol Methods. 1996;26(1):95–101.CrossRef Blanco M, et al. Polymerase chain reaction for detection of Escherichia coli strains producing cytotoxic necrotizing factor type 1 and type 2 (CNF1 and CNF2). J Microbiol Methods. 1996;26(1):95–101.CrossRef
39.
Zurück zum Zitat Blum G, et al. Gene clusters encoding the cytotoxic necrotizing factor type 1, Prs-fimbriae and alpha-hemolysin form the pathogenicity island II of the uropathogenic Escherichia coli strain J96. FEMS Microbiol Lett. 1995;126(2):189–95.PubMed Blum G, et al. Gene clusters encoding the cytotoxic necrotizing factor type 1, Prs-fimbriae and alpha-hemolysin form the pathogenicity island II of the uropathogenic Escherichia coli strain J96. FEMS Microbiol Lett. 1995;126(2):189–95.PubMed
40.
Zurück zum Zitat Landraud L, et al. Expression of cnf1 by Escherichia coli J96 involves a large upstream DNA region including the hlyCABD operon, and is regulated by the RfaH protein. Mol Microbiol. 2003;47(6):1653–67.CrossRefPubMed Landraud L, et al. Expression of cnf1 by Escherichia coli J96 involves a large upstream DNA region including the hlyCABD operon, and is regulated by the RfaH protein. Mol Microbiol. 2003;47(6):1653–67.CrossRefPubMed
41.
Zurück zum Zitat Lemonnier M, Landraud L, Lemichez E. Rho GTPase-activating bacterial toxins: from bacterial virulence regulation to eukaryotic cell biology. FEMS Microbiol Rev. 2007;31(5):515–34.CrossRefPubMed Lemonnier M, Landraud L, Lemichez E. Rho GTPase-activating bacterial toxins: from bacterial virulence regulation to eukaryotic cell biology. FEMS Microbiol Rev. 2007;31(5):515–34.CrossRefPubMed
42.
Zurück zum Zitat Guyer DM, et al. Identification of sat, an autotransporter toxin produced by uropathogenic Escherichia coli. Mol Microbiol. 2000;38(1):53–66.CrossRefPubMed Guyer DM, et al. Identification of sat, an autotransporter toxin produced by uropathogenic Escherichia coli. Mol Microbiol. 2000;38(1):53–66.CrossRefPubMed
43.
44.
Zurück zum Zitat Bonacorsi S, et al. Molecular analysis and experimental virulence of French and North American Escherichia coli neonatal meningitis isolates: identification of a new virulent clone. J Infect Dis. 2003;187(12):1895–906.CrossRefPubMed Bonacorsi S, et al. Molecular analysis and experimental virulence of French and North American Escherichia coli neonatal meningitis isolates: identification of a new virulent clone. J Infect Dis. 2003;187(12):1895–906.CrossRefPubMed
45.
Zurück zum Zitat Wohlgemuth S, et al. Reduced microbial diversity and high numbers of one single Escherichia coli strain in the intestine of colitic mice. Environ Microbiol. 2009;11(6):1562–71.CrossRefPubMed Wohlgemuth S, et al. Reduced microbial diversity and high numbers of one single Escherichia coli strain in the intestine of colitic mice. Environ Microbiol. 2009;11(6):1562–71.CrossRefPubMed
46.
Zurück zum Zitat Aidar-Ugrinovich L, et al. Serotypes, virulence genes, and intimin types of Shiga toxin-producing Escherichia coli (STEC) and enteropathogenic E. coli (EPEC) isolated from calves in Sao Paulo, Brazil. Int J Food Microbiol. 2007;115(3):297–306.CrossRefPubMed Aidar-Ugrinovich L, et al. Serotypes, virulence genes, and intimin types of Shiga toxin-producing Escherichia coli (STEC) and enteropathogenic E. coli (EPEC) isolated from calves in Sao Paulo, Brazil. Int J Food Microbiol. 2007;115(3):297–306.CrossRefPubMed
47.
Zurück zum Zitat Molina-Lopez J, et al. Drug resistance, serotypes, and phylogenetic groups among uropathogenic Escherichia coli including O25-ST131 in Mexico City. J Infect Dev Ctries. 2011;5(12):840–9.PubMed Molina-Lopez J, et al. Drug resistance, serotypes, and phylogenetic groups among uropathogenic Escherichia coli including O25-ST131 in Mexico City. J Infect Dev Ctries. 2011;5(12):840–9.PubMed
48.
Zurück zum Zitat Morales-Espinosa R, et al. UPEC strain characterization isolated from Mexican patients with recurrent urinary infections. J Infect Dev Ctries. 2016;10(4):317–28.CrossRefPubMed Morales-Espinosa R, et al. UPEC strain characterization isolated from Mexican patients with recurrent urinary infections. J Infect Dev Ctries. 2016;10(4):317–28.CrossRefPubMed
49.
Zurück zum Zitat Budic M, et al. Escherichia coli bacteriocins: antimicrobial efficacy and prevalence among isolates from patients with bacteraemia. PLoS ONE. 2011;6(12):e28769.CrossRefPubMedPubMedCentral Budic M, et al. Escherichia coli bacteriocins: antimicrobial efficacy and prevalence among isolates from patients with bacteraemia. PLoS ONE. 2011;6(12):e28769.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Abraham S, et al. Molecular characterization of Escherichia coli strains that cause symptomatic and asymptomatic urinary tract infections. J Clin Microbiol. 2012;50(3):1027–30.CrossRefPubMedPubMedCentral Abraham S, et al. Molecular characterization of Escherichia coli strains that cause symptomatic and asymptomatic urinary tract infections. J Clin Microbiol. 2012;50(3):1027–30.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Micenkova L, et al. Bacteriocin-encoding genes and ExPEC virulence determinants are associated in human fecal Escherichia coli strains. BMC Microbiol. 2014;14:109.CrossRefPubMedPubMedCentral Micenkova L, et al. Bacteriocin-encoding genes and ExPEC virulence determinants are associated in human fecal Escherichia coli strains. BMC Microbiol. 2014;14:109.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Micenkova L, et al. Microcin determinants are associated with B2 phylogroup of human fecal Escherichia coli isolates. Microbiologyopen. 2016;5(3):490–8.CrossRefPubMedPubMedCentral Micenkova L, et al. Microcin determinants are associated with B2 phylogroup of human fecal Escherichia coli isolates. Microbiologyopen. 2016;5(3):490–8.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Azpiroz MF, Poey ME, Lavina M. Microcins and urovirulence in Escherichia coli. Microb Pathog. 2009;47(5):274–80.CrossRefPubMed Azpiroz MF, Poey ME, Lavina M. Microcins and urovirulence in Escherichia coli. Microb Pathog. 2009;47(5):274–80.CrossRefPubMed
Metadaten
Titel
Cytotoxic Escherichia coli strains encoding colibactin and cytotoxic necrotizing factor (CNF) colonize laboratory macaques
verfasst von
Yan Feng
Anthony Mannion
Carolyn M. Madden
Alton G. Swennes
Catherine Townes
Charles Byrd
Robert P. Marini
James G. Fox
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Gut Pathogens / Ausgabe 1/2017
Elektronische ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-017-0220-y

Weitere Artikel der Ausgabe 1/2017

Gut Pathogens 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

RAS-Blocker bei Hyperkaliämie möglichst nicht sofort absetzen

14.05.2024 Hyperkaliämie Nachrichten

Bei ausgeprägter Nierenfunktionsstörung steigen unter der Einnahme von Renin-Angiotensin-System(RAS)-Hemmstoffen nicht selten die Serumkaliumspiegel. Was in diesem Fall zu tun ist, erklärte Prof. Jürgen Floege beim diesjährigen Allgemeinmedizin-Update-Seminar.

Gestationsdiabetes: In der zweiten Schwangerschaft folgenreicher als in der ersten

13.05.2024 Gestationsdiabetes Nachrichten

Das Risiko, nach einem Gestationsdiabetes einen Typ-2-Diabetes zu entwickeln, hängt nicht nur von der Zahl, sondern auch von der Reihenfolge der betroffenen Schwangerschaften ab.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.