Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Food Microbiology 102 (2022) 103902

Contents lists available at ScienceDirect

Food Microbiology
journal homepage: www.elsevier.com/locate/fm

Biofilm formation by Non-O157 Shiga toxin-producing Escherichia coli in


monocultures and co-cultures with meat processing surface bacteria
Yuan Fang, Jeyachchandran Visvalingam 1, Peipei Zhang, Xianqin Yang *
Agriculture and Agri-Food Canada Lacombe Research and Development Centre, 6000 C & E Trail, Lacombe, Alberta, T4L 1W1, Canada

A R T I C L E I N F O A B S T R A C T

Keywords: This study investigated the impact of meat processing surface bacteria (MPB) on biofilm formation by non-O157
Non-O157 STEC Shiga toxin-producing Escherichia coli (STEC), and potential links between biofilm formation by STEC and
Co-culture biofilms biofilm-related genes in their genomes. Biofilm development by 50 MPB and 6 STEC strains in mono- and co-
Genes related to biofilm formation
cultures was assessed by the crystal violet staining method, and their expression of curli and cellulose was
Meat processing surface bacteria
determined using the Congo red agar method. Genes (n = 141) associated with biofilm formation in the STEC
strains were profiled. Biofilm formation in general correlated with cellulose and curli expression in both mono-
and co-cultures. Most MPB strains had antagonistic effects on the biofilm formation of the STEC strains. Of the
genes investigated, 81% were common among the STEC strains and there seems to be a gene-redundancy in
biofilm formation. The inability of the O26 strain to form biofilms could be due to mutations in the rpoS gene.
Truncation in the mlrA gene in the O145 strain seems not affecting its biofilm formation alone or with MPB. The
O45 strain, despite having the greatest number of biofilm-related genes, did not form measurable biofilms.
Overall, biofilm formation of STEC was affected by curli-cellulose expression and companion strains.

1. Introduction meat can occur by contacting processing equipment surfaces, such as


conveyor belts (Wang et al., 2018; Yang et al., 2017a, 2017b). Biofilm
Shiga toxin-producing Escherichia coli (STEC) are major foodborne formation may play a role in the recurring contamination of meat with
pathogens, causing sporadic and outbreak cases of illness (Crim et al., E. coli O157:H7, leading to “high event period”, a time period when beef
2015). Hemolytic uremic syndrome can be developed in 5–10% cases of processing establishments experience an increased occurrence of prod­
STEC infections (Bielaszewska et al., 2007). E. coli O157:H7/NM and six uct contamination by E. coli O157:H7 (Wang et al., 2014). Biofilm is a
additional serotypes of E. coli, known as the “Big Six”, including O26, structured bacterial community of single or multiple species that adhere
O45, O103, O111, O121, and O145, are the most common serotypes to biotic or abiotic surfaces with self-produced extracellular polymeric
associated with human illnesses in North America (Crim et al., 2015). substances (EPS) (Costerton et al., 1999). Floors, walls, and drains in
Illnesses due to non-O157 STEC strains are increasing worldwide food processing facilities with routine sanitation have been reported to
(Hadler et al., 2011). Estimates from the US Center for Disease Control be prone to biofilm formation (Sinde and Carballo, 2000). Biofilms, in
and Prevention (CDC) indicated that illness cases caused by non-O157 general, are more resistant than their planktonic counterparts to bio­
STEC strains as a group outnumber the cases due to O157 (Hadler cides commonly used for cleaning and sanitizating, making the elimi­
et al., 2011). nation of biofilms in such environment challenging (Wang et al., 2012,
STEC, in general, colonize the colon of the gastrointestinal tract of 2013).
cattle, but do not cause symptoms in cattle, and as such are found in Bacteria develop biofilms in a complex and coordinated manner that
feces and on hides of animals presented for slaughter (Barkocy-gallagher involves sensing and responding to changes in, including but not limited
et al., 2003). Transmission of bacteria from hides to skinned carcasses is to, the cell density, nutrient, accompanying species/strains, and envi­
inevitable during the dressing process (Bell, 1997). Some bacteria may ronmental stress (Dourou et al., 2011; Uhlich et al., 2014). These tran­
also persist in/on meat processing equipment, where recontamination of sitions are coordinated by multiple genetic pathways, including flagella

* Corresponding author.
E-mail address: xianqin.yang@canada.ca (X. Yang).
1
Current address: Kane Biotech Inc, 290-100 Innovation Drive, Winnipeg, Manitoba, R3T 2N2, Canada.

https://doi.org/10.1016/j.fm.2021.103902
Received 17 March 2021; Received in revised form 10 August 2021; Accepted 10 September 2021
Available online 14 September 2021
0740-0020/Crown Copyright © 2021 Published by Elsevier Ltd. All rights reserved.
Y. Fang et al. Food Microbiology 102 (2022) 103902

motility, cell-cell signaling, bio-surfactant secretion, cellulose, and curli A. haemolyticus, F. columnare, P. ginsengisoli, and A. urinaeequi, which did
biosynthesis (Chapman et al., 2002; Serra et al., 2013a, 2013b). As not grow well in LB-NS and thus were grown in half-strength Brain Heart
important structural elements of biofilms, cellulose provides cohesion Infusion (BHI) broth (Oxoid). Stationary-phase cultures were obtained
and elasticity that together with cell network encased by curli supports by incubation at 25 ◦ C with shaking at 80 rpm for 24–120 h (Visva­
the formation of high ridges and radial wrinkles (Serra et al., 2013a). lingam et al., 2019b).
Biofilm formation by STEC and the abundance of genes associated with
biofilms are highly strain-dependent (Biscola et al., 2011; Wang et al., 2.2. Biofilm development and quantification
2012). Moreover, in real-world scenarios, biofilms may be inhabited by
multiple strains/species where spatial and metabolic interactions To prepare monoculture inoculum, bacterial cultures were diluted
contribute to biofilm properties (Chorianopoulos et al., 2010; Habimana using LB-NS to obtain a cell density of ca. 107 CFU/mL. For bacteria
et al., 2010). When STEC O157:H7 and Salmonella enterica were grown in half-strength BHI broth, 1 mL of culture was centrifuged at
co-cultured with microorganisms from meat processing plants, the bio­ 10,000×g for 10 min at 4 ◦ C, and then, the cell pellets were suspended in
film formation was substantially affected by the environmental bacteria, 1 mL of LB-NS and further diluted using LB-NS. The co-culture inocula
some of which favored the non-biofilm producer STEC O157:H7 to form were prepared by mixing two bacterial suspensions in equal volumes.
biofilms, and may enhance their persistence in meat processing plants Biofilms were developed using 96-well round-bottom microtiter
(Visvalingam et al., 2019a). Information on the effect of bacteria from plates (Nunc Immuno TSP lid; Fisher) fitted with pegged lids (Nunc;
meat processing surface on the biofilm development by non-O157 STEC Fisher) and quantified using the crystal violet (CV) dye method (Vis­
and the genetic determinants of their biofilm formation is scarce. valingam et al., 2019b). Briefly, 160 μL of bacterial culture was added
Therefore, the objectives of this study were to i) assess the into the wells of a 96-well plate in duplicate. LB-NS served as blank.
biofilm-forming ability of the “Big Six” non-O157 STEC in single-species Inoculated plates were fitted with pegged lids and incubated at 15 ◦ C for
cultures and co-cultures with bacteria isolated from meat processing up to 6 days. Biofilms attached to the pegs were quantified by their
surface, and ii) elucidate the genetic differences associated with the ability to take up CV on day 2 and 6. The pegs were washed twice using
biofilm formation in these non-O157 STEC strains. 200 μL/well of phosphate-buffered saline (PBS, Hardy Diagnostics,
Santa Maria, CA), followed by staining with 180 μL of 0.1% (w/vol) CV
2. Materials and methods solution for 20 min. Then, the pegs were washed three times with PBS
and immersed into 180 μL/well of 80% ethanol for 20 min to dissolve
2.1. Bacterial strains and culture conditions the CV absorbed by the biofilms on the pegs. The absorbance of the
resulting solution in each well was measured at 570 nm using a micro­
Six non-O157 STEC strains recovered from beef carcasses were plate reader (Polarstar Omega, BMG LABTECH GmbH, Ortenberg, Ger­
included, and these strains were kindly provided by the Meat Animal many). When the absorbance value of a sample at 570 nm (A570) was
Research Center of the US Department of Agriculture (USDA) (Table 1). greater than 4, the sample was further diluted by 2-fold in 80% ethanol,
STEC O26:H11, O45:H2, O103:H2, O111:H8, O121:H19 and O145:H28 and the absorbance of which was determined. The results were calcu­
were referred to as O26, O45, O103, O111, O121, and O145 in this lated as mean ± standard deviation (SD) of two independent replicates,
study, respectively. Meat processing surface bacteria (MPB) included 18 with each containing duplicate samples (n = 4).
Gram-negative aerobic bacteria (GNA), 8 Gram-positive aerobic bacteria Biofilm-forming ability of tested bacterial strains was classified to
(GPA), 5 lactic acid bacteria (LAB), 9 Enterobacteriaceae (ENT) (Wang different levels based on the method described previously, with modi­
et al., 2018), and 10 generic E. coli (GEC) (Tables 1 and 2) (Yang et al., fications (Stepanovic et al., 2007). Briefly, the A570 of three times the
2015, 2017a, 2017b). The GEC strains were genotyped by multiple-locus standard deviation of the blank was used as the cut-off value (ODc),
variable-number tandem-repeat analysis and defined as non-persistent which was 0.05 in this study. The A570 ranged from − 0.05 ± 0.03 to
or persistent if they were recovered only on a single sampling or more 12.17 ± 0.45. Based on the calculation of 2n × ODc, ,the strains were
than three sampling visits from the same facility, respectively (Table 1) regarded as non-biofilm former (A570 ≤ 0.05), weak biofilm former
(Yang et al., 2015). (0.05 < A570 ≤ 0.8), moderate biofilm former (0.8 < A570 ≤ 1.6), strong
All bacteria were cultivated in Lennox broth without salt (LB-NS) biofilm former (1.6 < A570 ≤ 3.2), very strong biofilm former (3.2 <
(Oxoid Canada Inc., Mississauga, ON, Canada) except for A570 ≤ 6.4), and extremely strong biofilm former (6.4 < A570 ≤ 12.8).

Table 1 2.3. Expression of cellulose/cellulose-like, and curli/curli-like substances


E. coli strains used in this study.
Group Strain ID Serotype Original strain Origin of strains
Expression of the extracellular polymeric substances cellulose and
used in this ID/genotype thin aggregative fimbriae (curli) by MPB and STEC was evaluated using
study the Congo red indicator (CRI) method (Visvalingam et al., 2017; Wang
Non-O157 O26 O26:H11 Imp 133.1 USDA et al., 2013). Overnight cultures were streaked on CRI plates, followed
STEC O45 O45:H2 F-A 222.5 by incubation at 15 ◦ C for up to 4 days. After incubation, strains were
O103 O103:H2 Mar 125B determined as positive for both cellulose and curli, for curli, for cellu­
O111 O111:H8 C4-462-2_7095
lose, or neither if the colonies on CRI plates were red, brown, pink, or
O121 O121:H19 V2-G2 1-C 16.3
O145 O145:H28 May 109
white, respectively (Uhlich et al., 2014; Zogaj et al., 2001). As the
method has been primarily validated for members of Enterobacteriaceae,
Persistent B0 136 (Yang et al.,
cellulose/cellulose like and curli/curli-like substances were used for
E. coli B1 390 2015, 2017a,
B2 Generic 179 2017b) MPB.
E. coli
B3 541 2.4. Whole genome sequencing and genome assembly of non-O157 STEC
B4 138
strains
Non- B5 518
persistent B6 533
E. coli B7 Generic 545 The six STEC strains were sequenced to explore the potential genes
E. coli contributing to their distinct biofilm forming abilities. Briefly, DNA was
B8 115 extracted from the overnight culture of each STEC strain using a Mas­
B9 235
terPure™ Complete DNA and RNA purification kit (Lucigen, Middleton,

2
Y. Fang et al. Food Microbiology 102 (2022) 103902

Table 2
Bacterial strains isolated from beef processing equipment surfaces and their production of curli/curli-like substances and cellulose/cellulose like substances.
Group Strain ID used in this study Genus/species Original strain ID Curli/like substances Cellulose/like substances Origin of strains

GNA B11 Acidovorax wohlfahrtii 03–11 – + Wang et al. (2018)


B12 Acinetobacter haemolyticus 03–04 – –
B13 Aeromonas sp. 03–09 + –
B14 Brevundimonas staleyi 25–3 ND
B15 Chryseobacterium shigense 25–54 ND
B16 Comamonas koreensis 25–64 – +
B17 Flavobacterium columnare 09–7 – –
B18 Janthinobacterium lividum 09–39 – +
B19 Massilia aurea 03–22 ND
B20 Moraxella osloensis 25–60 – –
B21 Paracoccus aminophilus 09–12 – +
B22 Pedobacter ginsengisoli 03–52 – –
B23 Pseudomonas poae 09–41 + –
B24 Psychrobacter sp. 03–65 – +
B25 Sphingomonas 03–14 ND
B26 Sphingopyxis sp. 03–68 ND
B27 Stenotrophomonas maltophilia 25–39 + –
B28 Acinetobacter lwoffii 09–3 – +

GPA B29 Microbacterium sp. 03–50 – –


B30 Sanguibacter insulinus 25–11 – –
B31 Luteococcus japonicus 03–33 + –
B32 Macrococcus caseolyticus 25–15 + +
B33 Microbacterium phyllosphaerae 09–58 ND
B34 Microbacterium phyllosphaerae 03–25 ND
B35 Plantibacter sp. 09–65 ND
B36 Pseudoclavibacter helvolus 25–1 ND

LAB B37 Aerococcus urinaeequi LAB25-13 – –


B38 Carnobacterium maltaromaticum LAB9-67 – –
B39 Enterococcus sp. LAB3-8 – –
B40 Streptococcus parauberis LAB25-2 ND
B41 Vagococcus fluvialis LAB25-11 ND

ENT B42 Buttiauxella agrestis ENT25-12 – +


B43 Citrobacter gillenii ENT09-29 + +
B44 Enterobacter sp. ENT03-16 – +
B45 Hafnia alvei ENT03-7 – +
B46 Buttiauxella noackiae ENT09-55 – +
B47 Raoultella sp. ENT25-16 + –
B48 Serratia sp. ENT25-20 – +
B49 Yersinia sp. ENT09-26 – +
B50 Salmonella sp. ENT25-1 – +

ND: not determined. No conclusion was made due to the color-reduction of CRI plates by bacteria.

WI, US). The sequencing library was prepared using a NEBNext® Ultra™ for each isolate/genome is listed in Table S1.
II DNA Library Prep Kit, and subsequently sequenced on an Illumina
HiSeq4000 instrument with sequencing depth >300 for 150 × 2 cycles
2.5. Analysis of genes associated with biofilm formation in the STEC
(Genome Quebec, QC, CA). The quality of raw sequencing reads was
genomes
examined using FastQC v0.11.8. The contamination of Phix sequences in
the reads was removed using Bowtie v2.3.4.3 (Langmead and Salzberg,
Genes encoding proteins related to biofilm formation in E. coli were
2012). Trimmomatic 0.39 was used to remove adapter sequences and
collected from GenBank and made into a local database, including genes
sequences with an average quality score <20 or length <100 bases
involved in the biosynthesis of colonic acid, curli, cellulose, β-1,6-N-
(Bolger et al., 2014). Each genome was assembled using SPAdes v3.14.0
acetylglucosamine (PGA), adhesion factors, and quorum sensing
with the size of kmers set at 21, 33, 55, 77, 99, and 127 (Bankevich et al.,
(Table S2). Annotated STEC genomes were searched against the local
2012). The quality of the assembled genomes was assessed using Quast
database using BLASTp (2.6.0+) to identify the genes in each genome,
v5.0.2 (Gurevich et al., 2013). Contigs with length <500 bp or coverage
with >70% identity and >70% coverage of the reference amino acid
<10 were removed (Douglass et al., 2019). The remaining contigs in
sequences (Joshi and Xu, 2007).
each genome were ordered using Mauve (Darling et al., 2004) by
mapping against respective serotypes of STEC, the complete genomes of
which are available in the database of the National Center for Biotech­ 2.6. Data analysis
nology Information (NCBI). The reference strains were E. coli O26:H11
(GenBank assembly accession no: GCA_000091005.1) (Smith et al., The A570 values for the same strain at different incubation times, or a
2017), O45:H2 (GCA_005037845.1), O103:H2 (GCA_000010745.1), MPB strain in mono-cultures and in combination with different STEC
O111:H8 (GCA_003288295.1) (Sekizuka et al., 2019), O121:H19 strains at the same incubation time were analyzed by one-way analysis
(GCA_002844165.1) (Robertson et al., 2018), and O145:H28 of variance (ANOVA). Statistical differences among bacterial combina­
(GCA_000520035.1). The open reading frames (ORFs) in each genome tions and between incubation times were determined by Tukey’s test
were searched using Prodigal (v2.6.3) with default settings (Hyatt et al., using SPSS software, version 21.0 (SPSS Inc., Chicago, IL, USA). To
2010). The raw sequencing reads and draft genomes were submitted to determine the correlation between extracellular property and biofilm
GenBank under the BioProject PRJNA706412, and the accession number forming ability, the A570 values of groups of strains that associated with
the different numbers of curli/curli-like substances and/or cellulose/

3
Y. Fang et al. Food Microbiology 102 (2022) 103902

cellulose-like substances were pairwise compared using Kruskal-Wallis showed STEC strain dependent biofilm formation on day 6 (Fig. 3). The
test. A significance level of P < 0.05 was used for all statistical analysis. pattern of biofilm formation of co-cultures of O145, O103, O26 and O45
The interactions between two strains on biofilm formation were with LAB was similar to that of the respective STEC monocultures.
regarded as synergistic, no effect or antagonistic if the A570 value of co- However, the co-cultures of O111 or O121 with various LAB strains all
culture is larger than, equals to, or smaller than the higher A570 value of had significantly lower A570 values except for the combination of B41
the two relevant monocultures (Ren et al., 2015). (V. fluvialis) and O111, compared to their monoculture counterparts. In
contrast to LAB, the biofilm production of STEC-ENT co-cultures was
3. Results consistent with the biofilm-forming ability of the ENT strains, in a STEC
strain independent manner (Fig. 3). This trend in that more co-culture
3.1. Biofilm formation by monocultures of STEC and their co-cultures biofilms of O111 and O121 were in the weak category than those of
with GPA O145 and O103 was consistent with their respective co-cultures with
GPA and GNA.
The biofilm-forming ability of STEC strains in monocultures varied
from non biofilm-forming (O45) to extremely strong biofilm-forming 3.4. Biofilm formation by co-cultures of STEC and persistent or non-
(O103 and O145), with the latter two strains having A570 values > 8.0 persistent E. coli
on day 6 (Table 3). The five biofilm forming strains all had significantly
higher (P < 0.05) A570 values on day 6 than on day 2. Different from co-cultures of STEC with GPA, LAB or ENT, 30% (two
Generally, GPA produced no biofilms (A570<0.05) on day 2 and weak persistent and one non-persistent strains) of GEC and STEC formed very/
biofilms (A570<0.8) on day 6, except for B32 (M. caseolyticus), which extremely strong co-culture biofilms on day 2, irrespective of the STEC
produced strong biofilms on day 6 (Fig. 1). On day 2, the A570 values for strains (Fig. 4). Co-culture GEC and STEC O103 and O145 were uniform
the co-cultures of B32 and the STEC strains were higher than the A570 on forming very/extremely strong biofilms; however, the biofilm pro­
values for the co-cultures of other GPA and STEC strains. Of the 48 co- duction of the co-cultures including other serotypes of STEC exhibited a
cultures on day 6, 24 developed extremely strong biofilms (A570 > high degree of variation depending on the strains of GEC involved,
6.4), 62.5% of which involved either O103 or O145. B33 particularly for non-persistent GEC. Biofilms of persistent GEC (B0-4) in
(M. phyllosphaerae) and O45, both of which were non-biofilm formers on monocultures and co-cultures with STEC were very/extremely strong in
their own, formed extremely strong biofilms in their co-cultures on day all cases. However, biofilm formation of non-persistent GEC (B5-9)
6. On the other hand, the A570 values for the day 6 co-culture biofilms of varied by non-persistent GEC and STEC. Some non-persistent GEC (B6
O111 or O121 with B30 (S. insulinus) or B36 (P. helvolus), a non-biofilm and B7) developed very/extremely strong biofilms in monocultures and
former on its own, were much lower than the A570 values for their together with STEC. Two non-persistent GEC (B5 and B8) did not form
respective STEC mono-cultures, which were both in the strong biofilm measurable biofilms by themselves or when co-cultures with O111 or
category. Compared to the O103 and O145, O111 and O121 were more O121 after 6 days, despite that O111 and O121 both formed strong
negatively impacted by the companion GPA strains. biofilms at equivalent times.

3.5. Interactions between STEC and MPB on biofilm formation


3.2. Biofilm formation by co-cultures of STEC and GNA
The interactions between STEC and MPB in biofilm formation were
Different from GPA, a higher fraction of GNA developed measurable
largely antagonistic, as was observed with 78%, 90%, 81%, and 77% of
biofilms on both day 2 and day 6. Unlike the uniformity of co-cultures of
the GNA, LAB, ENT, and GEC strains, respectively (Fig. 5). The greatest
O145 or O103 with GPA in their strong biofilm formation, their
antagonistic effects were observed in members of GNA and ENT, in
respective co-cultures with GNA show a much higher degree of variation
particular B18 (J. lividum) and B19 (M. aurea), as well as B42 (B. agrestis)
in this regard, with A570 ranging from much lower to much higher than
and B44 (Enterbacter sp.), for which no synergistic effects were observed
that of the respective monocultures (Fig. 2). Even so, as with the GPA-
in any of the combinations with STEC. The greatest synergistic effects
STEC co-culture biofilms, the co-culture biofilms by O103 or O145
were found in the co-cultures with GPA strains, namely B33
with GNA were mostly stronger than the co-culture biofilms by the other
(M. phyllosphaerae) and B34 (M. phyllosphaerae), with differences in the
STEC strains. Among the GNA, B18 (J. lividum) and B11 (A. wohlfahrtii)
A570 values being >10.
developed non- or weak mono-culture and STEC-co-culture biofilms
even with those strong biofilm forming STEC strains.
3.6. Cellulose and curli expression by non-O157 STEC and MPB strains

3.3. Biofilm formation by co-cultures of STEC and LAB or ENT E. coli O145, O103, O121, and O111 expressed both curli and cel­
lulose, while O26 expressed only cellulose and O45 exhibited only curli
Despite not being able to form detectable biofilms in monocultures expression. Of the 40 MPB strains, 11 had pigmentation and as such, CRI
on both day 2 and day 6, the co-cultures of the LAB strains (B37-41) could not be used to determine curli or cellulose production. For the
remaining MPB strains, curli (curli-like substance) and/or cellulose
Table 3 (cellulose-like substance) production was found in all ENT strains, 9/13
Biofilm formation by non-O157 Shiga toxin-producing E. coli strains. of GNA, 2/4 of GPA, and none of LAB, respectively. Of ENT, B43
Serotype Curli Cellulose Biofilm mass (A570)a (C. gillenii) and B47 (Raoultella sp.) were positive for both curli and
cellulose, and curli, respectively. All the other strains expressed only
Day 2 Day 6
cellulose. The extent of biofilm formation by mono- and co-cultures, as
O26 – + 0.04 ± 0.02 1.5 ± 0.04C* determined by A570 values, had a strong correlation (P < 0.05) with
O45 0 0D
these two types of extracellular substance production (Fig. 6).
+ –
O103 + + 0.06 ± 0.01 8.22 ± 1.09A*
O111 + + 0.01 ± 0 4.49 ± 0.8B*
O121 + + 0.03 ± 0.01 5.09 ± 0.29B* 3.7. Genome analysis of non-O157 STEC strains
O145 + + 0.6 ± 0.06 8.62 ± 0.49A*
a
Means in the same column that do not share a common superscript are Of the 141 genes that are potentially associated with biofilm for­
significantly different (P < 0.05). Asterisk indicates significant differences be­ mation in E. coli, 114 were found in all six STEC strains including the
tween means in the same row (P < 0.05). clusters bcs, csg, and wca encoding proteins involved in cellulose, curli,

4
Y. Fang et al. Food Microbiology 102 (2022) 103902

Fig. 1. Biofilm formation by non-O157 STEC strains in co-cultures with Gram-positive aerobic (GPA) bacteria recovered from beef processing equipment surfaces.
The biofilm formation was assessed by CV staining after incubation at 15 ◦ C for 2 and 6 days. Asterisks indicate significant differences in A570 values between the two
incubation days for the same strains. Significant differences among the GPA in monoculture and in combination with different STEC strains are labeled with different
capital letters (P < 0.05). The horizon dash lines are set at A570 at 1.6 (black) and 6.4 (red), representing the cut-off values for strong and extremely strong biofilms,
respectively. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

and colonic acid production, as well as genes encoding for a large The two extremely strong biofilm formers, O103 and O145, each had
number of known function or putative adhesins and fimbriae (Table S2). genes for the biosynthesis of surfactin and colicin (Fig. 7). The O145
The tibA coding for an adhesin/invasion, commonly found in entero­ strain had a truncated MlrA (Fig. S1) and the O26 strain differed by 40
toxigenic E. coli (Sherlock et al., 2005), was the only one that was not amino acids in its RpoS from the wild type strain ATCC 43895 and the
found in any of the strains. The 26 genes, which were differentially other STEC strains (Fig. S2).
abundant in the six STEC genomes, distributed differently, even between
the strains with similar biofilm forming phenotypes (Fig. 7). No unique 4. Discussion
genes were found specific to O45, the non-biofilm former, and in fact,
O45 had the least number of biofilm related genes missing in the genome Biofilm formation is an essential strategy for bacteria to survive
(5/141). under adverse conditions, including food-processing environments
Some components of the regulatory and functional genes related to (Burmølle et al., 2006; Habimana et al., 2010; Wang et al., 2013). Bio­
cellulose, c-di-GMP and N-acetyl-D-glucosamine (PGA) biosynthesis films developed by mixed-species rather than single species represent
were missing in some strains. The pgaABCD operon for PGA was present the most frequent form of microbiological contamination and persis­
in the genomes of E. coli O45, O103, and O145 but missing in O26, tence in food industries (Yuan et al., 2020). Observations made on
O111, and O121. The yhjR part of the bcs operon was missing only in monoculture biofilms might not reflect the mixed-species biofilm for­
E. coli O145. As part of the synthesis of signalling molecule c-di-GMP, mation as interspecies interactions may alter the behavior of the indi­
dgcT encoding for diguanylate cyclase was only present in the genomes vidual bacterial strains in biofilm formation (Burmølle et al., 2014). This
of E. coli O103 and E. coli O45, whereas dosCP encoding for phospho­ observation was confirmed in this study, which shows that biofilm for­
diesterase was only absent in E. coli O103. However, additional 9 genes mation of non-O157 STEC was differentially affected by bacterial strains
encoding for diguanylate cyclase were found in all six STEC genomes isolated from beef processing equipment surfaces. Importantly, this
(Table S2). study provides pertinent information on the biofilm formation in meat
Fimbriae encoding genes were more heterogeneous and diverse than processing environment by evaluating biofilm formation of 50 bacterial
genes coding for cellulose, curli, PGA, and quorum sensing (Fig. 7; strains isolated from beef processing plants, which was considered as a
Table S2). Genes coding for flagella filament (flhDC and fliC), cell representative composition of microbiome of meat processing environ­
motility (motAB and cheAC), and related genes were all present in the ment (Wang et al., 2018). The findings that biofilm formation of STEC
STEC strains. Genes related to cell-cell communication were present in strains was negatively or positively influenced by companion strains
all STEC genomes in most cases. lsrG encoding a cytoplasmic kinase highlight the importance, from a food safety standpoint, of addressing
responsible for phosphorylation of AI-2 into an active molecule biofilms under conditions simulating their natural ecological niches.
following its import into the cell was absent in O26. The gene encoding Production of cellulose and curli determines the morphology and
the autotransporter Ag43, which mediates cell-cell aggregation and in- biofilm strength in Enterobacteriaceae (Zogaj et al., 2001). Genome
turn leads to cell-aggregates, was not found in O111 or O121. analysis has demonstrated that functional amyloid systems are

5
Y. Fang et al. Food Microbiology 102 (2022) 103902

Fig. 2. Biofilm formation by non-O157 STEC strains in co-cultures with Gram-negative aerobic bacteria (GNA) recovered from beef processing equipment surface.
The biofilm formation was assessed by CV staining after incubation for 2 and 6 days. Asterisks indicate significant differences in A570 values between the two in­
cubation days for the same strains. Significant differences among the GNA in a single culture and combination with different STEC are labeled with different su­
perscript (P < 0.05). The horizon dash lines are set at A570 at 1.6 (black) and 6.4 (red), representing the cut-off values for strong and extremely strong biofilms,
respectively. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

wide-spread in bacteria (Dueholm et al., 2012), and that amyloids are c-di-GMP is controlled by the antagonistic activities between diguany­
abundant in natural biofilms (Larsen et al., 2007). Thus, this study used late cyclase with GGDEF domains and the phosphodiesterase with EAL
the Congo red assay to determine the extracellular substance production domains (Simm et al., 2004). However, E. coli encodes multiple proteins
in non-Enterobacteriaceae species as well as in Enterobacteriaceae. The containing GGDEF or EAL domains, making the synthesis and signaling
ability to uptake Congo red by MPB and STEC had a positive correlation network of c-di-GMP more complicated (García et al., 2004; Povolotsky
with the degree of their biofilm formation in mono- and co-cultures, as and Hengge, 2015; Weber et al., 2006). The redundancy of genes
determined by A570. This suggests that CRI can be a useful method for encoding diguanylate cyclases and phosphodiesterase was also observed
screening bacterial strains for biofilm forming potentials. in the six STEC genomes in this study and missing some of the genes
Despite the wide-spread of genes encoding functional amyloids in (dgcT/ycdT, dosP, dosC) in O111, O121, O103 and O145 did not impact
bacterial genomes (Biscola et al., 2011; Dueholm et al., 2012), the their curli or cellulose production or biofilm formation. In addition,
production of curli and cellulose varies with species and even strains, in production of curli and cellulose by Enterobacteriaceae was affected by
the case of E. coli (Visvalingam et al., 2017; Wang et al., 2012). The growth conditions through the differential expression of csgD and/or
strain-dependent curli and cellulose production was also noted in this adrA (García et al., 2004), and the level of c-di-GMP (Reinders et al.,
study (O26, curli-negative; O45, cellulose-negative; the other four 2015). These factors may have contributed to the findings that the O45
strains positive for both curli and cellulose), even though both O45 and strain did not produce cellulose on CRI despite having all the essential
O26 harbored essential genes for the biosynthesis of curli and cellulose. genes.
CsgD is a transcriptional regulator, which positively regulates the In mixed-species biofilms, microorganisms interact through jamming
expression of the structural genes (csgBC) and the export of proteins for of quorum sensing molecules (Jahid et al., 2018) and the production of
assembling curli (Brombacher et al., 2003). The diguanylate cyclase secondary metabolites or toxins that inhibit/promote the growth of
ArdA indirectly activates cellulose production (Zogaj et al., 2001). The coexisting microorganisms (Chorianopoulos et al., 2010; Habimana
transcription of CsgD is controlled by the alternative Sigma factor RpoS et al., 2010). Biofilm formation of a non-biofilm producer E. coli O157:
and MlrA, the latter of which binds to the former to enhance the H7 was enhanced by A. calcoaceticus in dual-species biofilms (Habimana
expression of CsgD; and mutations in RpoS and insertions in MlrA by the et al., 2010). In contrast, H. alvei and/or Serratia sp. inhibited the bio­
Stx1-encoding prophage have been attributed to the limited curli films formed by S. enterica and Listeria monocytogenes (Alavi et al., 2013;
expression and subsequent biofilm formation in some E. coli O157 Visvalingam et al., 2019b) and non-O157 STEC strains (this study). The
strains (Uhlich et al., 2013). The inability of the O26 strain to produce presence of unknown compounds in the culture supernatant of H. alvei
curli in this study may be attributable to the multiple mutations in the contributed to the inhibition of metabolic activity and biofilm produc­
RpoS in O26. Interestingly, a truncated MlrA did not seem to affect the tion by S. Typhimurium (Chorianopoulos et al., 2010). Production of
curli or cellulose production, or biofilm formation by O145. AdrA is antimicrobial compounds by Serratia plymuthica is one of the factors
responsible for the synthesis of the secondary signaling molecule contributing to its inhibitory effects on the biofilm formation by E. coli
c-di-GMP, which activates cellulose biosynthesis (bcsABEC and bcsEFG) MG1655, which is regulated by the quorum-sensing molecule
(Povolotsky and Hengge, 2015; Simm et al., 2004). The homeostasis of N-acyl-L-homoserine lactone (AHL) (Moons et al., 2006). Such

6
Y. Fang et al. Food Microbiology 102 (2022) 103902

Fig. 3. Biofilm formation by non-O157 STEC strains in co-cultures with lactic acid bacteria (LAB; B37-41) or Enterobacteriaceae (ENT; B42-50) recovered from beef
processing equipment surface. The biofilm formation was assessed by CV staining after incubation for 2 and 6 days. Asterisks indicate significant differences in A570
values between the two incubation days for the same strains. Significant differences among the LAB and ENT in a single and combination with different STEC are
labeled with different superscript (P < 0.05).). The horizon dash lines are set at A570 at 1.6 (black) and 6.4 (red), representing the cut-off values for strong and
extremely strong biofilms, respectively. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

antagonistic effects on the biofilm formation were also found in the aggregation and enhanced the biofilm formation by Pseudomonas fluo­
dual-species biofilms of Aeromonas sp. (B13) and E. coli O103, O111, rescens (Kjaergaard et al., 2000). Whether or not these factors play a role
O121 or O145, as well as of A. urinaeequi (B37) or C. maltaromaticum in the different patterns of biofilm formation in the STEC-MPB co-­
(B38) and E. coli O111 or O121 in this study. E. coli do not produce AHLs, cultures would warrant further investigations.
but produce autoinducer-2 (AI-2) as quorum sensing molecules (Walters Cell to cell adhesion also plays a role in biofilm formation (Van
and Sperandio, 2006). However, they can detect AHLs from other spe­ Houdt and Chris, 2005; Wang et al., 2004). The PGA polymers, fimbriae,
cies by a receptor-like protein (SdiA) (Lindsay and Ahmer, 2005), which membrane auto-transporters, and adhesins mediate cell-cell or cell to
can modulate their gene expression (Van Houdt et al., 2006). Different surface adhesion (Hammer and Bassler, 2003; Schembri et al., 2003;
from AHLs, AI-2 was employed to increase the biofilm formation in Wang et al., 2004). Type 1 fimbriae contributed to surface colonization
E. coli by stimulation of flagella motility via the motility quorum-sensing and biofilm formation of E. coli K12 (Beloin et al., 2004); however, the
regulator MqsR (González Barrios et al., 2006). Internalization of disruption of type 1 fimbriae did not affect the biofilm formation
extracellular AI-2 requires the phosphorylation by LsrK kinase, and then significantly (Reisner et al., 2003). Homologous sequences of fim were
the phosphorylated AI-2 is further processed by LsrG and LsrF (Xavier found in many E. coli, including yra, yfc, yad, and yeh, which promoted
and Bassler, 2005). The six STEC genomes harbored all genes for the biofilm formation in the absence of type 1 fimbriae (Korea et al., 2010).
AI-2 system and SdiA except for E. coli O26, which did not have LsrG. On the other hand, EhaA was physically shielded by the expression of
Despite this, different patterns in the biofilm formation by different type 1 fimbriae (Wells et al., 2008); flagella encoded by fliC were not
STEC strains and the same MPB were noted. In general, compared to the essential for biofilm formation when the curli was expressed (Pri­
A570 values for the respective strong biofilm forming STEC strains gent-Combaret et al., 2000). The depolarization of PGA resulted in the
(O145, O103, O121, and O111) in monocultures, the A570 values for the dispersal of biofilms of E. coli (Itoh et al., 2005). The pga operon was
O145 and O103 in co-cultures with MPB were less affected than those present in the extremely strong biofilm formers E. coli O103 and O145
for the MPB co-cultures with O121 and O111, where in many cases the and non-biofilm former E. coli O45, but not in the other two strong
co-cultures behaved similarly with the non-biofilm forming MPB (B5, biofilm formers E. coli O111 and O121. It suggested that other adhesion
B6, B11, B16, B20, B24, B30, B36–40, B44). The exact reason for this mechanisms are compensated to the PGA-dependent biofilm formation
behavior is unclear. One noted difference in the genomes was that O145 in E. coli O111 and O121.
and O103 each harbored a gene encoding for surfactin and colicin, the In conclusion, this study found that production of cellulose and curli
former of which is a biosurfactant and the latter a bacteriocin and both was positively correlated with the degree of biofilm formation in both
of which can inhibit biofilm formation of other bacterial species (Banat mono- and co-cultures of STEC and MPB. Although a plethora of genes
et al., 2014; Rendueles et al., 2014). In addition, O121 and O111 did not encoding for proteins involved in essential steps in biofilm formation
harbor flu, a gene encoding for the autotransporter Ag43. The exact role were present in all STEC genomes investigated; finesse of biofilm pro­
of Ag43 in the biofilm formation of E. coli is a matter for debate (Danese duction is likely on the regulatory level, rather than the presence or
et al., 2000; Taghadosi et al., 2017). However, Kjaergaard et al. absence of structural genes. Phenotypic characterization of biofilm
demonstrated that Ag43 induced intra- and inter-species cell formation under relevant conditions is then indispensable. Some MPB,

7
Y. Fang et al. Food Microbiology 102 (2022) 103902

Fig. 4. Biofilm formation by non-O157 STEC strains in co-cultures with persistent (B0-4) and non-persistent (B5-9) generic E. coli recovered a beef plant. The biofilm
formation was assessed by CV staining after incubation for 2 and 6 days. Asterisks indicate significant differences in A570 values between the two incubation days for
the same strains. Significant differences among the generic E. coli in a single culture and combination with different STEC are labeled with different superscript (P <
0.05). The horizon dash lines are set at A570 at 1.6 (black) and 6.4 (red), representing the cut-off values for strong and extremely strong biofilms, respectively. (For
interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

Fig. 5. Interactions between non-O157 STEC strains and MPB on the biofilm formation. The interactions were determined as synergistic, antagonistic, and neutral if
the A570(co-culture – mono-culture) is greater than, smaller than and equals to zero, respectively. Strains of B0-4, B5-9, B11–B28, B29–B36, B37–41, B42-50, are groups of
persistent and non-persistent E. coli, GNA, GPA, LAB, and ENT, respectively.

particularly some members of Gram-negative aerobic bacteria, lactic and health effects.
acid bacteria, and Enterobacteriaceae have strong antagonistic effects on
the biofilm formation by various STEC strains, which could be further
explored as biocontrol agents for biofilms, depending on their and safety

8
Y. Fang et al. Food Microbiology 102 (2022) 103902

Fig. 6. Correlation between the biofilm formation and expression of curli and cellulose in mono-cultures (Panel A) and dual-cultures (B). Phenotype for mono-
cultures is defined as 0 (no curli or cellulose), 1 (curli or cellulose), and 2 (curli and cellulose). For dual-cultures in panel B, number 1–4 was defined as the
number of positive determinants expressed by the two bacterial strains in the dual-culture.

Fig. 7. The presence (Y; light blue) or absence (N; dark blue) of genes related to biofilm formation which showed difference in distribution in the six STEC genomes.
(For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

Declaration of competing interest Appendix A. Supplementary data

None. Supplementary data to this article can be found online at https://doi.


org/10.1016/j.fm.2021.103902.
Acknowledgments
References
Funding for this study was provided by Agriculture and Agri-Food
Canada (A1637 and A1603) and the Beef Cattle Research Council of Alavi, D., Edin, H., Truelstrup Hansen, L., 2013. Kinetics of biofilm formation and
desiccation survival of Listeria monocytogenes in single and dual species biofilms with
Canada (FOS.01.17). Pseudomonas fluorescens, Serratia proteamaculans or Shewanella baltica on food-grade
stainless steel surfaces. Biofouling 29, 1253–1268.

9
Y. Fang et al. Food Microbiology 102 (2022) 103902

Banat, I.M., De Rienzo, M.A., Quinn, G.A., 2014. Microbial biofilms: biosurfactants as Itoh, Y., Wang, X., Hinnebusch, B.J., Preston, J.F., Romeo, T., 2005. Depolymerization of
antibiofilm agents. Appl. Microbiol. Biotechnol. 98, 9915–9929. β-1,6-N-acetyl-D-glucosamine disrupts the Integrity of diverse bacterial biofilms.
Bankevich, A., Nurk, S., Antipov, D., Gurevich, A.A., Dvorkin, M., Kulikov, A.S., Lesin, V. J. Bacteriol. 187, 382–387.
M., Nikolenko, S.I., Pham, S., Prjibelski, A.D., Pyshkin, A.V., Sirotkin, A.V., Jahid, I.K., Mizan, M.F.R., Myoung, J., Ha, S.-D., 2018. Aeromonas hydrophila biofilm,
Vyahhi, N., Tesler, G., Alekseyev, M.A., Pevzner, P.A., 2012. SPAdes: a new genome exoprotease, and quorum sensing responses to co-cultivation with diverse foodborne
assembly algorithm and its applications to single-cell sequencing. J. Comput. Biol. pathogens and food spoilage bacteria on crab surfaces. Biofouling 34, 1079–1092.
19, 455–477. Joshi, T., Xu, D., 2007. Quantitative assessment of relationship between sequence
Barkocy-gallagher, G.A., Arthur, T.M., Rivera-betancourt, M., Nou, X., Shackelford, S.D., similarity and function similarity. BMC Genom. 8, 222.
Wheeler, T.l., Koohmaraie, M., 2003. Seasonal prevalence of Shiga toxin–producing Kjaergaard, K., Schembri, M.A., Hasman, H., Klemm, P., 2000. Antigen 43 from
Escherichia coli, Including O157:H7 and Non-O157 serotypes, and Salmonella in Escherichia coli induces inter- and intraspecies cell aggregation and changes in colony
commercial beef processing plants. J. Food Protect. 66, 1978–1986. morphology of Pseudomonas fluorescens. J. Bacteriol. 182, 4789–4796.
Bell, R.G., 1997. Distribution and sources of microbial contamination on beef carcasses. Korea, C.-G., Badouraly, R., Prevost, M.-C., Ghigo, J.-M., Beloin, C., 2010. Escherichia coli
J. Appl. Microbiol. 82, 292–300. K-12 possesses multiple cryptic but functional chaperone–usher fimbriae with
Beloin, C., Valle, J., Latour-Lambert, P., Faure, P., Kzreminski, M., Balestrino, D., distinct surface specificities. Environ. Microbiol. 12, 1957–1977.
Haagensen, J.A.J., Molin, S., Prensier, G., Arbeille, B., Ghigo, J.-M., 2004. Global Langmead, B., Salzberg, S.L., 2012. Fast gapped-read alignment with Bowtie 2. Nat.
impact of mature biofilm lifestyle on Escherichia coli K-12 gene expression. Mol. Methods 9, 357–359.
Microbiol. 51, 659–674. Larsen, P., Nielsen, J.L., Dueholm, M.S., Wetzel, R., Otzen, D., Nielsen, P.H., 2007.
Bielaszewska, M., Köck, R., Friedrich, A.W., von Eiff, C., Zimmerhackl, L.B., Karch, H., Amyloid adhesins are abundant in natural biofilms. Environ. Microbiol. 9,
Mellmann, A., 2007. Shiga toxin-mediated hemolytic uremic syndrome: time to 3077–3090.
change the diagnostic paradigm? PloS One 2, e1024. Lindsay, A., Ahmer, B.M.M., 2005. Effect of sdiA on biosensors of N-acylhomoserine
Biscola, F.T., Abe, C.M., Guth, B.E., 2011. Determination of adhesin gene sequences in, lactones. J. Bacteriol. 187, 5054–5058.
and biofilm formation by, O157 and non-O157 Shiga toxin-producing Escherichia coli Moons, P., Van Houdt, R., Aertsen, A., Vanoirbeek, K., Engelborghs, Y., Michiels, C.W.,
strains isolated from different sources. Appl. Environ. Microbiol. 77, 2201–2208. 2006. Role of quorum sensing and antimicrobial component production by Serratia
Bolger, A.M., Lohse, M., Usadel, B., 2014. Trimmomatic: a flexible trimmer for Illumina plymuthica in formation of biofilms, including mixed biofilms with Escherichia coli.
sequence data. Bioinformatics 30, 2114–2120. Appl. Environ. Microbiol. 72, 7294–7300.
Brombacher, E., Dorel, C., Zehnder, A.J.B., Landini, P., 2003. The curli biosynthesis Povolotsky, T.L., Hengge, R., 2015. Genome-based comparison of cyclic di-GMP
regulator CsgD co-ordinates the expression of both positive and negative signaling in pathogenic and commensal Escherichia coli strains. J. Bacteriol. 198,
determinants for biofilm formation in Escherichia coli. Microbiol. 149, 2847–2857. 111–126.
Burmølle, M., Ren, D., Bjarnsholt, T., Sørensen, S.J., 2014. Interactions in multispecies Prigent-Combaret, C., Prensier, G., Le Thi, T.T., Vidal, O., Lejeune, P., Dorel, C., 2000.
biofilms: do they actually matter? Trends Microbiol. 22, 84–91. Developmental pathway for biofilm formation in curli-producing Escherichia coli
Burmølle, M., Webb, J.S., Rao, D., Hansen, L.H., Sørensen, S.J., Kjelleberg, S., 2006. strains: role of flagella, curli and colanic acid. Environ. Microbiol. 2, 450–464.
Enhanced biofilm formation and increased resistance to antimicrobial agents and Reinders, A., Hee, C.-S., Ozaki, S., Mazur, A., Boehm, A., Schirmer, T., Jenal, U., 2015.
bacterial invasion are caused by synergistic interactions in multispecies biofilms. Expression and genetic activation of cyclic di-GMP-specific phosphodiesterases in
Appl. Environ. Microbiol. 72, 3916–3923. Escherichia coli. J. Bacteriol. 198, 448–462.
Chapman, M.R., Robinson, L.S., Pinkner, J.S., Roth, R., Heuser, J., Hammar, M., Reisner, A., Haagensen, J.A.J., Schembri, M.A., Zechner, E.L., Molin, S., 2003.
Normark, S., Hultgren, S.J., 2002. Role of Escherichia coli curli operons in directing Development and maturation of Escherichia coli K-12 biofilms. Mol. Microbiol. 48,
amyloid fiber formation. Science 295, 851–855. 933–946.
Chorianopoulos, N.G., Giaouris, E.D., Kourkoutas, Y., Nychas, G.J., 2010. Inhibition of Ren, D., Madsen, J.S., Sørensen, S.J., Burmølle, M., 2015. High prevalence of biofilm
the early stage of Salmonella enterica serovar Enteritidis biofilm development on synergy among bacterial soil isolates in cocultures indicates bacterial interspecific
stainless steel by cell-free supernatant of a Hafnia alvei culture. Appl. Environ. cooperation. ISME J. 9, 81–89.
Microbiol. 76, 2018–2022. Rendueles, O., Beloin, C., Latour-Lambert, P., Ghigo, J.M., 2014. A new biofilm-
Costerton, J.W., Stewart, P.S., Greenberg, E.P., 1999. Bacterial biofilms: a common cause associated colicin with increased efficiency against biofilm bacteria. ISME J. 8,
of persistent infections. Science 284, 1318–1322. 1275–1288.
Crim, S.M., Griffin, P.M., Tauxe, R., Marder, E.P., Gilliss, D., Cronquist, A.B., Cartter, M., Robertson, J., Lin, J., Levett, P.N., Nadon, C., Nash, J., Berry, C., 2018. Complete genome
Tobin-D’Angelo, M., Blythe, D., Smith, K., Lathrop, S., Zansky, S., Cieslak, P.R., sequence of an Escherichia coli O121:H19 strain from an outbreak in Canada
Dunn, J., Holt, K.G., Wolpert, B., Henao, O.L., Centers for Disease, C., Prevention, associated with flour. Genome Announc. 6, e01561, 1517.
2015. Preliminary incidence and trends of infection with pathogens transmitted Schembri, M.A., Hjerrild, L., Gjermansen, M., Klemm, P., 2003. Differential expression of
commonly through food - foodborne Diseases Active Surveillance Network, 10 U.S. the Escherichia coli autoaggregation factor antigen 43. J. Bacteriol. 185, 2236–2242.
sites, 2006-2014. MMWR (Morb. Mortal. Wkly. Rep.) 64, 495–499. Sekizuka, T., Lee, K., Kimata, K., Isobe, J., Kuroda, M., Iyoda, S., Ohnishi, M., Sata, T.,
Danese, P.N., Pratt, L.A., Dove, S.L., Kolter, R., 2000. The outer membrane protein, Watahiki, M., 2019. Complete genome sequence of an enterohemorrhagic Escherichia
Antigen 43, mediates cell-to-cell interactions within Escherichia coli biofilms. Mol. coli O111:H8 strain recovered from a large outbreak in Japan associated with
Microbiol. 37, 424–432. consumption of raw beef. Microbiol Resour Announc 8.
Darling, A.C., Mau, B., Blattner, F.R., Perna, N.T., 2004. Mauve: multiple alignment of Serra, D.O., Richter, A.M., Hengge, R., 2013a. Cellulose as an architectural element in
conserved genomic sequence with rearrangements. Genome Res. 14, 1394–1403. spatially structured Escherichia coli biofilms. J. Bacteriol. 195, 5540–5554.
Douglass, A.P., O’Brien, C.E., Offei, B., Coughlan, A.Y., Ortiz-Merino, R.A., Butler, G., Serra, D.O., Richter, A.M., Klauck, G., Mika, F., Hengge, R., 2013b. Microanatomy at
Byrne, K.P., Wolfe, K.H., 2019. Coverage-versus-length plots, a simple quality cellular resolution and spatial order of physiological differentiation in a bacterial
Control step for de Novo yeast genome sequence assemblies. Genetics 9, 879–887. biofilm. mBio 4, e00103, 00113.
Dourou, D., Beauchamp, C.S., Yoon, Y., Geornaras, I., Belk, K.E., Smith, G.C., Nychas, G.- Sherlock, O., Vejborg, R.M., Klemm, P., 2005. The TibA adhesin/invasin from
J.E., Sofos, J.N., 2011. Attachment and biofilm formation by Escherichia coli O157: enterotoxigenic Escherichia coli is self recognizing and induces bacterial aggregation
H7 at different temperatures, on various food-contact surfaces encountered in beef and biofilm formation. Infect. Immun. 73, 1954–1963.
processing. Int. J. Food Microbiol. 149, 262–268. Simm, R., Morr, M., Kader, A., Nimtz, M., Römling, U., 2004. GGDEF and EAL domains
Dueholm, M.S., Albertsen, M., Otzen, D., Nielsen, P.H., 2012. Curli functional amyloid inversely regulate cyclic di-GMP levels and transition from sessility to motility. Mol.
systems are phylogenetically widespread and display large diversity in operon and Microbiol. 53, 1123–1134.
protein structure. PloS One 7, e51274. Sinde, E., Carballo, J., 2000. Attachment of Salmonella spp. and Listeria monocytogenes to
García, B., Latasa, C., Solano, C., Portillo, F.G.-d., Gamazo, C., Lasa, I., 2004. Role of the stainless steel, rubber and polytetrafluorethylene: the influence of free energy and
GGDEF protein family in Salmonella cellulose biosynthesis and biofilm formation. the effect of commercial sanitizers. Food Microbiol. 17, 439–447.
Mol. Microbiol. 54, 264–277. Smith, A.M., Bosco, K.J., Nicol, M.P., Kleynhans, J., McCulloch, M., Duze, S.T., Ismail, A.,
González Barrios, A.F., Zuo, R., Hashimoto, Y., Yang, L., Bentley, W.E., Wood, T.K., 2006. Allam, M., Tau, N.P., Keddy, K.H., 2017. Genome sequence for Shiga Toxin-
Autoinducer 2 controls biofilm formation in Escherichia coli through a novel motility producing Escherichia coli O26:H11, associated with a cluster of hemolytic-uremic
quorum-sensing regulator (MqsR, B3022). J. Bacteriol. 188, 305–316. syndrome cases in South Africa, 2017. Genome Announc. 5.
Gurevich, A., Saveliev, V., Vyahhi, N., Tesler, G., 2013. QUAST: quality assessment tool Stepanovic, S., Vukovic, D., Hola, V., Bonaventura, G.d., Djukic, S., Cirkovic, I.,
for genome assemblies. Bioinformatics 29, 1072–1075. Ruzicka, F., 2007. Quantification of biofilm in microtiter plates: overview of testing
Habimana, O., Heir, E., Langsrud, S., Asli, A.W., Moretro, T., 2010. Enhanced surface conditions and practical recommendations for assessment of biofilm production by
colonization by Escherichia coli O157:H7 in biofilms formed by an Acinetobacter staphylococci. APMIS 115, 891–899.
calcoaceticus isolate from meat-processing environments. Appl. Environ. Microbiol. Taghadosi, R., Shakibaie, M.R., Ghanbarpour, R., Hosseini-Nave, H., 2017. Role of
76, 4557–4559. antigen-43 on biofilm formation and horizontal antibiotic resistance gene transfer in
Hadler, J.L., Clogher, P., Hurd, S., Phan, Q., Mandour, M., Bemis, K., Marcus, R., 2011. non-O157 Shiga toxin producing Escherichia coli strains. Iran. J. Microbiol. 9, 89–96.
Ten-year trends and risk factors for non-O157 Shiga toxin–producing Escherichia coli Uhlich, G.A., Chen, C.-Y., Cottrell, B.J., Nguyen, L.-H., 2014. Growth media and
found through Shiga toxin testing, Connecticut, 2000–2009. Clin. Infect. Dis. 53, temperature effects on biofilm formation by serotype O157:H7 and non-O157 Shiga
269–276. toxin-producing Escherichia coli. FEMS Microbiol. Lett. 354, 133–141.
Hammer, B.K., Bassler, B.L., 2003. Quorum sensing controls biofilm formation in Vibrio Uhlich, G.A., Chen, C.Y., Cottrell, B.J., Hofmann, C.S., Dudley, E.G., Strobaugh, T.P.,
cholerae. Mol. Microbiol. 50, 101–104. Nguyen, L.H., 2013. Phage insertion in mlrA and variations in rpoS limit curli
Hyatt, D., Chen, G.-L., LoCascio, P.F., Land, M.L., Larimer, F.W., Hauser, L.J., 2010. expression and biofilm formation in Escherichia coli serotype O157: H7. Microbiology
Prodigal: prokaryotic gene recognition and translation initiation site identification. 159, 1586–1596.
BMC Bioinf. 11, 119.

10
Y. Fang et al. Food Microbiology 102 (2022) 103902

Van Houdt, R., Aertsen, A., Moons, P., Vanoirbeek, K., Michiels, C.W., 2006. N-acyl-l- Typhimurium enhanced bacterial resistance to sanitization due to extracellular
homoserine lactone signal interception by Escherichia coli. FEMS (Fed. Eur. polymeric substances. J. Food Protect. 76, 1513–1522.
Microbiol. Soc.) Microbiol. Lett. 256, 83–89. Wang, X., Preston 3rd, J.F., Romeo, T., 2004. The pgaABCD locus of Escherichia coli
Van Houdt, R., Chris, W.M., 2005. Role of bacterial cell surface structures in Escherichia promotes the synthesis of a polysaccharide adhesin required for biofilm formation.
coli biofilm formation. Res. Microbiol. 156, 626–633. J. Bacteriol. 186, 2724–2734.
Visvalingam, J., Ells, T.C., Yang, X., 2017. Impact of persistent and non-persistent Weber, H., Pesavento, C., Possling, A., Tischendorf, G., Hengge, R., 2006. Cyclic-di-GMP-
generic Escherichia coli and Salmonella sp. recovered from a beef packing plant on mediated signalling within the σS network of Escherichia coli. Mol. Microbiol. 62,
biofilm formation by E. coli O157. J. Appl. Microbiol. 123, 1512–1521. 1014–1034.
Visvalingam, J., Wang, H., Ells, T.C., Yang, X., 2019a. Facultative anaerobes shape Wells, T.J., Sherlock, O., Rivas, L., Mahajan, A., Beatson, S.A., Torpdahl, M., Webb, R.I.,
multispecies biofilms composed of meat processing surface bacteria and Escherichia Allsopp, L.P., Gobius, K.S., Gally, D.L., Schembri, M.A., 2008. EhaA is a novel
coli O157:H7 or Salmonella enterica serovar typhimurium. Appl. Environ. Microbiol. autotransporter protein of enterohemorrhagic Escherichia coli O157:H7 that
85, e01123, 01119. contributes to adhesion and biofilm formation. Environ. Microbiol. 10, 589–604.
Visvalingam, J., Zhang, P., Ells, T.C., Yang, X., 2019b. Dynamics of biofilm Formation by Xavier, K.B., Bassler, B.L., 2005. Regulation of uptake and processing of the quorum-
Salmonella Typhimurium and beef processing plant bacteria in mono- and dual- sensing autoinducer AI-2 in Escherichia coli. J. Bacteriol. 187, 238–248.
species cultures. Microb. Ecol. 78, 375–387. Yang, X., He, A., Badoni, M., Tran, F., Wang, H., 2017a. Mapping sources of
Walters, M., Sperandio, V., 2006. Quorum sensing in Escherichia coli and Salmonella. Int. contamination of Escherichia coli on beef in the fabrication facility of a commercial
J. Med. Microbiol. 296, 125–131. beef packing plant. Food Contr. 75, 153–159.
Wang, H., He, A., Yang, X., 2018. Dynamics of microflora on conveyor belts in a beef Yang, X., Tran, F., Youssef, M.K., Gill, C.O., 2015. Determination of sources of Escherichia
fabrication facility during sanitation. Food Contr. 85, 42–47. coli on beef by multiple-locus variable-number tandem repeat analysis. J. Food
Wang, R., Bono, J.L., Kalchayanand, N., Shackelford, S., Harhay, D.M., 2012. Biofilm Protect. 78, 1296–1302.
formation by Shiga toxin-producing Escherichia coli O157:H7 and Non-O157 strains Yang, X., Wang, H., He, A., Tran, F., 2017b. Microbial efficacy and impact on the
and their tolerance to sanitizers commonly used in the food processing environment. population of Escherichia coli of a routine sanitation process for the fabrication
J. Food Protect. 75, 1418–1428. facility of a beef packing plant. Food Contr. 71, 353–357.
Wang, R., Kalchayanand, N., King, D.A., Luedtke, B.E., Bosilevac, J.M., Arthur, T.M., Yuan, L., Hansen, M.F., Roder, H.L., Wang, N., Burmolle, M., He, G., 2020. Mixed-species
2014. Biofilm formation and sanitizer resistance of Escherichia coli O157:H7 strains biofilms in the food industry: current knowledge and novel control strategies. Crit.
isolated from “high event period” meat contamination. J. Food Protect. 77, Rev. Food Sci. Nutr. 60, 2277–2293.
1982–1987. Zogaj, X., Nimtz, M., Rohde, M., Bokranz, W., Römling, U., 2001. The multicellular
Wang, R., Kalchayanand, N., Schmidt, J.W., Harhay, D.M., 2013. Mixed biofilm morphotypes of Salmonella typhimurium and Escherichia coli produce cellulose as the
formation by Shiga toxin–producing Escherichia coli and Salmonella enterica serovar second component of the extracellular matrix. Mol. Microbiol. 39, 1452–1463.

11

You might also like