Sie sind auf Seite 1von 16

Journal of Agriculture and Food Research 2 (2020) 100045

Contents lists available at ScienceDirect

Journal of Agriculture and Food Research


journal homepage: www.journals.elsevier.com/journal-of-agriculture-and-food-research/

Quantitative microbial risk assessment for Salmonella: Inclusion of whole


genome sequencing and genomic epidemiological studies, and advances in
the bioinformatics pipeline
Jinyao Chen a, c, Shraddha Karanth a, Abani K. Pradhan a, b, *
a
Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
b
Center for Food Safety and Security Systems, University of Maryland, College Park, MD, 20742, USA
c
West China School of Public Health, Sichuan University, Chengdu, Sichuan, China

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

Keywords: Despite the implementation of extensive measures to control food-borne illness caused by Salmonella since the
Salmonella beginning of the 20th century, it remains one of the most important and widespread food-borne pathogens.
Quantitative microbial risk assessment Quantitative microbial risk assessment (QMRA) has become essential to evaluating and mitigating the public
Whole genome sequencing
health risk of Salmonella. However, current QMRA models do not incorporate the minute changes in microbial
Food safety
Genomic epidemiology
behavior arising from differences in the microbial genome. Genomic analysis, particularly whole genome
sequencing (WGS), has undergone significant progress over the past two decades, facilitating rapid data accu-
mulation. This presents an unprecedented opportunity to incorporate genomic data into a QMRA for Salmonella.
This review aims to explore the applicability of WGS and other genomic technologies in Salmonella QMRA, and to
outline the added value and current limitations and promise of genomics, particularly bacterial WGS, in sup-
porting the integration of this data in a framework to assess the risk of Salmonella. Our review indicated that
although comparative genomic studies for Salmonella using WGS has seen a considerable amount of interest over
the past decade, the incorporation of genomic data into QMRA remains a difficult and arduous prospect. However,
with the progress in bioinformatics and genomic epidemiology, genomic data could certainly be utilized in QMRA
to improve and update risk assessment models and risk management strategies for Salmonella, as well as other
foodborne pathogens of interest.

1. Salmonella variance: Food safety implications several heterogeneous environments; consumable animal sources, spe-
cifically chicken and eggs, have been reported as major sources of human
The implementation of several preventative and control measures Salmonella infection [5–7]. However, recent outbreak data have also
against Salmonella since the beginning of the 20th century has failed to identified vegetables, nuts, and fruits, such as melons and papayas, as
make a significant impact on its worldwide prevalence rates. Salmonella possible novel routes for human infection [5,8,9].
is a major food-borne pathogen, with high morbidity and mortality rates The genus Salmonella is composed of two distinct species: S. bongori
and demonstrated major economic loss worldwide [1–3]. According to and S. enterica, the latter of which is divided into six subspecies. These
the World Health Organization (WHO), as of 2015, non-typhoidal Sal- subspecies have been classified into more than 50 serogroups and 2500
monella is considered to be 1 of the 4 key global causes of diarrheal serovars based on the O (somatic) and H (flagellar) antigens [10].
diseases, and affects an estimated 550 million people worldwide every Although different serotypes do not actually imply pathogenicity, a
year, including 220 million children under the age of 5 years. This report limited number of serotypes have been associated with a majority of the
estimated the global impact of Salmonella to be approximately 31–211 cases of human infections. Among these, Salmonella enterica serovar
million illnesses and 36,341–89,045 deaths, with a disease burden of Enteritidis (referred to hereafter as S. Enteritidis) is the most frequently
2.4–6.2 million disability-adjusted life years (DALYs) [4]. Globally, Sal- detected serovar, with incidence rates of 39.5% and 14.5% in the EU (in
monella prevalence and survival has been identified and reported in 2013) and the U.S. (in 2012), respectively, followed by Salmonella

* Corresponding author. Department of Nutrition and Food Science, University of Maryland, 0112 Skinner Building College Park, MD, 20742, USA.
E-mail address: akp@umd.edu (A.K. Pradhan).

https://doi.org/10.1016/j.jafr.2020.100045
Received 30 December 2019; Received in revised form 23 March 2020; Accepted 23 April 2020
2666-1543/© 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

enterica serovar Typhimurium (referred to as S. Typhimurium hereafter) and 2 (T3SS-1 and T3SS-2) are encoded within SPI-1 and SPI-2, respec-
(including its monophasic variant; incidence rates of 28.8% and 11.6% in tively, and it has been proven that deletion of the SPI-1 or SPI-2 regions
the EU and U.S. in 2013 and 2012, respectively) [11,12]. S. Enteritidis is can abrogate the virulence and ability of Salmonella to invade, colonize,
commonly associated with poultry and its products, whereas S. Typhi- or replicate within the host cells [30,33]. On the other hand, SPI-3 en-
murium has been isolated from a wider range of species, including pigs codes mgtC, which enables Salmonella survival under Mg2þ starvation
and cattle, in addition to poultry [13]. However, recently, several Sal- conditions and is therefore required for survival within macrophages, as
monella serovars other than the two considered to be major players in the well as to induce systemic infection in mouse cells [24,30]. SPI-4 encodes
food safety domain (S. Typhimurium and Enteritidis) have been more a type I secretion system (T1SS) and mediates adhesion, whereas SPI-5
frequently implicated in food-borne outbreaks. For example, an unusual includes SopB, which encodes the phosphatase portion of T3SS-1 [34].
serovar of Salmonella (ultimately recognized and named as S. Stanley) While little information is available about the other SPI, studies have
was identified as the causative organism of a cross-border outbreak shown that SPI-7 is exclusively present in the host-specific S. Typhi and
(comprising 10 European Union countries) with over 700 non-travel absent in S. Typhimurium [35], and SPI-8, -9, and -10 encode a pseudo
related human cases that occurred between 2011–2012 [13]. Similarly, bacteriocin and degenerate integrase, a type I secretion system like SPI-4,
there has been an increase in the incidence of multi-state outbreaks and a chaperone usher fimbrial operon, respectively [30,35,36].
linked to S. enterica serovars Montevideo, Infantis, Javiana, Braenderup, Salmonella attachment to the target cell occurs over several steps
Adelaide, and Heidelberg, and other unusual serovars in the U.S [14–17]. mediated by various adhesins encoded by fimbrial genes, such as type 1
fimbriae (fim), plasmid encoded fimbriae (pef), long polar fimbriae (lpf),
2. Diversity in Salmonella virulence and pathogenicity profiles or thin aggregative fimbriae (Agf). Virulence plasmids, which contain the
7.8-kb Salmonella plasmid virulence (spv) locus, also play a significant
All serovars belonging to Salmonella enterica share a common genome role in the virulence of Salmonella. However, virulence plasmids show a
structure, and the same core genome [10,18,19]. For instance, the remarkable diversity in the combination of virulence factors they encode,
serovars Typhi and Typhimurium have 11% difference in their genome, which appears to support their host specificity and ability to cause
which are otherwise 99% similar in their sequences of housekeeping gastroenteritis or systemic disease [37,38].
genes [10,15]. However, there is enormous variation in the virulence, It is common knowledge that effector proteins encoded by a complex
host range, and epidemiology of different serotypes of Salmonella. For network of functional virulence genes allow Salmonella to survive under
example, only specific serotypes (S. Typhi, S. Paratyphi A and C, and S. various conditions effected by immune cells such as macrophages, neu-
Sendai) cause enteric fever, while most serotypes cause only gastroen- trophils, and dendritic cells. Specific virulence determinants may be
teritis. Similarly, a few other non-typhoidal serovars such as S. Choler- clustered together on polymorphic pathogenicity islands or located on
aesuis and S. Dublin are more likely to cause bacteremia than diarrhea transmissible genetic elements such as plasmids or phages that facilitate
[20,21]. Furthermore, some serovars are host adaptive while others are the transmission of genes involved in pathogenesis, thereby increasing
more host-specific [22,23]. the diversity in virulence profile among strains [30,39,40]. Based on
Salmonella pathogenicity islands (SPIs) are DNA segments encoding recent findings that multiple individual virulence genes have a variable
virulence genes that are absent from the corresponding region of the distribution in Salmonella, it would be safe to conclude that a complicated
Escherichia coli K12 genome sequence. These gene-clusters are thought to combination of genes contribute to the overall virulence diversity, which
have been acquired by Salmonella from other species through horizontal presents a big challenge for virulence profiling and, by extension, for risk
gene transfer [24]. The presence of virulence determinants such as type assessment.
III secretion systems (T3SS) allows Salmonella to be intracellular patho-
gens, distinguishing them from commensal E. coli [10,24]. Salmonella 3. Whole genome sequencing (WGS) in microbiology:
undertakes several evolutionary measures, including gene acquisition Advantages and limitations in food safety
through horizontal transfer (associated with SPIs, transposable elements,
phages, and plasmids) and the loss of genes or a loss of their function, Laboratory identification and differentiation of bacterial infectious
which impacts host range. The latter might be the case with typhoidal agents are critical for timely detection of foodborne outbreaks, identifi-
serovars: comparative molecular analyses have suggested that the ge- cation of the transmission vehicles, and retraction of contaminated food
nomes of the human-specific serovars Typhi and Paratyphi A show from circulation [41]. Investigations into a foodborne outbreak relies on
considerable genome degradation and the accumulation of pseudogenes, the ability to subtype the etiological agent to differentiate it from cases
while being characterized by a unique complement of virulence de- caused by non-related agents. Moreover, possible co-circulation of several
terminants, including the type IVb pilus operon [25,26]. strains of foodborne pathogens among the population could confound
So-called virulence factors determine the virulence of bacterial epidemiologic investigations [42]. In this regard, the emerging WGS-based
strains. Actually, terms such as gene virulence and virulence factors are techniques, which provide complete data to enable the elucidation of
not strictly defined [20,27,28]. In the case of Salmonella, the list of phylogenetic relationships and monitoring of disease-causing lineages
candidate virulence factors increases with the ongoing increase in over time, offer a resolution and evolutionary context far exceeding that of
knowledge on the molecular mechanisms underlying its pathogenicity. traditional phenotypic molecular subtyping methods [43].
Virulence factors for Salmonella now include virulence plasmids, which WGS bypasses the need for a genetic map by using bacterial clones to
are genes encoding toxins, fimbriae, flagella, antimicrobial resistance, produce a large amount of redundant sequence read data across the
and other factors essential for pathogenicity [24,27,29]. So far, 21 SPIs genome and utilizing computational technology to assemble the
have been identified for Salmonella, and the continuous process of evo- sequence reads; subsequently, WGS has become the most popular and
lution may add more to this list. advanced sequencing method in the last decade [44,45]. More recently,
SPI-1~SPI-5 are classic pathogenicity islands that are commonly high-throughput next-generation sequencing technology has paved the
shared by a majority of the serovars of Salmonella. SPI-1 contains a way for cost-efficient and rapid WGS which can be used for real-time
number of functional genes, and is therefore responsible for the cyto- detection [46]. Before the maturation of WGS, different DNA profiling
toxicity of macrophages, invasion of epithelial cells, inflammation and techniques such as multiple-locus variable number of tandem repeat
fluid secretion in the ileum, and cytokine secretion, thereby playing a analysis (MLVA), pulsed-field gel electrophoresis (PFGE), and multi-locus
pivotal role in both gastroenteritis and systemic infection [30,31]. Genes sequence typing (MLST) were found to have drawbacks such as diffi-
within the SPI-2 region are not essential for gastroenteritis but are culties in standardization and comparatively lower resolution [47,48].
indispensable for systemic infection, as they support the intracellular Moreover, WGS could greatly improve the speed, cost, analytical spec-
survival of bacteria within host cells [32]. Type-III secretion systems 1 trum, and resolution of obtained data [49,50]. A study comparing the

2
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

capability of various methods used to subtype S. Enteritidis revealed that purposes has shifted to post-sequencing bioinformatics analysis of WGS
WGS resolved all outbreak clusters and provided robust inference results data. The failure to properly harmonize and interpret available WGS data
with higher epidemiological correlation compared to PFGE and MLVA; will lead to data fragmentation to the point that phenotypic traits such as
moreover, WGS was shown to cluster regularly interspaced short palin- strain pathogenicity may be regarded as being different for different
dromic repeats in combination with multi-virulence-locus sequence strains due to the use of distinct approaches of sequence assembly and
typing (CRISPR-MVLST) [41], showing the high applicability of this annotation [12]. Moreover, the WGS revolution has resulted in the
method in molecular subtyping of complex gene sequences. identification of a large number of genes with unknown function to date;
With the realization of WGS in microbiology, the robust and higher therefore, gene annotation and functional evaluation should be con-
resolution genomic data provided has yielded important insights into the ducted concurrently with sequencing. Another factor to be considered in
transmission pathways of several significant pathogens including Sal- the post-sequencing bioinformatics process is that WGS and mathemat-
monella. For example, WGS has been utilized in several recent epidemi- ical modeling appear to show opposing tendencies, one towards greater
ological investigations for microbial source attribution, in detail and better distinctions and the other towards simplicity and gen-
microbiologically linking current outbreaks with historic cases of food- erality [63].
borne illness, in differentiating outbreak cases with sporadic cases of Several public repositories of sequencing data for Salmonella are
illness, and most recently, in geographic source attribution [42,51–59]. currently available, with published sequences being made available for
However, the challenge of converting primary sequence data into useful comparative genomic analysis. Table 1 lists several databases and ini-
public health action, in the form of microbial risk assessments and pre- tiatives for Salmonella genomic analyses, including the National Center
diction of antimicrobial resistance patterns, remains primarily unmet. for Biotechnology Information's (NCBI) GenBank database, the U.S. FDA
This could be due to the lack of a comprehensive bioinformatics pipeline GenomeTrakr network, the NCBI Pathogen Detection database, the
and information technology (IT) support to analyze the data and obtain Collaborative Management Platform for Detection and Analyses of (Re)-
the specific information required for modeling and bacterial feature Emerging and Foodborne Outbreaks in Europe (COMPARE), and the
prediction [60–62]. Global Microbial Identifier (GMI), among others.
Additionally, there are a number of online resources for species-
4. Databases and initiatives for Salmonella genomic analyses specific typing and subtyping, such as the Salmonella In-Silico Typing
Resource (SISTR; https://lfz.corefacility.ca/sistr-app/), the direct iden-
Currently, the bottleneck in implementing WGS for public health tification of specific pathogenic genes from assembled genomes, as the

Table 1
Several databases and initiatives for Salmonella whole genomic analyses.
Database Website Contributors/organizers/initiators Purpose

National Center for Biotechnology https://www.ncbi.nlm.nih United States Food and Drug Administration's (U.S. Lists over 10,935 complete (and annotated) genome
Information's (NCBI) GenBank .gov/genome/genomes/152 FDA) Center for Food Safety and Applied Nutrition assemblies of Salmonella uploaded by a worldwide
database (CFSAN), the United States Department of network of labs, programs, and initiatives that are
Agriculture (USDA) Food Safety Inspection Service responsible for the generation of WGS data (and its
(FSIS), the U.S. Centers for Disease Control and related metadata).
Prevention (U.S. CDC), the CDC's National
Antimicrobial Resistance Monitoring System for
Enteric Bacteria (NARMS), the European Molecular
Biology Laboratory (EMBL), and the DNA Data
Bank of Japan (DDBJ), among others.
GenomeTrakr https://www.fda.gov/f U.S. FDA The first open-source network of its kind that
ood/whole-genome-seq utilizes WGS for pathogen identification. As of
uencing-wgs-program/g December 2019, GenomeTrakr participants include
enometrakr-network 15 federal laboratories, 25 state public health and
academic laboratories, 1 U.S. hospital lab, 2 other
labs located in the U.S., and 21 laboratories outside
the U.S., and several other laboratories, which are
authorized to collect WGS data and metadata of
foodborne pathogens, subsequently sharing them
via publicly accessible databases at the NCBI.
Collaborative Management Platform http://www.compare European Union's Horizon 2020 A large EU project comprising a multidisciplinary
for Detection and Analyses of (Re-) -europe.eu/ research network of 29 European participants from
Emerging and Foodborne Outbreaks 10 EU countries and Australia, with the aim of
in Europe (COMPARE) speeding up the detection of, and response to,
disease outbreaks among humans and animals
worldwide through the use of new genome
technology.
Global Microbial Identifier (GMI) http://www.globalmicr EU pilot program A network of approximately 160 representatives
obialidentifier.org/About from 32 countries working to develop a global
-GMI system to aggregate, share, mine, and use
microbiological genomic data to address global
public health and clinical challenges.
Pathosystems Resource Integration https://www.patricbrc. Wholly or in part funded by the U.S. National A genomics-centric relational database and source
Center (PATRIC) org/portal/portal/pat Institute of Allergy and Infectious Diseases, U.S. for the analysis of bacterial gene expression data.
ric/Home National Institutes of Health, and the Department
of Health and Human Service
Center for Genomic Epidemiology http://www.genom National Food Institute, Denmark Aims to provide the scientific foundation for a
icepidemiology.org/ future central database of genome data, a platform
of spatio-temporal tools for the analyses of such data
along with epidemiological information, as well as a
web-based interface capable of facilitating the
exchange of required microbial data.

3
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Antimicrobial Resistance Identification by Assembly (ARIBA; https://g information, comparative genomic research could also support in-
ithub.com/sanger-pathogens/ariba), or the identification of gene fam- vestigations into the molecular differences between isolates from
ilies that correlate with pathogenic phenotypes to distinguish them from different hosts, types of food, Salmonella serovars, and geographical and
relatively harmless microorganisms, such as the PathogenFinder (htt spatial locations, as well as the differences between serovars that could
ps://cge.cbs.dtu.dk/services/PathogenFinder/) [64]. However, many of lead to the manifestation of different types of disease. This could also aid
these databases and tools do not contain sufficient metadata regarding in the understanding of virulence genes and their combinations. To date,
the date, time, or conditions under which the isolates were obtained to comparative genomic analyses have been performed on various groups of
make useful conclusions about the behavior of pathogens in food or in isolates of Salmonella, with the corresponding phenotypic traits being
their human hosts. assessed by comparative phenotypic or proteomic studies [15,21,71–73].
There are, however, publicly available databases to look into the Therefore, gaining a thorough understanding of virulence genes and gene
virulence genes of Salmonella, and their functionality. The virulence profiles has been employed as the first step towards integrating Salmo-
factor database (VFDB) (http://www.mgc.ac.cn/VFs/) is a constantly nella WGS data with food safety risk assessment. With this in mind,
updated database of various virulence factors (and their corresponding WGS-assisted comparative genomic studies published alongside (or not)
genetic sequences) present in pathogenic bacteria, including E. coli, phenotypic analyses or proteomic studies of Salmonella were collected
Salmonella, and Vibrio [65]. Pathogenicity Island Databases (PAIDB) is a and listed in Table 2; the studies were retrieved using various combi-
database dedicated to providing comprehensive information on all an- nations of the keywords “whole genome sequence”, “comparative
notated and predicted pathogenicity islands (PAIs) in prokaryotic ge- genomic study”, “virulence phenotype”, “proteomic analysis”, and “Sal-
nomes, containing 223 types of PAIs with 1331 accessions [66]. monella.” Antimicrobial resistance genes were not considered at this
Alternatively, the “Virulence Search” database (http://www.hp point.
a-bioinfotools.org.uk/pise/virfactfind_small.html), developed by Public Current efforts in the area of Salmonella genomics involve the use of
Health England, is an engine that was designed to predict the potential WGS, followed by comparative genome analysis, in elucidating the
virulence factors in unannotated genomes [67]. Most of the above evolutionary relationships between bacterial species. However, as the
pathogen and pathogenicity prediction tools rely heavily on a structured evolution and phylogeny of the various Salmonella serovars was not the
database of pathogens to inform their pathogenicity inference. This primary focus of this literature review, only studies that investigated the
severely hinders their performance on unknown, unrecognized, and un- genetic variations in virulence determinants were included. Among the
mapped sequences [68]. Recent approaches to analyzing and interpreting studies included in Table 2, nine focused on genomic variety and
WGS data has shifted towards the inference of pathogenic phenotypes phenotypic diversity, six of which reported on a phenotypic analysis
using a number of machine learning- and deep-learning-based tools. coupled with WGS. All nine studies indicated the presence of consider-
Examples include the Pathogenicity Prediction for Bacterial Genomes able genomic variation within the Salmonella genus; in fact, some of these
(PaPrBaG; https://github.com/crarlus/paprbag) [69], which trains on a studies confirmed that Salmonella Typhi and Paratyphi A, serovars
wide range of species with known pathogenicity phenotype to overcome restricted to human hosts, characteristically displayed genome degra-
genetic divergence, and the Deep Learning Approach to Pathogenicity dation and pseudogene accumulation, as well as acquisition of horizon-
Classification (DeePAC; https://gitlab.com/rki_bioinformatics/DeePaC) tally transferred gene elements [10,15]. Most studies concluded that
[68], which infers pathogenic phenotypes without the aid of databases of horizontal gene transfer occurs frequently among all Salmonella strains,
known pathogens. and that the composition of mobile genetic elements with virulent plas-
The status of the above databases and programs suggests that despite mids and phages varies within and across serovars (inter- or
the rapid development of WGS and basic comparative genome analyses, intra-serovar) [10,14,15,20,21,71,72]. Two of the six studies that con-
there is a significant lag in the interpretation of raw data in a phenotypic ducted phenotypic analyses conducted mutant or knock-out tests for the
context. Moreover, there are no standard procedures or experimental genes of interest. While one established the hmpA and katE genes as being
protocols for the exploration of phenotypic characteristics. While some responsible for the loss of fitness under nitrosative and oxidative stress in
components necessary for phenotypic prediction and comparison based S. Typhimurium, which might be associated with host adaptation [15],
on WGS data have been developed, there currently exists no single in- the other determined that the gene st313-td contributed to the intracel-
tegrated platform to provide this kind of analysis. Moreover, there is no lular survival of S. Typhimurium in murine macrophages, and that its
specific repository of widely applicable genomic and phenotypic analyses deletion would attenuate the virulence of the isolates [73]. Among
for Salmonella, although a predictive genomic platform for E. coli studies that focused on the phenotypic and genotypic variations among
(https://lfz.corefacility.ca/superphy/) is under development [70]. various Salmonella strains, three focused on metabolic traits such as the
Comparative genomic analyses of Salmonella would be a crucial first step utilization of different carbon sources [72], β-glucuronidase activity
in the evolution of these platforms; this could help in establishing the [71], catalase activity, and growth patterns in the presence of tetrathi-
framework to pool genotypic and phenotypic information obtained by onate and under nitrosative stress [15]. Alternately, three other studies
Salmonella researchers worldwide. looked directly into the virulent characteristics of various Salmonella
serovars [19,21,73] by analyzing their ability to infect mice, via in vitro
5. Comparative genomic analysis of Salmonella tests [21,73], and using a test to determine virulence against Caeno-
rhabditis elegans [19]. Noteworthy studies include those conducted by
At this point, filtering genome sequences to locate genes relevant to Fricke et al. (2011) and den Bakker et al. (2011), wherein the phenotypic
its virulence is an essential step towards integrating molecular data into variations in utilization of carbon substrates between genetically similar
risk assessment. However, the physiological impact of the occurrence, Salmonella isolates and the effect of genes related to β-glucuronidase
expression, and interaction of such genes poses a major challenge. activity on host adaption, respectively, were observed [71,72]. These
Despite the availability of several tools (both old-school and emerging) studies implied that phenotype-based risk assessment research should
for genome sequence analyses, data mining for virulence genes and focus on the metabolic aspect of various Salmonella serovars in addition
predicting their observable phenotypes is still in its infancy. Although to their virulence characteristic. However, the above studies failed to
genetic analyses and comparisons are required, determining the pheno- directly link the genotype and/or phenotype geographically and spatially
typic characteristics (such as adhesion, invasiveness of intestinal and to outbreak and sporadic cases. This is despite specific genes being
epithelial cells, antimicrobial resistance, and animal virulence) encoded linked to increased virulence; examples of such genes include st313-td,
in these genetic sequences by performing functional (proteomic) or which is correlated with increased invasiveness of S. Typhimurium [73],
phenotypic assays in combination with gene expression analyses would and SPI-2, whose defection was linked to the incompetence of S. Ken-
be more preferable. In the face of a lack of experimental phenotypic tucky [14].

4
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 2
Comparative genomic studies of Salmonella by WGS.
Authors, year, Description of the isolates Phenotypic analysis Main findings of the study Virulence genes specifically focused
country on

1 Jacobsen et al. 45 sequenced Salmonella genomes N/A (1) The core and pan-genome of SPI-6 and SPI-7 found only in serovar
(2011) [10], that were publicly available at the Salmonella were estimated to S. Typhi; SPI-3 exists in several similar
Denmark time of the study. contain around 2,800 and 10,000 but distinct versions.
gene families, respectively.
(2) Gene content is often, but not
uniformly, correlated to serotype.
(3) The SPIs were among genes that
showed variations; variations were
also observed in phage insertion
sites and transposable elements.
2 Fricke et al. 17 non-typhoidal Salmonella strains The Biolog phenotype microarray (1) The evolution of known
(2011) [72], (classified into 12 serovars) isolated system, which allows the testing of Salmonella sublineages is mediated
U.S. from human, animal, and food nearly 1,200 metabolic and chemical mostly by the loss of coding
sources. sensitivity phenotypes, was used to sequences with known metabolic
assay the utilization of different functions and the acquisition of
carbon sources. horizontally transferred phage and
plasmid DNA.
(2) Phenotypic variations between
closely related Salmonella isolates
were observed for utilization of
carbon substrates.
(3) Phenotypes are frequently lost
and/or acquired in different
Salmonella sublineages and thus
correlate poorly with the
evolutionary relationships within
this species.
3 den Bakker MLSA and SNP analysis of 16 newly β-glucuronidase activity was (1) Salmonella consists of at least All clade B isolates contained SPI-18
et al. (2011) sequenced and 30 publicly available determined by growing the isolates two subpopulations (clade A and and a cytolethal distending toxin islet.
[71], U.S. genomes resulted in the division of in EC MUG broth. clade B) that differ specifically in The combination of these two islands
Salmonella into two clades, A and B. genes involved in host and tissue was previously thought to be
An additional set of 123 isolates was tropism, and utilization of host- exclusive to typhoid-associated
assigned to clades A and B by specific carbon and nitrogen serovars.
targeting subpopulation-specific sources, suggesting differences in
genes and SNPs with qPCR. adhesion, colonization properties,
and metabolic capabilities.
(2) Presence of β-glucuronidase in
clade B isolates suggests an
adaptation of this clade to the
vertebrate gastrointestinal
environment.
4 Hayden et al. 114 S. Typhimurium strains (1) Bacterial growth in the presence (1) A phylogenetic tree of S. Strains closely related to strains
(2016) [15], collected between 1946 and 2012 of tetrathionate and nitrosative Typhymurium was constructed. associated with human
U.S. from humans and food animals in stress. (2) In the U.S., salmonellosis is immunodeficiency virus (HIV)-
the U.S. and abroad. (2) Mutation of hmpA and katE was caused by diverse S. Typhimurium infected individuals in sub-Saharan
tested in strains that showed growth strains circulating worldwide. Africa were found. Phenotyping
under nitrosative and catalase (3) No specific link was found studies linked hmpA and katE variants
activity, respectively. between strains or lineages and in subclade 1a specific to these strains
specific outbreaks or sporadic to loss of fitness under nitrosative and
events. oxidative stress, respectively.
5 Herrero- 295 S. Typhimurium and 50 S. (1) Mouse infection; (2) epithelial The gene st313-td is mainly The apparently restricted presence of
Fresno et al. Dublin isolates from 10 countries cell infection; (3) macrophage associated with invasive strains, st313-td in human isolates suggests a
(2014) [73], were analyzed for the presence of infection; (4) knock-out and st313-td contributes to S. potential host specificity for this gene
Denmark st313-td. Six S. Typhimurium strains complementation study: the gene Typhimurium intracellular survival as suggested for its carrier, S.
from around the world were st313-td was cloned into wild type S. within murine macrophages, and Typhimurium ST313.
sequenced. Typhimurium as well as knocked out not needed for invasion of the
in S. Typhimurium ST313 followed epithelial cells, and st313-td knock-
by complementation. out was attenuated in virulence in a
systemic murine infection.
6 Dhanani et al. 25 Salmonella isolates classified into N/A (1) The study isolates showed Conserved T3SS and T4SS could be
(2015) [14], 5 serovars from broiler chicken variations in genes coding for candidates for pathogenicity
Canada commercial farms in Canada; some adhesins, flagella, iron acquisition, prediction.
of the isolates included in “Diarra T3SS, T4SS, and drug resistance, The lack of SPI-2-associated genes
et al. (2014) [85]”. suggesting the diversity of SopD2, PipB2, SspH2, and SrfH in
Salmonella serovars found in broiler isolates of S. Kentucky might explain
chicken. in part their inability to induce
(2) Existence of mobile genetic diseases in humans.
elements indicates that isolates
from a given serovar could acquire
and transfer genetic elements.
7 Blondel et al. 44 completed or in-progress N/A (1) The Salmonella T6SS loci were Data indicate that T6SS is widely
(2009) [111], Salmonella genomes from a public acquired by independent lateral distributed in some serovars (such as
Chile database. transfer events and evolved to S. Dublin) and absent in serovars
(continued on next page)

5
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 2 (continued )
Authors, year, Description of the isolates Phenotypic analysis Main findings of the study Virulence genes specifically focused
country on

Three representatives of each branch contribute to the adaptation of Enteritidis, Gallinarum, Agona,
in previously described T6SS various serotypes to different Javiana, and Paratyphi B, among
phylogenetic trees were used as lifestyles and environments, others.
baits. including animal hosts. However,
these were not essential for
virulence.
(2) The authors identified 3 novel
T6SS loci, designated as SPI-19,
SPI-20, and SPI-21.
8 Marzel et al. A case-control study of 48345 The 22 S. Typhimurium strains were (1) A different composition of
(2016) [21], culture-confirmed non-typhoidal tested for (1) growth in minimal mobile genetic elements (plasmids
Israel Salmonella infections that occurred media; (2) biofilm formation; (3) and phages) or SNPs in global
in Israel between 1995 and 2012. motility; (4) and replication within regulators was identified in
WGS data was used to analyze 22 S. macrophages. multiple cases.
Typhimurium isolates from 11 Four pairs of isolates that showed (2) Four NTS serovars (Mbandaka,
patients with persistent, recurring genetic and phenotypic differences in Bredeney, Infantis and Virchow)
cases of salmonellosis. vitro were chosen to perform a were significantly more frequently
streptomycin pre-treated mouse associated with persistence than
colitis model test. others in this study.
(3) Though clear directionality of
neither genetic nor phenotypic
changes between early and later
same-patient isolates were
identified, differences in phenotype
and virulence between early- and
late-stage same-patient isolates
were found, specifically in-host
microevolution and its potential to
shape Salmonella pathogenicity.
9 Hoffmann 44 S. Heidelberg isolates including Caenorhabditis elegans virulence (1) Substantial loss and gain of 107 previously characterized
et al. (2014) 30 with nearly indistinguishable assay. plasmids and phages between these virulence genes (84 pathogenicity and
[19], U.S. PFGE patterns from various sources S. Heidelberg isolates that were 23 fimbrial markers) and plasmids:
between 1982 and 2011. This study consistent with previously SPI-1~5, the lambda-like prophage
included nine isolates associated generated tree topologies, Gifsy-2, and/or islets, T3SS, sopE, and
with the 2011 S. Heidelberg suggesting rapid exchange of these adhesins were present in all S.
outbreak in the U.S. plasmids. Heidelberg isolates. On the other
(2) S. Heidelberg isolates carrying hand, SPI-7, prophage Gifsy-1, Gifsy-
components of the T4SS were more 3, Fels-1, and spv were not identified
pathogenic in C. elegans model than in any of the 44 isolates.
those not carrying the system,
indicating T4SS plays a role in the
pathogenicity of S. Heidelberg.

SPI – Salmonella pathogenicity islands.


NTS – non-typhoidal Salmonella.
PFGE – pulsed-field gel electrophoresis.
TxSS – Type-x secretion system (where x denotes the number; for example, T4SS).

Undoubtedly, the high discriminatory capacity of WGS has cemented corresponding phenotype, coding for the plasmid-borne pef operon and
its position as the new gold-standard method for comparative genomic amino acid transportation genes, which are responsible for nitrogen
studies of isolates. However, since a search of literature pertaining to utilization, epithelial cell invasion, and biofilm formation, between the
comparative genomic studies using WGS provided us with a limited prevalent PTs 4, 6, 8, and 13a and non-prevalent PT11 strains [74].
number of studies, we expanded our search to other comparative Therefore, these genes could be of considerable interest as targets in
genomic studies carried out by comparative genomic hybridization future studies. On the other hand, Betancor et al. (2009) [39] and Yim
(CGH; Table 3), as well as to studies that investigated multiple virulence et al. (2010) [76] reported that phage SE20 and phage-associated genes
factors and phenotypic traits using polymerase chain reaction (PCR; might contribute to the epidemiological nature of S. Enteriditis. In fact,
Table 4). Microarrays comprising multiple genes from the genome of a Betancor's study detected major differences in the in vitro infection ability
typical Salmonella serovar such as S. Typhimurium LT2 or S. Typhi CT18, with strains with indistinguishable genetic patterns being based on the
those combined with specific genes from other serovars, as well as Salmonella pan-genome [39], which reflects the considerable variability
microarrays comprising genes from the Salmonella pan-genome are in the Salmonella genotype and phenotype, and highlights the need for
available commercially [25,40,74]. However, the results of CGH and PCR high-resolution WGS as comparative tool for Salmonella identification
using these microarrays should be interpreted with caution as and characterization.
microarray-based analyses cannot detect mutated genes, or those that Studies conducted after 2000 using traditional PCR primarily focused
have not yet been identified by sequencing [75]. on specific virulence genes in Salmonella. However, as the analyzed iso-
Fourteen CGH studies focusing on virulence gene expression and lates were obtained from different sources present in the same
phenotype comparison and eleven studies analyzing multiple virulence geographic area, a majority of these studies could not identify specific
factors using PCR (since 2000) were included in this review. The results source-related or prevalence-related virulence profiles. Among these
of the CGH studies corroborated those of the WGS studies with respect to studies, phenotypic analyses were performed in only two studies: one
the variability in the virulence, potency, and survival characteristics of study conducted using a mouse lethality assay [77] reported a conflicting
Salmonella. Pan et al. (2009) found differences in the genes, and the finding that outbreak strains may be avirulent, while the other, which

6
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 3
Comparative genomic studies of Salmonella by Comparative genomic hybridization (CGH) with or without other methods.
Authors, Description of the isolates CGH used, and the Phenotypic analysis Main findings of the study Virulence genes specifically
year, country microarray description focused on

1 Betancor 266 S. Enteritidis isolates The 266 S. Enteritidis Human epithelial cell (1) 233 chromosomal genes Phages play a crucial role in
et al. (2009) from different sources. isolates were genotyped invasion analysis was coding for metabolism and the generation of genetic
[39], using RAPD-PCR. performed for the 29 bacterial fitness, and the diversity in S. Enteritidis.
Uruguay 29 epidemic-spanning S. isolates, spanning different virulence plasmid analyzed Phage SE20 may be a key
Enteritidis field isolates periods, genetic profiles, were found as variable among marker for the emergence of
were subjected to CGH and sources of isolation. S. Enteritidis isolates. particular isolates capable of
using a Salmonella pan-array (2) Strains with causing epidemics.
based on the S. Typhi CT18 indistinguishable genetic
genome, complemented patterns still showed major
with strain-specific genes differences in their infection
from other strains. ability; the approach used
was insufficient to detect the
genetic basis of differential
behavior.
2 Yim et al. The 29 isolates studied in The CGH design and results (1) Motility tests; (2) Caco- (1) A great heterogeneity of Loss of several phage-
(2010) [76], “Betancor et al. (2009) were the same as in 2 adhesion and invasion phenotypes was found, with associated genes improved
Uruguay [39]” 15 isolates from “Betancor et al. (2009) assays; (3) Bacterial isolates belonging to identical the virulence of S. Enteritidis,
human gastroenteritis, 5 [39]”. survival in egg albumen; genetic profiles showing very rendering strains more prone
from invasive disease, and (4) Chicken virulence dissimilar behaviors. to cause an epidemic.
9 from nonhuman sources assays. (2) The pre-epidemic isolates
in Uruguay. showed low levels of fitness in
most of the assays evaluated.
(3) The subset of isolates from
human gastroenteritis
showed higher homogeneity
of phenotypes and higher
levels of fitness than isolates
from any other sources.
3 Pan et al. 37 S. Enteritidis strains CGH with a Salmonella pan- (1) Epithelial cell adhesion Genetic and corresponding 400 genes that differentiated
(2009) [74], from five phage types genome microarray and invasion assays. phenotypic differences exist prevalent (4, 6, 8 and 13a)
UK (prevalent and non- covering seven Salmonella (2) Bio-forming assay and between strains of the and sporadic (11) PTs, of
prevalent) from different genomes accompanied with colony morphology. prevalent PTs 4, 6, 8 and 13a which 35 were unique to
sources in UK. Pulsed field gel (3) Phenotype microarray and non-prevalent PT11 prevalent PTs, including six
electrophoresis (PFGE) and for nitrogen utilization and strains: (1) strains from plasmid-borne genes and four
multi-locus sequence typing osmotic stress. prevalent PTs were less chromosomal genes encoding
(MLST). (4) Inactivation of amino susceptible to urea stress and putative amino acid
acid transporter genes and utilized amino acids more transporters.
complementation. efficiently than PT11 strains.
(2) in epithelial cell
association assays, PT11
strains were less invasive than
other prevalent PTs.
(3) Most strains from
prevalent PTs were better
biofilm formers than PT11.
4 Morales et al. A field isolate of S. CGH with a DNA microarray Phenotype microarrays It is not known how the A single band shift within a
(2005) Enteritidis PT4, a wild-type developed from sequence of assay the growth of results of phenotype phage type may indicate an
[112], U.S. S. Enteritidis PT13A strain, S. Enteritidis PT4 bacteria by measurement microarray analysis correlate evolutionary event in this
and a biofilm-forming S. accompanied with PCR. of respiratory activity with virulence genes pathogen that has potential
Enteritidis PT13A. under variable culture expression. for increasing human disease.
conditions.
5 Anjum et al. 40 clinical and field CGH with a DNA microarray Most genes within SPI-1 to
(2005) [18], Salmonella isolates from 12 developed based on S. Typhi SPI-5 and SPI-6, SPI-9, and
UK common serovars. CT18 chromosome. three fimbrial operons (fim,
bcf, and stb) were conserved
within all Salmonella strains
included in this study.
6 Porwollik 22 S. enterica and 1 CGH with a DNA microarray The scale of gene flux among Outer membrane proteins
et al. (2002) S. bongori. developed based on S. and within the various strains (Sops) is variable, and SPI-
[79], U.S. Typhimurium LT2 genome. is significant. 1~5 is quite homologues.
7 Porwollik 79 isolates from common A microarray based on S. Gene contents sometimes The entire plasmid of pSLT
et al. (2004) prevalent serovars Typhimurium LT2 differed more within a including spv is present in all
[80], U.S. supplemented with serovar than between S. Typhimurium strains
annotated chromosomal serovars. included in this study.
ORFs from the serovar
Typhi CT18, also containing
LT2 virulence plasmid pSLT
and R46
8 Figueiredo 106 Salmonella strains from A customized Salmonella (1) Cells distention/cells Isolates of the same serotype (1) Virulence determinants
et al. (2015) poultry (n ¼ 39), swine virulence gene microarray apoptosis analysis usually harbor the same located in SPIs 1–6 and
[25], UK (n ¼ 14), cattle (n ¼ 1), and targeting 114 Salmonella (2) epithelial cells virulence profile. adhesion were highly
processed food (meat cuts, virulence determinants adhesion/invasion assays. conserved; prophage genes
present on the chromosome (3) Galleria mellonella shows great variability; genes
(continued on next page)

7
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 3 (continued )
Authors, Description of the isolates CGH used, and the Phenotypic analysis Main findings of the study Virulence genes specifically
year, country microarray description focused on

hamburgers or sausages) (n ¼ 90; SPIs, n ¼ 52, and infection model, was in virulence plasmid are
(n ¼ 52). other chromosomal genes, detected for S. Infantis present in all S. Enteritidis
n ¼ 38), phage related Sal147, Sal199, Sal280, and part of S. Typhimurium.
(n ¼ 12), and plasmid and S. Typhimurium LT2. (2) S. Infantis Sal147, was
related (n ¼ 12). unable to invade HT-29 cells,
WGS for S. Infantis Sal147 unlike Sal280, virulence was
(absence of SPI-1 genes), decreased corresponding to
and S. Typhimurium Sal199 the absence of SPI-1 genes.
(presence of cdtB of toxin); (3) The increased virulence
PCR of SPI-11 genes and and cytotoxic effect by in vitro
typhoid-associated cells apoptosis analysis and in
virulence genes. vivo mortality of G. mellonella
by S. Typhimurium Sal199
with respect to S.
Typhimurium LT2 might be
associated with expression of
the CdtB toxin genes.
9 Zou et al. 24 Salmonella isolates CGH composing of 69 gene- 58% of the virulence- Regardless of the serotype,
(2011) [81], including Roma tomato specific oligonucleotide associated genes (40/69) Salmonella isolates exhibited
U.S. outbreak strains, and probes including (SPI-1 to were present in all 24 most variability among the
isolates from turkey farms SPI-5) virulence-associated Salmonella isolates tested iacP, avrA, sopE, sirA and invH
and chicken eggs. genes using S. Typhimurium while the remaining genes genes belonging to the SPI-1
LT2 genome NC_003197 as were variable in their class.
the reference. distribution.
10 Huehn et al. 523 Salmonella strains (S. PCR for 10 potential (1) The virulotype did not (1) Virulence determinants
(2010) [84], Enteritidis, Typhimurium, virulence factors (avrA, vary significantly with host located in the SPIs 1–5 were
Europe Infantis, Virchow, and ssaQ, mgtC, siiD, sopB, gipA, source or geographical highly conserved among the
Hadar) collected from sodC1, sopE1, spvC, and location. five serovars except for SPI-3,
various host sources and bcfC) for 523 isolates. (2) The highest variation was virulence genes in phage and
public health and A representative subset of observed in genes located on plasmid were variable.
veterinary institutes across 77 strains was investigated prophages. Each serovar (2) Some genes for
nine European countries. using a DNA microarray demonstrated its own specific pathogenicity, e.g. spv, were
designed to detect 102 fimbrial profile. only found in S. Enteritidis,
virulence and 49 resistance Typhimurium.
determinants.
11 Suez et al. Low-passage clinical strains CGH using a Salmonella (1) The pathogenicity of (1) A substantial degree of (1) SPI 1–5, 9, 13, 14; five
(2013) [83], isolated from blood microarray (the STv7E invasive isolates intra-serovar variation in fimbrial operons (bcf, csg, stb,
Israel (n ¼ 66), stool (n ¼ 68) or Salmonella microarray). representing the 12 virulence-associated sth, sti); three colonization
unknown source (N ¼ 1) serovars was investigated phenotypes was identified. factors (misL, bapA, sinH);
from 12 non-typhoidal in the mouse model. (2) Not all previous identified and the invasion gene, pagN
Salmonella (NTS) serovars (2) Virulence-associated virulence factors are were conserved in all isolates.
obtained from the Israeli phenotypes were necessary for invasiveness. (2) Except that tcfA that was
National Salmonella compared between (3) Intrigue results were found to be more frequently
reference Center. invasive and found that blood isolates were associated with invasive
gastroenteritis isolates in not generally more invasive isolates of S. Schwarzengrund
epithelial cells lines and than stool isolates of the same than with gastroenteritis
macrophages. serovar, and superior isolates, the study did not
intraepithelial cell replication identify a clear source-related
of gastroenteritis isolates distribution of virulence
compared to invasive isolates genes.
was observed in some (3) Various NTS virulence
serovars. factors and six typhoid-
associated virulence genes
displaying a wider
distribution among NTS than
was previously known.
12 Litrup et al. 58 Salmonella isolates from A DNA micro-array (1) Differences in content of Strains from the supposedly
(2010) [26], human faces and 10 from containing 280 probes pathogenicity related genes less virulent serovar S. Derby
Denmark human blood, and pigs covering genes related to between serovars with lacked a combination of
(n ¼ 3), cattle (n ¼ 1) and pathogenicity, phages, various host preferences and virulence genes (truncated
hedgehog (n ¼ 1). antimicrobial resistance, pathogenicity towards SPI-3, irsA, msgA, and some
fimbria, mobility, serovars, humans were not observed. prophage genes).
and metabolism with MLST. (2) Except for 4 outbreak
strains of S. 4,5,12:b: the
characterization of strains
based on pathogenicity
related genes fits well with
the serovar assignment.
13 Reen et al. 12 S. Dublin, Agona, and The S. Typhimurium LT2 Only limited intraserovar SPI1~SPI5 were present in
(2005) [86], Typhimurium isolates microarray, the same with diversity was found among all isolates. Plasmid pSLT was
Ireland recovered from bovine and “Porwollik et al., 2004”, the nine S. Dublin, three S. absent from all S. Agona
milk filter samples in including SPI1~SPI5 and Agona, and six S. isolates, and only
southern Ireland, and 2 pSLT. Typhimurium isolates. homologues of the spv genes
clinical isolates from were present in eight of the
nine S. Dublin strains, and
(continued on next page)

8
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 3 (continued )
Authors, Description of the isolates CGH used, and the Phenotypic analysis Main findings of the study Virulence genes specifically
year, country microarray description focused on

Canada and 4 archival present in the six S.


isolates. Typhimurium isolates.
14 Beutlich 38 Salmonella strains PCR (10 virulence genes: (1) Only minor serovar-
et al. (2011) isolated between 2002 and avrA, bcfC, gipA, mgtC, ssaQ, specific differences among
[113], 2006 in eight European sopB, sopC1, sopE1, spvC, and virulence patterns were
Europe countries from humans, spi4_D). detected.
animals, and food. 9 representative Salmonella (2) Some virulence patterns
isolates were analyzed by were serovar specific, e.g., all
DNA microarray (101 genes S. Newport and most S.
including SPIs, prophages, Typhimurium strains shared a
plasmids, islets, and fimbrial common virulotype.
clusters). (3) SGI1-positive isolates
carry more or less identical
virulence gene repertoires.

PFGE – Pulsed field gel electrophoresis.


MLST – Multi-locus sequence typing.
RAPD-PCR – Random amplification of polymorphic DNA polymerase chain reaction.
NTS – Non-typhoidal Salmonella.
SPI – Salmonella pathogenicity islands.

analyzed the production of aerobactin and colicin in various Salmonella limited to certain serotypes that are responsible for gastroenteritis.
serovars [78], reported a difference in the production of the siderophore Although a number of these studies give us an insight into the genetic
and bacteriocin between the Typhimurim and Kentucky serovars. factors controlling Salmonella virulence behavior, many of these studies
Several CGH studies focused on the construction of a Salmonella pan- are relatively low-throughput, as the genes chosen were identified based
genome, with the intention of locating the core and variable genes in the on earlier findings. Recent studies, on the other hand, are exploring a
genome, which are not very well defined [18,79,80]. For example, more comprehensive approach towards obtain a systems-level under-
although a majority of studies suggested that genes in the SPI-1~SPI-6 standing of bacterial physiology by analyzing the expression of proteins
region were conserved, Zou et al. (2011) reported significant variability related to bacterial pathobiology under stress conditions that mimic
among genes belonging to the SPI-1 class irrespective of the serotype in those experienced by bacteria during human colonization and infection
twenty-four Salmonella isolates [81]. This defect in the genes belonging (Table 5). A major area of interest has been to identify the regulatory
to the SPI-1 class was also reported by Figueiredo et al. (2015) and systems that allow bacterial cells to identify and overcome host envi-
Majtan et al. (2007) [25,82] Additionally, Figueiredo et al. (2015) re- ronmental and immune signals. For example, Tran et al. (2016) con-
ported that S. Infantis Sal147 displayed decreased invasiveness in the ducted a whole proteome analysis on wild-type and mutant S.
absence of the SPI-1 region [25]. Studies conducted with Salmonella Typhimurium strains to identify proteins regulated by, and the overall
serovars obtained from different geographical sources such as the U.S. importance of, the RstA/RstB and PhoPQ two-component regulatory
[71], UK [25], and Israel [83] suggested that the presence of systems (TCS) on bacterial pathogenesis, finding that while the PhoPQ
typhoid-associated virulence genes was not exclusive to typhoid-related regulatory system was more extensive than the RstA/RstB system (14.9%
serovars; in fact, these studies concluded that these genes displayed a vs. 6.92%), the latter played a more significant role in regulating Sal-
wider distribution among non-typhoidal Salmonella strains than was monella motility and invasion [87]. This was similar to the results found
previously known, which brought to the fore questions regarding the by Liu et al. (2015) [88]. On the other hand, Liu et al. (2017) and Karash
pathogenic mechanism of Salmonella as well as the predictability of et al. (2017) performed a proteogenomic analysis of S. Typhimurium
previous in vitro and even in vivo invasion assays. Correspondingly, under stress conditions (such as the production of reactive oxygen spe-
clinical trials conducted by Suez et al. (2013) obtained interesting results cies) initiated by macrophages, finding a significant downregulation in
about the phenotypic/epidemiological traits, such as the lower level of histidine biosynthesis in the former, and upregulation in proteins
invasiveness of the same serovar in blood isolates compared to stool responsible for DNA repair, aromatic amino acid biosynthesis, Fe–S
isolates and the greater fitness of gastroenteritis isolates compared to cluster biosynthesis, iron homeostasis and H2O2 scavenging in the latter
invasive isolates grouped into some serovars [83]. study [89,90]. Li et al. (2019), on the other hand, focused on changes in
The results of studies conducted by Zou et al. (2011) [81] using 24 Salmonella metabolic pathways during host infection as indicators of its
Salmonella strains from the U.S., Litrup et al. (2010) [26] with 73 isolates survival and proliferation capabilities. The authors observed significant
from Denmark, and Huehn et al. (2010) [84] using a larger sample of 523 upregulation of PhoPQ-regulated proteins, such as PgtE, PqaN, and
Salmonella strains obtained from across Europe indicated that strains PhoN, and metal transporters such as SitAB, but downregulation of Sal-
belonging to the same serovar could be grouped together via virulence monella flagellar proteins, such as fliC and fliFGLMNI, and chemotaxis
profiling. This indicated that the broader virulence-associated gene proteins such as Tsr, Tcp, Tar, and Trg, at 18 h post-infection of an in vitro
complemented the serovar classification. However, as these studies used culture [91]. These, and additional findings by the authors indicate that
microarrays that were only focused on known virulence factors, the invasive Salmonella employ a non-motile lifestyle to support rapid pro-
significance of the former statement and the extent of correlation be- liferation, and also display significant shifts in metabolism and respira-
tween the two should be further investigated using WGS. tion cycles to optimize on the available conditions within the host,
Mobile genetic elements such as plasmid genes have generally shown similar to the results obtained by Liu et al. (2015) and Noster et al.
greater variability in distribution among strains compared to phage genes. (2019). These studies provide a fairly detailed overview of the genotypic
Moreover, the results of several studies have shown that genes occurring in and phenotypic controllers of bacterial physiology, which in turn can
the plasmid (such as spv) are more frequently observed in the common help in fine-tuning the predictive models for bacterial dose-response [88,
Salmonella serovars such as Enteritidis and Typhimurium [80,84–86]. This 92].
conforms to the established idea that virulence plasmids are usually

9
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 4
Comparative multiple virulence factor studies of Salmonella by PCR.
Authors, Description of the isolates Genes detected by PCR Phenotypic analysis Main findings of the study Virulence genes specifically
year, focused on
country

1 Capuano 114 Salmonella strains of human PCR for mobile virulence 59 different virulence profiles The genes showing the
et al. (2013) origin (71/114) or isolated from genes (gipA, gtgB, sopE, sspH1, were highlighted. highest homology were
[22], Italy food (43/114) collected from sspH2, sodC1, gtgE, spvC, pefA, those related to the
2010 to 2012, including only the mig5, rck, srgA). presence of prophages
S. Enteritidis, S. Typhimurium, (gipA, gtgB, sopE, sspH1,
S. 4 [5],12:i: . sspH2, sodC1, gtgE), while
the genes related to the
presence of plasmids were
less frequently detected
(spvC, pefA, mig5, rck, srgA).
2 Jaradat et al. 18 S. Enteriditis isolates A phylogenic tree based on Isolates (1) The most virulent strains
(2014) [77], obtained from raw poultry 16S rRNA sequencing was representing each were isolated from poultry
North samples compared with 7 constructed to cluster the S. cluster were further samples and had an LD50 of
Jordan outbreak strains. Enteritidis isolates. studied for their 1.55  105 CFU.
virulence in normal (2) Some of the outbreak
adult Swiss mice. isolates were highly virulent
while others were avirulent,
which might be due to a
variation in susceptibility of the
mouse to different S. Enteritidis
isolates.
3 Campioni 128 S. Enteritidis strains isolated ERIC-PCR and PFGE for The strains isolated from Almost all strains (97.6%)
et al. (2012) from human feces (67) and food clustering. sporadic cases were harbored all 13 virulence
[114], Brazil (61) between 1986 and 2010 in PCR for SPI-1, SPI-2 and undistinguishable from the genes.
Brazil. virulence-associated plasmid outbreak strains.
(13 genes: invA, sipA, sipD,
sopB, sopD, sopE2, ssaR, sifA,
spvB, prot6E, flgK, fljB flgL).
4 Rowlands 237 Salmonella strains isolated PCR for virulence genes of The pathogenic potential is The invA was detected in all
et al. (2014) from food sources between 1983 spvC, invA, sefA and pefA. highlighted with the presence strains; spvC and pefA were
[115], Brazil and 2007, among them 42 were of the four virulence genes in S. found in 48.1% and 44.3%
isolated from foods associated Enteritidis. of strains, respectively; sefA
with foodborne salmonellosis was detected in 31.6% of
and 195 from food samples not the strains and only among
associated with outbreaks or S. Enteritidis.
sporadic cases.
5 Mohamed 309 S. Typhimurium and S. Pulsed field gel Production of Significant difference was Chilling has no effect on the
et al. (2014) Kentucky isolates recovered electrophoresis (PFGE) for aerobactin and found in the production of tested virulence factors.
[78], U.S. from pre- and post-chill whole isolates comparison, and PCR colicin. aerobactin and colicin between
broiler carcasses. for three virulence factors S. Typhimurium and S.
(InvA, pagC, spvC). Kentucky isolates (S.
Kentucky > S. Typhimurium).
6 Diarra et al. 193 Salmonella isolates PFGE for isolates comparison The invA and spvC were
(2014) [85], recovered from commercial PCR for two virulence factors found in 97.9% and 25.9%
Canada farms (InvA, spvC) of the isolates, respectively,
with 33 (71.7%) of the 46 S.
Typhimurium isolates and
17 (65.4%) of the 26 S.
Enteritidis isolates carrying
both invA and spvC.
7 Graziani 112 strains of S. Napoli isolated PFGE for cluster analysis, PCR 10 distinct virulence profiles All the S. Napoli strains
et al. (2011) in Italy from human cases, foods for 10 virulence genes, (VP) were tested, but VP1-2-3 lacked spvC, the avrA gene
[23], Italy of animal origin, and the including five targets (avrA, included 99 out of 112 strains. in SPI, present in about
environment. ssaQ, mgtC, spi_4D, and sopB) These three profiles are similar 100% of the main serovars,
located on the SPI 1–5, three to each other, with single-gene was detected in 5 out of
targets (gipA, sodC1, and variations; compared to VP1, 112 S. Napoli strains only.
sopE1) on prophages, spvC on VP2 lacks the bcfC gene,
plasmid, and one (bcfC) on a whereas the VP3 lacks the
fimbrial cluster. spi_4D gene.
8 Saroj et al. 110 Salmonella isolates of 6 PCR (SPI-8 and SPI-10). SPI-8 and SPI-10 are serovar
(2008) serovars from sprouts, fish, food specific, which is contradictory
[116], India and blood, in India. to previous statement “SPI-8
and SPI-10 are widely
distributed among Salmonella
serovars”.
9 Majtan et al. 14 S. Typhimurium, 7 S. Oligonucleotide microarray Minimal variability of
(2007) [82], Kentucky and 3 S. Enteritidis and PCR confirmation (16 virulence factors encoded
Slovakia from patients suffering from pathogenicity-related markers by SPIs, two S.
various forms of including genes coding for Typhimurium isolates were
gastroenterocolitis. effector proteins associated defective in spaO and three
with SPIs and fimbriae). S. Kentucky strains were
(continued on next page)

10
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 4 (continued )
Authors, Description of the isolates Genes detected by PCR Phenotypic analysis Main findings of the study Virulence genes specifically
year, focused on
country

missing sptP located within


SPI-1.
10 Chen et al. 12 Salmonella and 7 E. coli a PCR based microarray (10 Each Salmonella isolate
(2005) isolates recovered from retail Salmonella virulence genes harbored virulence genes
[117], U.S. ground meat and food animals. including virulence plasmid, including sipA, sipB, sipC,
SPI-1 and SPI-5, flagella). and sop that reside on SPI-1,
SPI-5, as well as other
virulence factors such as
pagC, with variance in
virulence plasmid and invA.
11 Biswas et al. 67 Salmonella strains belonging PCR for bapA BapA gene was found to be
(2011) to 34 serovars isolated from highly conserved in Salmonella.
[118], India diverse sources in India
including both reference strains
and field isolates from different
sources.

PFGE – Pulsed field gel electrophoresis.


MLST – Multi-locus sequence typing.
PCR – Polymerase chain reaction.
NTS – Non-typhoidal Salmonella.
SPI – Salmonella pathogenicity islands.

6. Status and potential of integration of WGS with QMRA for Moreover, the occurrence of frequent horizontal gene transfer among
salmonella pathogen strains could cause strain discrepancy, which could cause false-
negative PFGE results in the event of an actual transmission, which in
QMRA is a widely applied method that partially or wholly in- turn could contribute significantly to the acquisition of false-negative
corporates factors from the farm-to-consumption paradigm to identify results in Salmonella [46]. The first significant application of WGS that
measures (or control steps) that can be taken to prevent and reduce mi- led to source identification of a Salmonella foodborne outbreak was re-
crobial contamination along the (farm-to-fork) pathway. QMRA for Sal- ported in 2011 [16]. Epidemiological investigations into this multistate
monella has been carried out worldwide in eggs and in common food outbreak (300 illnesses reported from 44 states and the District of
animals including meats such as broiler chicken, turkey, pork, and beef Columbia in the U.S.) of S. Montevideo which occurred between 2009
[93–96], nuts [97], leafy greens [98], and more specifically, retail, and 2010 (http://www.cdc.gov/salmonella/montevideo/) indicated
catering [99], and pet food [8]. QMRA methods, driven and challenged black and red peppers used to prepare salami as being the source of the
by better data and the inclusion of uncertainty and variability, have illness. Subsequent investigations into the supply chain of the spices
advanced greatly over the past two decades, allowing risk assessors to established the link between patients, the food product manufacturer,
describe the physical and biological processes of Salmonella across the and the ingredients supplier, which was confirmed by WGS of 35 strains
farm-to-fork pathway. of Salmonella [16].
The nature of risk of Salmonella infection in humans and animals Given the evident genotypic plasticity and anticipated diversity of
depends on a variety of bacterial and host factors and their complex virulence of Salmonella, the robust and high-resolution genomic data
interactive environment. This is a point that current risk assessment provided by WGS has yielded new insights into the pathogenic trans-
strategies have not sufficiently addressed, despite the increase in focus on mission pathways. These, in turn, have provided researchers with the
the importance of the biological variations in Salmonella on predictive possibility of correlating disease manifestation with the diversity in
microbiology [100]. The variations in pathogenicity and host adaptive- pathogenic genomic sequences within a geographic area or food category
ness of Salmonella might directly and immensely influence the to anticipate changes in pathogenicity, thereby facilitating research into
dose-response evaluation step, as well as other steps in risk assessment the behavior of Salmonella along the food chain. An example of this is
[101,102]. As mentioned previously, Salmonella is classified into various when Duchene et al. (2016) estimated the evolutionary rates of Salmo-
serovars. However, to date, published risk assessment of Salmonella has nella and other pathogenic species to understand the time-scale of genetic
not integrated serovar data or specified a certain serovar, although some diversification, which in turn would help in understanding major
have mentioned that the difference in pathogenic potential presented as a evolutionary and ecological processes [105]. Ultimately, in 2018, re-
significant uncertainty factor [8,94–96,101,102]. searchers from CFSAN presented a framework for combining the counts
Serovar is determined by the expression of a few surface antigens that, of nucleotide differences (in terms of single nucleotide polymorphisms
in theory, can only provide limited information about the pathogenicity of (SNPs)) with evolutionary phylogenetic trees and bootstrap support to
different Salmonella strains. Furthermore, classification by serovar may be interpret WGS data and analytical results, in order to determine if more
influenced by the relative occurrence of different serovars in different than one species of bacteria arose from the same source of contamination
geographic locations [10]. In other words, surface antigens are only in- [106].
direct markers that tell us little about the pathogenic phenotype, providing Despite these advances in WGS and WGS-related bioinformatics
limited information for epidemiological investigations. The fact that ge- support, currently available Salmonella serovar data does not have suf-
netic differences within a serovar can have profound influence on the ficient granularity for integration into risk assessment. Therefore, at this
pathogenicity of the isolate has already been noted; for example, S. Par- point, grouping Salmonella according to genotypes attributable to human
atyphi B strains from systemic infections always lack the avrA gene but health risk would be immensely more meaningful to public health than
contain sopE1, whereas those from enteric infections generally display the species- or serovar-level classification and analyses, indicating that
opposite genetic pattern [103]. Compared to surface antigens, the geno- genomic analyses will allow for better incorporation of variability among
type of an organism at dozens or hundreds of loci provides a greater po- bacterial strains in QMRA models [101,104]. To date, very few papers
tential for revealing its human health risk [102,104]. have managed to directly integrate genomic data into health risk

11
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 5
Comprehensive proteomic or genoproteomic studies of Salmonella.
Authors, Description of the isolates Primary purpose of the study – Methods used Main findings of the study
year, microbial function to be elucidated
country

1 Tran et al. S. Typhimurium wild type strain Comparative analysis of the Proteins extracted from bacterial cell The RstA/RstB and PhoPQ systems
(2016) [87], ATCC14028, and ΔPhoPQ, functional networks under the control cultures grown under minimal were found to regulate 80 and 85
China & U.S. ΔrstB::Cm, and Δudp::Km mutants. of the PhoP/PhoQ and RstA/RstB conditions and subjected to liquid metabolic pathways, respectively.
two-component regulatory systems chromatography-tandem mass The PhoPQ system overrepresented
(TCS), which regulates pathogenesis. spectrophotometry (LC-MS/MS). 20 biological pathways, while the
RstA/RstB pathway overrepresented
only 6.
8 proteins were repressed by both
pathways, including 4 ribosomal
proteins (RpmB, RpsT, RpsO, and
RplY) and 2 primary metabolic
process-related proteins (FabZ and
TtcA).
RstA/RstB system was found to
specifically regulate the expression of
proteins responsible for pyrimidine
metabolism, iron acquisition,
Salmonella motility, and invasion.
2 Liu et al. S. Typhimurium SL1344 wild-type, Comprehensive proteomic expression Bacterial proteins isolated from Salmonella employs a borad range of
(2015) [88], ΔentC and ΔfeoAB mutants. of S. Typhimurium upon invasion and infected human epithelial (HeLa) cells adaptation mechanisms to survive in
China infection of nonphagocytic epithelial and subjected LC-MS/MS. host intracellular environments.
cells. Since Salmonella encounters severe
metal ion starvation in host epithelial
cells and upregulates iron,
manganese, and zinc-uptake systems
(EntC, IroN, IroC, CirA, ZnuA, ZnuC,
SitABCD, and FepA) at 6 h post-
infection.
Salmonella also upregulates the
PhoPQ regulatory system, and several
proteins from SPI-2 such as the SsaQn,
PipB, SseL, PgtE, and PagN, which are
important for its intracellular survival
and proliferation.
3 Liu et al. S. Typhimurium S344 wild type Comprehensive characterization of Bacterial proteins isolated from At 18 h post-infection, significant
(2017) [89], strain, and mutant ΔpfkAB, Δgnd, bacterial metabolism, motility, and infection HeLa cells and subjected to downregulation was observed in
China ΔpflB, ΔpurM, ΔdeoC strains. chemotaxis in host intracellular LC-MS/MS. Salmonella flagellar (FliC, FliF, FliG,
conditions. FliL, FliM, etc.) and chemotaxis
(CheA, CheW, CheZ, and CheY)
proteins, indicating loss of motility
and chemotaxis.
Late in the infection stages,
additionally, the tricarboxylic acid
(TCA) cycle and aerobic and
anaerobic respiration pathways were
repressed, leading to utilization of
glycolysis, pentose phosphate
pathway, mixed acid fermentation,
and nucleotide metabolism.
These results indicate the specific
mechanisms and factors responsible
for bacterial fitness during host
infection.
4 Karash et al. S. Typhimurium ATCC 14028s Determine Salmonella proteomic and Analysis conducted using electrospray 121 and 125 proteins were
(2017) [90], wild type strain, mutagenized with genotypic response to hydrogen ionization (ESI)-LC-MS/MS. upregulated and downregulated,
U.S. E. coli SM10 λpir. peroxide produced by macrophages respectively, under hydrogen
during invasion and infection. peroxide (oxidative) stress conditions.
Specifically, this study found that the
expression of amino acid biosynthesis
proteins aroB, aroD, aroE_2, aroK, and
aroA were significantly associated
with fitness under the stress
condition.
Additionally, proteins encoded by
ahpCF, tsaA, tpx, sodA, sodB, sodCI,
and sodCII played a significant role in
oxygen scavenging, and hence
promoting bacterial survival in within
the macrophage system.
5 Li et al. S. Typhimurium wild type strains Compare proteomic expression of S. Salmonella proteins isolated from Overall, many proteomic features
(2019) [91], SL1344 and SL14028s. Typhimurium from infected infected HeLa and human were shared between the two types of
China host cells, indicating a similarity in
(continued on next page)

12
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Table 5 (continued )
Authors, Description of the isolates Primary purpose of the study – Methods used Main findings of the study
year, microbial function to be elucidated
country

macrophages against those from macrophage (RAW 264.7) cell lines intracellular environmental cues. This
infected epithelial cells (HeLa). analyzed by LC-MS/MS. included the induction of metal ion
transportation and utilization
pathways and repression of TCA and
anaerobic respiration pathways.
However, the time required for
intracellular adaptation varied,
especially in the cases of SPI-1 T3SS
and SPI-2.
Uniquely, Salmonella expressed
protein signatures indicative of
histidine biosynthesis when infecting
macrophage cultures, indicating a
path for Salmonella suppression in this
host cell type.
6 Noster et al. Wild type strain - S. Typhimurium Compare the intracellular proteomic Salmonella proteins isolated from Infection-associated proteins such as
(2019) [92], NCTC12023, and isogenic mutant response of wild-type S. Typhimurium infected macrophage culture and SlyA/B, PhoP/N, Fur, Hfq, YdcR,
Germany strains ΔssaV::FRT, defective in to mutant strains with aberrant subjected to liquid chromatography- SodC1 and PntA, were upregulated in
SPI2-T3SS, and ΔsseF::FRT, Salmonella-induced filament (SIF) mass spectrophotometry. the ΔsseF mutant, allowing for
defective in the SPI2-T3SS effector architecture against nutritional stress increased intracellular replication
protein SseF. and host cell defense mechanism. under macrophage stress conditions.
Induction of SIFs required for
sufficient nutritional supply.
While nutritional limitations slightly
reduces ΔsseF mutant replication, the
mutant ΔssaV displays the additional
inability to neutralize host defense
mechanisms.

SPI – Salmonella pathogenicity island.


T3SS – Type III secretion system.
LC-MS/MS – Liquid chromatography – tandem mass spectrophotometry.
TCA – tricarboxylic acid cycle.

assessment [60,107–109]. Previously, Pielaat et al. (2015) explored the data alone to predict pathogenicity could give rise to further uncertainty
integration of WGS data in hazard characterization of Shiga as the existence of a particular virulence or antibiotic resistance gene is
toxin-producing E. coli (STEC) O157 [60] by conducting a Genome Wide not necessarily indicative of phenotypic expression. Phenotypic hetero-
Association Study (GWAS), wherein genetic data (virulence SNPs) was geneity may also be caused by mutations and acquisition of genes
correlated with phenotypic data (in vitro adherence to epithelial cells as a through horizontal gene transfer, as well as by epigenetic mechanisms
proxy for virulence). On the other hand, Gale et al. built a dose–response that are not accompanied by changes in the genome content [101].
model for RNA viruses by breaking down initial infection in the gastro- It can be safely summarized that the challenge of converting primary
intestinal tract into the component steps at the molecular level and WGS data into useful public health risk assessment remains unmet, and
assigning probabilities to each step, which led to the conclusion that the that experience with such data is quite limited at this point. In addition,
molecular mechanisms are not sufficiently well understood at present to there is no doubt that gene expression studies, transcriptomics, prote-
enable quantitative estimation of probabilities on the basis of omics data omics, metabolomics, high-throughput phenotyping approaches, and
[107]. More recently, Njage and colleagues employed machine learning other “omics” studies, coupled with genomic studies, would be more
approaches to link genome composition with clinical outcome in informative in the future QMRA of Salmonella.
humans, by identifying virulence genes that were important predictors of
higher frequency of illness for Listeria monocytogenes and STEC to update 7. Future recommendations
the hazard characterization paradigm of the risk assessment framework
[108,109]. However, the potential of such an estimation must not be Based on the above findings, it is safe to conclude that multiple in-
neglected. All of these studies took a baby step towards the application of dividual virulence genes are variably distributed in Salmonella. It is quite
genomic data of E. coli, Listeria and RNA virus (whose genomic content unrealistic to expect a single locus to be responsible for all pathogenic
and phenotypic traits are simpler than those of Salmonella) in a proba- traits; instead, a complicated combination of genes is more likely to
bilistic framework. In the case of Salmonella, high levels of genome contribute to the overall virulence phenotype. It can be concluded that
similarity do not necessarily imply similar behavior in the food chain or comparative studies analyzing the differences in genome content among
similar levels of pathogenic phenotype, since small genetic changes may different “groups” of Salmonella must be performed to assess the impor-
result in large phenotypic differences [39,110], which increases the tance of differences in virulence characteristics among strains. A more
complexity of the data integration process. realistic approach to relate phenotypic differences with genetic de-
The fields of QMRA and genomic analyses have both seen significant terminants would be to compare closely related bacterial lineages that
development over the past 15 years [60,101,104,108–110]. Genomic exhibit distinct biological properties. In this way, genes that are directly
approaches offer the potential to both inform QMRA and transform how associated with special pathogenic phenotypes could be maximally
it is conducted. QMRA of Salmonella if further challenged by the focused. Furthermore, a majority of the studies continue to focus on the
increasingly comprehensive descriptions of the complex, dynamic commonly detected gastroenteritis serovars (viz. S. Typhimurium and S.
behavior of bacteria within the context of the host provided by WGS data. Enteriditis); therefore, future research should include more serovars.
Therefore, there is a long way to go before WGS data can be routinely Due to the lack of both standard sequencing and post-bioinformatics
used in QMRA of Salmonella. It should also be noted that the use of WGS methods for WGS and corresponding phenotypic analysis, researchers are

13
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

currently employing different platforms and bioinformatics tools for genomic technologies in food safety risk assessment is currently in flux
WGS analysis (and for the analysis of its metadata). Metadata that would without a clearly defined path and methodology. However, with the
be required to perform such comparative research would include viru- accumulation and standardization of WGS data, the normalization and
lence gene profiling, phylogenetic analyses, phenotypic characteristics, availability of advanced proteomics, and increasing utilization of ma-
and the resulting epidemiological traits. The main limiting factor, how- chine learning and advanced data analytic techniques, it would be
ever, would not be the cost of sequencing; rather, it would be how to possible to make the best use of all available data in order to improve the
assemble, process, and handle large amounts of WGS data (as well as its efficiency of QMRA in the near future. This will undoubtedly provide
metadata) in a standardized way that will make the information useful, in new insights and inform actions to mitigate the risk caused by Salmonella
this case for food safety risk assessment. Therefore, further research must in chicken, as well as other pathogenic agents in food.
be directed at the following focal points:
Declaration of competing interest
 Metadata that could be of value in determining the role of food in the
epidemiology of isolates is rare and, when present, incomplete. None.
Detailed epidemiological information corresponding to molecular
data should be collected and provided in the future. References
 Genomics can be used to identify genes responsible for a variety of
factors. Experimental data remains essential for improving the un- [1] European Food Safety Authority EFSA Panel on Biological Hazards (BIOHAZ),
“Scientific opinion on monitoring and assessment of the public health risk of
derstanding of health risk posed by foodborne pathogens. ‘Salmonella Typhimurium-like’ strains, EFSA J. 8 (10) (2010) 1–48.
 Methods to directly screen for virulence-associated phenotypes in [2] M.D. Kirk, S.M. Pires, R.E. Black, M. Caipo, J.A. Crump, B. Devleesschauwer, et al.,
animal models are required to take a more comprehensive approach World Health Organization estimates of the global and regional disease burden of
22 foodborne bacterial, protozoal, and viral diseases, 2010: a data synthesis, PLoS
to define genes essential for in vivo infection. Med. 12 (12) (2015) 1–21.
 It is unrealistic and meaningless to attempt the mapping of microbial [3] E.L. Snary, A.N. Swart, T. Hald, Quantitative microbiological risk assessment and
WGS data consisting of thousands of loci onto a probabilistic scale for source attribution for Salmonella: taking it further, Risk Anal. 36 (3) (2016)
433–436.
risk. Considering the genotype-phenotype diversity, another potential [4] World Health Organization, WHO, WHO Estimates of the Gobal Burden of
way is to model the variability of multiple virulence genes identified Foodborne Diseases. Foodborne Disease Burden Epidemiology Reference Group
by WGS among the various Salmonella isolates within one food 2007 - 2015, World Health Organization, ” Switzerland, 2018.
[5] P. Antunes, J. Mour~ao, J. Campos, L. Peixe, Salmonellosis: the role of poultry
category or one serovar. Quantification and calibration of the dif-
meat, Clin. Microbiol. Infect. 22 (2) (2016) 110–121.
ferences (such as SNP variants) between genomes will facilitate the [6] M.K. Fatica, K.R. Schneider, Salmonella and produce: survival in the plant
categorization of Salmonella, allowing us to treat them accordingly. environment and implications in food safety, Virulence 2 (6) (2011) 573–579.
 Genome Wide Association Studies (GWAS), which have been devel- [7] B.R. Jackson, P.M. Griffin, D. Cole, K.A. Walsh, S.J. Chai, Outbreak-associated
Salmonella Enterica serotypes and food commodities, United States, 1998-2008,
oped in other areas (but not in foodborne microorganisms), could Emerg. Infect. Dis. 19 (8) (2013) 1239–1244.
facilitate the management of large sets of phenotype and genotype [8] E. Lambertini, R.L. Buchanan, C. Narrod, R.M. Ford, R.C. Baker, A.K. Pradhan,
data, analysis of phenotypic variation and heritability, and most Quantitative assessment of human and pet exposure to Salmonella associated with
dry pet foods, Int. J. Food Microbiol. 216 (2016) 79–90.
importantly, generation of a refined set of candidate single nucleotide [9] C.D.C. United States Centers for Disease Control and Prevention, Reports of
polymorphisms (SNPs). Salmonella Outbreak Investigations from 2019, 2019. Retrieved from U.S. Centers
 The main biological process or (potentially) food processing chain can for Disease Control and Prevention, https://www.cdc.gov/salmonella/outbreaks-
2019.html. December 20, 2019.
be broken down into its component steps such as (surviving the [10] A. Jacobsen, R.S. Hendriksen, F.M. Aaresturp, D.W. Ussery, C. Friis, The
various) process conditions and time required to initiate growth or Salmonella Enterica pan-genome, Microb. Ecol. 62 (3) (2011) 487–504.
invasion of gut epithelium, which would guide the data requirements [11] C.D.C. United States Centers for Disease Control and Prevention, National
Salmonella Surveillance Annual Report, 2012, 2012 atlanta, ga. Retrieved from
(e.g. specific gene expression data and phenotypic data as cells grow). U.S. Centers for Disease Control and Prevention, https://www.cdc.gov/ncezid/
 Recent outbreaks in the U.S. have been linked to vegetables, nuts, and dfwed/pdfs/salmonella-annual-report-2012-508c.pdf. (Accessed 20 December
fruits, in addition to traditional sources (food animals). Therefore, 2016).
[12] European Food Safety Authority, EFSA and European Center for Disease
Salmonella strains could have significant phenotype diversity and
Prevention and Control, The European Union summary report on trends and
potential genomic variability in host specificity or colonization that sources of zoonoses, zoonotic agents and food-borne outbreaks in 2011, EFSA J.
could be worth looking into. The potential findings might contribute 11 (4) (2013) 3129.
significantly to the risk assessment of these food sources. [13] M. Hugas, P.A. Beloeil, Controlling Salmonella along the food chain in the
European Union - progress over the last ten years, Euro. Surv. 19 (19) (2014).
[14] A.S. Dhanani, G. Block, K. Dewar, V. Forgetta, E. Topp, R.G. Beiko, M.S. Diarra,
8. Conclusion Genomic comparison of non-typhoidal Salmonella Enterica serovars Typhimurium,
Enteritidis, Heidelberg, Hadar and Kentucky isolates from broiler chickens, PLoS
One 10 (6) (2015) 1–24.
It should be mentioned that the primary aim of this review was to [15] H.S. Hayden, S. Matamouros, K.R. Hager, M.J. Brittnacher, L. Rohmer,
pool genomic data with its corresponding phenotypic data in order to M.C. Radey, et al., Genomic analysis of Salmonella Enterica serovar Typhimurium
potentially conduct a quantitative modeling study to correlate genotypes characterizes strain diversity for recent U.S. salmonellosis cases and identifies
mutations linked to loss of fitness under nitrosative and oxidative stress, mBio 7
with their corresponding phenotypes. However, as our literature search (2) (2016), e00154.
has identified gaps and inconsistencies in this research such as the [16] E.K. Lienau, E. Strain, C. Wang, J. Zheng, A.R. Ottesen, C.E. Keys, et al.,
limited number of related studies, diversity in the methodology of Identification of a salmonellosis outbreak by means of molecular sequencing,
N. Engl. J. Med. 364 (2011) 981–982.
phenotypic testing, and variability in data obtained from even the most [17] United States Centers for Disease Control and Prevention, CDC, List of Selected
common epithelial cell invasion assays, conducting a quantitative anal- Multistate Foodborne Outbreak Investigations, 2019. Retrieved from U.S. Centers
ysis at this point might not be feasible. Although comparative genomic for Disease Control and Prevention, https://www.cdc.gov/foodsafety/out
breaks/multistate-outbreaks/outbreaks-list.html. December 20, 2019.
studies for Salmonella using WGS has seen a considerable amount of in- [18] M.F. Anjum, C. Marooney, M. Fookes, S. Baker, G. Dougan, A. Ivens,
terest over the past decade, a larger number of studies have reported the M.J. Woodward, Identification of core and variable components of the Salmonella
use of comparative genomic hybridization (CGH) and PCR-based tech- enterica subspecies I genome by microarray, Infect. Immun. 73 (12) (2005)
7894–7905.
niques for comparative genomic analysis of Salmonella. The reviewed
[19] M. Hoffmann, S. Zhao, J. Pettengill, Y. Luo, S.R. Monday, J. Abbott, et al.,
studies that attempted to correlate phenotypic differences in different Comparative genomic analysis and virulence differences in closely related
serovars with genetic variations in order to gain insights into the effect of Salmonella enterica serotype Heidelberg isolates from humans, retail meats, and
genome variations on apparent differences in prevalence and other fea- animals, Genom. Biol. Evol. 6 (5) (2014) 1046–1068.

tures provide limited information. Additionally, the transition to

14
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

[20] J. Fierer, D.G. Guiney, Diverse virulence traits underlying different clinical [51] T. Inns, P. Ashton, S. Herrera-Lyon, J. Lighthill, S. Foulkes, T. Jombart, et al.,
outcomes of Salmonella infection, J. Clin. Invest. 107 (7) (2001) 775–780. Prospective use of whole genome sequencing (WGS) detected a multi-country
[21] A. Marzel, P.T. Desai, A. Goren, Y.I. Schorr, I. Nissan, S. Porwollik, et al., Persistent outbreak of Salmonella Enteritidis, Epidemiol. Infect. (2017) 289–298.
infections by nontyphoidal Salmonella in humans: epidemiology and genetics, [52] V. Kozyreva, J. Crandall, A. Sabol, A. Poe, P. Zhang, J. Concepcion-Acevedo, et al.,
Clin. Infect. Dis. 62 (7) (2016) 879–886. Laboratory investigation of Salmonella enterica serovar Poona outbreak in
[22] F. Capuano, A. Mancusi, R. Capparelli, S. Esposito, Y.T.R. Proroga, California: comparison of pulsed-field gel electrophoresis (PFGE) and whole
Characterization of drug resistance and virulotypes of Salmonella strains isolated genome sequencing (WGS) results, PLoS Curr. 22 (2016) 8.
from food and humans, Foodb. Pathog. Dis. 10 (11) (2013) 963–968. [53] S. Simon, E. Trost, J. Bender, S. Fuchs, B. Malorny, W. Rabsch, et al., Evaluation of
[23] C. Graziani, L. Busani, A.M. Dionisi, A. Caprioli, S. Ivarsson, I. Hendenstrom, et al., WGS based approaches for investigating a food-borne outbreak caused by
Virulotyping of Salmonella Enterica serovar Napoli strains isolated in Italy from Salmonella enterica serovar Derby in Germany, Food Microbiol. (2018) 46–54.
human and nonhuman sources, Foodb. Pathog. Dis. 8 (9) (2011) 997–1003. [54] B. Diep, C. Barretto, A. Portmann, C. Fournier, A. Karczmarek, G. Voets, et al.,
[24] A.J. Van Asten, J.E. Van Dijk, “Distribution of ‘classic’ virulence factors among Salmonella serotyping; comparison of the traditional method to a microarray-
Salmonella spp.” FEMS Immunol, Med. Microbiol. 44 (3) (2005) 251–259. based method and an in silico platform using whole genome sequencing data,
[25] R. Figueiredo, R. Card, C. Nunes, M. AbuOun, M.C. Bagnall, J. Nunez, et al., Front. Microbiol. 2554 (2019).
Virulence characterization of Salmonella enterica by a new microarray: detection [55] K. Kubota, W. Wolfgang, D. Baker, D. Boxrud, L. Turner, E. Trees, et al., PulseNet
and evaluation of the cytolethal distending toxin gene activity in the unusual host and the changing paradigm of laboratory-based surveillance for foodborne
S. Typhimurium, PLoS One 10 (8) (2015) 1–14. diseases, Publ. Health Rep. 22S-28S (2019).
[26] E. Litrup, M. Torpdahl, B. Malorny, S. Huehn, H. Christensen, E.M. Nielsen, [56] L. Siira, U. Naseer, K. Alfsnes, N. Hermansen, H. Lange, L. Brandal, Whole genome
Association between phylogeny, virulence potential and serovars of Salmonella sequencing of Salmonella Chester reveals geographically distinct clusters, Norway,
enterica, Infect. Genet. Evol. 10 (7) (2010) 1132–1139. 2000 to 2016, Euro. Surv. 24 (4) (2019).
[27] J.E. Gomez, A. Clatworthy, D.T. Hung, Probing bacterial pathogenesis with [57] J. Jones, L. Wang, O. Ceric, S. Nemser, D. Rotstein, D. Jurkovic, et al., Whole
genetics, genomics, and chemical biology: past, present, and future approaches, genome sequencing confirms source of pathogens associated with bacterial
Crit. Rev. Biochem. Mol. Biol. 46 (1) (2011) 41–66. foodborne illness in pets fed raw pet food, J. Vet. Diagn. Invest. (2019) 235–240.
[28] T.M. Wassenaar, W. Gaastra, “Bacterial virulence: can we draw the line?”, FEMS [58] C. Jenkins, T. Dallman, K. Grant, Impact of whole genome sequencing on the
Microbiol. Lett. 201 (1) (2001) 1–7. investigation of food-borne outbreaks of Shiga toxin-producing Escherichia coli
[29] A.C. Fluit, “Towards more virulent and antibiotic-resistant Salmonella?” FEMS serogroup O157:H7, England, 2013-2017, Euro. Surv. 24 (4) (2019).
Immunol, Med. Microbiol. 43 (1) (2005) 1–11. [59] S.G. Schjørring, Cross-border outbreak of listeriosis caused by cold-smoked
[30] R.C. Laughlin, L.A. Knodler, R. Barhoumi, H.R. Payne, J. Wu, G. Gomez, et al., salmon, revealed by integrated surveillance and whole genome sequencing (WGS),
Spatial segregation of virulence gene expression during acute enteric infection Denmark and France, 2015 to 2017, Euro. Surv. 22 (50) (2017).
with Salmonella enterica serovar Typhimurium, mBio 5 (1) (2014) 1–11. [60] A. Pielaat, M.P. Boer, L.M. Wijnands, A.H. van Hoek, E. Bouw, G.C. Barker, et al.,
[31] C. Smith, A.M. Stringer, C. Mao, M.J. Palumbo, J.T. Wade, Mapping the regulatory First step in using molecular data for microbial food safety risk assessment; hazard
network for Salmonella enterica serovar Typhimurium invasion, mBio 7 (5) (2016) identification of Escherichia coli O157:H7 by coupling genomic data with in vitro
1–14. adherence to human epithelial cells, Int. J. Food Microbiol. 213 (2015) 130–138.
[32] E. Fass, E.A. Groisman, Control of Salmonella pathogenicity island-2 gene [61] G. Satta, M. Lipman, G.P. Smith, C. Arnold, O.M. Kon, T.D. McHugh,
expression, Curr. Opin. Microbiol. 12 (2) (2009) 199–204. Mycobacterium tuberculosis and whole genome sequencing: how close are we to
[33] R. Figueira, D.W. Holden, Functions of the Salmonella pathogenicity island 2 (SPI- unleashing its full potential? Clin. Microbiol. Infect. 24 (6) (2018) 604–609.
2) type III secretion system effectors, Microbiol. 158 (5) (2012) 1147–1161. [62] L. Collineau, P. Boerlin, C.A. Carson, B. Chapman, A. Fazil, B. Hetman, et al.,
[34] G. Cao, M. Allard, E. Strain, R. Stones, S. Zhao, E. Brown, J. Meng, Genetic Integrating whole-genome sequencing data into quantitative microbial risk
diversity of Salmonella pathogenicity islands SPI-5 and SPI-6 in Salmonella assessment of foodborne antimicrobial resistance: a review of opportunities and
Newport, Foodb. Pathog. Dis. 11 (10) (2014) 798–807. challenges, Front. Microbiol. 10 (2019) 1107.
[35] J. Kaur, S.K. Jain, Role of antigens and virulence factors of Salmonella enterica [63] M. Bersanelli, E. Mosca, D. Remondini, E. Giampieri, C. Sala, G. Castellani, et al.,
serovar Typhi in its pathogenesis, Microbiol. Res. 167 (4) (2012) 199–210. Methods for the integration of multi-omics data: mathematical aspects, BMC
[36] T.A. Agbor, B.A. McCormick, Salmonella effectors: important players modulating Bioinf. 17 (2) (2016) 15.
host cell function during infection, Cell Microbiol. 13 (12) (2011) 1858–1869. [64] S. Cosentino, M. Voldby Larsen, F. Møller Aarestrup, O. Lund, PathogenFinder –
[37] C. Gyles, P. Boerlin, Horizontally transferred genetic elements and their role in distinguishing friend from foe using bacterial whole genome sequence data, PLoS
pathogenesis of bacterial disease, Vet. Path. 51 (2) (2014) 328–340. One 8 (10) (2013), e77302.
[38] A.I. Moreno Switt, H.C. den Bakker, C.A. Cummings, L.D. Rodriguez-Rivera, [65] L. Chen, Z. Xiong, L. Sun, J. Yang, Q. Jin, VFDB 2012 update: toward the genetic
G. Govoni, M.L. Raneiri, et al., Identification and characterization of novel diversity and molecular evolution of bacterial virulence factors, Nucleic Acids Res.
Salmonella mobile elements involved in the dissemination of genes linked to 40 (2012) D641–D645.
virulence and transmission, PLoS One 7 (7) (2012), e41247. [66] S.H. Yoon, Y.K. Park, S. Lee, D. Choi, T.K. Oh, C.G. Hur, et al., Towards
[39] L. Betancor, L. Yim, M. Fookes, A. Martinez, N.R. Thomson, A. Ivens, et al., pathogenomics: a web-based resource for pathogenicity islands, Nucleic Acids Res.
Genomic and phenotypic variation in epidemic-spanning Salmonella enterica 35 (2007) D395–D400.
serovar Enteritidis isolates, BMC Microbiol. 9 (2009) 237. [67] A.P. Underwood, A. Mulder, S. Gharbia, J. Green, Virulence Searcher: a tool for
[40] X. Didelot, R. Bowden, T. Street, T. Golubchik, C. Spencer, G. McVean, et al., searching raw genome sequences from bacterial genomes for putative virulence
Recombination and population structure in Salmonella enterica, PLoS Genet. 7 (7) factors, Clin. Microbiol. Infect. 11 (9) (2005) 770–772.
(2011), e1002191. [68] J.M. Bartoszewicz, A. Seidel, R. Rentzsch, B.Y. Renard, DeePAC: predicting
[41] X. Deng, H.C. den Bakker, R.S. Hendriksen, Genomic epidemiology: whole- pathogenic potential of novel DNA with reverse complement neural networks,
genome-sequencing-powered surveillance and outbreak investigation of Bioinformatics 36 (1) (2019) 81–89.
foodborne bacterial pathogens, Annu. Rev. Food Sci. Technol. 7 (2016) 353–374. [69] C. Deneke, R. Rentzsch, B. Renard, PaPrBaG: a machine learning approach for the
[42] J. Ronholm, N. Nasheri, N. Petronella, F. Pagotto, Navigating microbiological food detection of novel pathogens from NGS data, Sci. Rep. 7 (2017) 39194.
safety in the era of whole-genome sequencing, Clin. Microbiol. Rev. 29 (4) (2016) [70] M.D. Whiteside, C.R. Laing, A. Manji, P. Kruczkiewicz, E.N. Taboada, V.P. Gannon,
837–857. SuperPhy: predictive genomics for the bacterial pathogen Escherichia coli, BMC
[43] E. Franz, L. Mughini-Gras, T. Dallman, Significance of whole genome sequencing Microbiol. 16 (1) (2016) 65.
for surveillance, source attribution and microbial risk assessment of foodborne [71] H.C. den Bakker, A.I. Moreno-Switt, G. Goroni, C.A. Cummings, M.L. Ranieri,
pathogens, Curr. Opin. Food Sci. 8 (2016) 74–79. Retrieved June 5, 2017, http:// L. Degoricija, et al., Genome sequencing reveals diversification of virulence factor
www.sciencedirect.com/science/article/pii/S221479931630056X. content and possible host adaptation in distinct subpopulations of Salmonella
[44] S. Goodwin, J.D. McPherson, W.R. McCombie, Coming of age: ten years of next- enterica, BMC Genom. 12 (2011) 425.
generation sequencing technologies, Nat. Rev. Genet. 17 (6) (2016) 333–351. [72] W.F. Fricke, M.K. Mammel, P.F. McDermott, C. Tartera, D.G. White, J.E. Leclerc, et
[45] J.C. Kwong, N. McCallum, V. Sintchenko, B.P. Howden, Whole genome al., Comparative genomics of 28 Salmonella enterica isolates: evidence for CRISPR-
sequencing in clinical and public health microbiology, Pathology 47 (3) (2015) mediated adaptive sublineage evolution, J. Bacteriol. 193 (14) (2011) 3556–3568.
199–210. [73] A. Herrero-Fresno, I. Wallrodt, P. Leekitcharoenphon, J.E. Olsen, F.M. Aarestrup,
[46] X. Deng, N. Shariat, E.M. Driebe, C.C. Roe, B. Tolar, E. Trees, et al., Comparative R.S. Hendricksen, The role of the st313-Td gene in virulence of Salmonella
analysis of subtyping methods against a whole-genome-sequencing standard for Typhimurium ST313, PLoS One 9 (1) (2014), e84566.
Salmonella Enterica serotype Enteritidis, J. Clin. Microbiol. 53 (1) (2015) 212–218. [74] Z. Pan, B. Carter, J. Nunez-Garcia, M. Abuoun, M. Fookes, A. Ivens, et al.,
[47] C.U. K€oser, M.J. Ellington, E.J. Cartwright, S.H. Gillespie, N.M. Brown, Identification of genetic and phenotypic differences associated with prevalent and
M. Farrington, et al., Routine use of microbial whole genome sequencing in non-prevalent Salmonella Enteritidis phage types: analysis of variation in amino
diagnostic and public health microbiology, PLoS Pathog. 8 (8) (2012), e1002824. acid transport, Microbiol. 155 (10) (2009) 3200–3213.
[48] P. Pilo, Improving exploitation of whole genome sequencing data for public [75] D.M. Raskin, R. Seshadri, S.U. Pukatzki, J.J. Mekalanos, Bacterial genomics and
health, forensic microbiology and biosafety, EBio Med. 2 (11) (2015) 1566–1567. pathogen evolution, Cell 124 (4) (2006) 703–714.
[49] A. van Belkum, P.T. Tassios, L. Dijkshoorn, S. Haeggman, B. Cookson, N.K. Fry, et [76] L. Yim, L. Betancor, A. Martinez, G. Giossa, C. Bryant, D. Maskell,
al., Guidelines for the validation and application of typing methods for use in J.A. Chabalgoity, Differential phenotypic diversity among epidemic-spanning
bacterial epidemiology, Clin. Microbiol. Infect. 13 (3) (2007) 1–46. Salmonella enterica serovar enteritidis isolates from humans or animals, Appl.
[50] S.H. Xie, A.L. Liu, Y.Y. Chen, L. Zhang, H.J. Zhang, B.X. Jin, et al., DNA damage Environ. Microbiol. 76 (20) (2010) 6812–6820.
and oxidative stress in human liver cell L-02 caused by surface water extracts [77] Z.W. Jaradat, L. Abedel Hafiz, M.M. Ababneh, Q.O. Ababneh, W. Al Mousa, A. Al
during drinking water treatment in a waterworks in China, Environ. Mol. Nabulsi, et al., Comparative analysis of virulence and resistance profiles of
Mutagen. 51 (3) (2010) 229–235.

15
J. Chen et al. Journal of Agriculture and Food Research 2 (2020) 100045

Salmonella Enteritidis isolates from poultry meat and foodborne outbreaks in green vegetables consumed at salad bars, based on modeling supply chain
northern Jordan, Virulence 5 (5) (2014) 601–610. logistics, J. Food Protect. 73 (10) (2010) 1830–1840.
[78] T. Mohamed, S. Zhao, D.G. White, S. Parveen, Molecular characterization of [99] C.O. Møller, M.J. Nauta, D.W. Schaffner, P. Dalgaard, B.B. Christensen,
antibiotic resistant Salmonella Typhimurium and Salmonella Kentucky isolated T.B. Hansen, et al., Risk assessment of Salmonella in Danish meatballs produced in
from pre- and post-chill whole broilers carcasses, Food Microbiol. 38 (2014) 6–15. the catering sector, Int. J. Food Microbiol. 196 (2015) 109–125.
[79] S. Porwollik, R.M.Y. Wong, M. McClelland, Evolutionary genomics of Salmonella: [100] T.P. Oscar, General regression neural network and Monte Carlo simulation model
gene acquisitions revealed by microarray analysis, Proc. Natl. Acad. Sci. Unit. for survival and growth of Salmonella on raw chicken skin as a function of
States Am. 99 (13) (2002) 8956–8961. serotype, temperature, and time for use in risk assessment, J. Food Protect. 72 (10)
[80] S. Porwollik, E.F. Boyd, C. Choy, P. Cheng, L. Florea, E. Proctor, M. Mclelland, (2009) 2078–2087.
Characterization of Salmonella enterica subspecies I genovars by use of [101] S. Brul, J. Bassett, P. Cook, S. Kathariou, P. McClure, P.R. Jasti, R. Betts, “‘Omics’
microarrays, J. Bacteriol. 186 (17) (2004) 5883–5898. technologies in quantitative microbial risk assessment, Trends Food Sci. Technol.
[81] W. Zou, S.F. Al-Khaldi, W.S. Branham, T. Han, J.C. Fuscoe, J. Han, et al., 27 (1) (2012) 12–24.
Microarray analysis of virulence gene profiles in Salmonella serovars from food/ [102] A. Pielaat, G.C. Barker, P. Hendricksen, P. Hollman, A. Peijnenburg, B.H. Ter
food animal environment, J. Infect. Dev. Countries 5 (2) (2011) 94–105. Kuile, A foresight study on emerging technologies: state of the art of omics
[82] T. Majtan, L. Majtanova, J. Timko, V. Majtan, Oligonucleotide microarray for technologies and potential applications in food and feed safety. Report 1: review
molecular characterization and genotyping of Salmonella spp. strains, on the state of art of omics technologies in risk assessment related to food and feed
J. Antimicrob. Chemother. 60 (5) (2007) 937–946. safety, EFSA Support. Publ. 10 (10) (2013) 1–126.
[83] J. Suez, S. Porwollik, A. Dagan, A. Marzel, Y.I. Schorr, P.T. Desai, et al., Virulence [103] R. Prager, W. Rabsch, W. Streckel, W. Voigt, E. Tietze, H. Tschape, Molecular
gene profiling and pathogenicity characterization of non-typhoidal salmonella properties of Salmonella enterica serotype Paratyphi B distinguish between its
accounted for invasive disease in humans, PLoS One 8 (3) (2013), e58449. systemic and its enteric pathovars, J. Clin. Microbiol. 41 (9) (2003) 4270–4278.
[84] S. Huehn, R.M. LaRagione, M. Anjum, M. Saunders, M.J. Woodward, C. Bunge, et [104] T.M. Wassenaar, Risk assessment prediction from genome sequences: promises
al., Virulotyping and antimicrobial resistance typing of Salmonella enterica and dreams, J. Food Protect. 67 (9) (2004) 2053–2057.
serovars relevant to human health in Europe, Foodb. Pathog. Dis. 7 (5) (2010) [105] S. Duch^ene, K.E. Holt, F.-X. Weill, S.L. Hello, J. Hawkey, D.J. Edwards,
523–535. M. Fourment, E.C. Holmes, Genome-scale rates of evolutionary change in bacteria,
[85] M.S. Diarra, P. Delaquis, H. Rempel, S. Bach, C. Harlton, M. Aslam, et al., Microb. Genom. 2 (11) (2016).
Antibiotic resistance and diversity of Salmonella enterica serovars associated with [106] A.W. Pightling, J.B. Pettengill, Y. Luo, J.D. Baugher, H. Rand, E. Strain,
broiler chickens, J. Food Protect. 77 (1) (2014) 40–49. Interpreting whole-genome sequence analyses of foodborne bacteria for regulatory
[86] F.J. Reen, E.F. Boyd, S. Porwollik, B.P. Murphy, D. Gilroy, S. Fanning, applications and outbreak investigations, Front. Microbiol. 9 (2018) 1482.
M. Mclelland, Genomic comparisons of Salmonella enterica serovar Dublin, Agona, [107] P. Gale, A. Hill, L. Kelly, J. Bassett, P. McClure, Y. LeMarc, I. Soumpasis,
and Typhimurium strains recently isolated from milk filters and bovine samples Applications of omics approaches to the development of microbiological risk
from Ireland, using a Salmonella microarray, Appl. Environ. Microbiol. 71 (3) assessment using RNA virus dose-response models as a case study, J. Appl.
(2005) 1616–1625. Microbiol. 117 (6) (2014) 1537–1548.
[87] T.-K. Tran, Q.-Q. Han, Y. Shi, L. Guo, A comparative proteomic analysis of [108] P.M.K. Njage, C. Henri, P. Leekitcharoenphon, M.-Y. Mistou, R.S. Hendricksen,
Salmonella Typhimurium under the regulation of the RstA/RstB and PhoP/PhoQ T. Hald, Machine learning methods as a tool for predicting risk of illness applying
systems, Biochem. Biophys. Acta (BBA) Proteins Proteom. 1864 (12) (2016) next-generation sequencing data, Risk Anal. 39 (6) (2019) 1397–1413.
1686–1695. [109] P.M.K. Njage, P. Leekitcharoenphon, T. Hald, Improving hazard characterization
[88] Y. Liu, Q. Zhang, M. Hu, K. Yu, J. Fu, F. Zhou, X. Liu, Proteomic analyses of in microbial risk assessment using next-generation sequencing data and machine
intracellular Salmonella enterica serovar Typhimurium reveal extensive bacterial learning: predicting clinical outcomes in shigatoxigenic Escherichia coli, Int. J.
adaptations to infected host epithelial cells, Infect. Immun. 83 (7) (2015) Food Microbiol. 292 (2019) 72–82.
2897–2906. [110] C. Fong, D.C. Ko, M. Wasnick, M. Radey, S.I. Miller, M. Brittnacher, GWAS
[89] Y. Liu, K. Yu, F. Zhou, T. Ding, Y. Yang, M. Hu, X. Liu, Quantitative proteomics analyzer: integrating genotype, phenotype and public annotation data for genome-
charts the landscape of Salmonella carbon metabolism within host epithelial cells, wide association study analysis, Bioinformatics 26 (4) (2010) 560–564.
J. Proteome Res. 16 (2) (2017) 788–797. [111] C.J. Blondel, J.C. Jimenez, I. Contreras, C.A. Santiviago, Comparative genomic
[90] S. Karash, R. Liyanage, A. Qassab, J. Lay Jr., Y. Kwon, A comprehensive analysis uncovers 3 novel loci encoding type six secretion systems differentially
assessment of the genetic determinants in Salmonella Typhimurium for resistance distributed in Salmonella Serotypes, BMC Genom. 10 (2009) 354.
to hydrogen peroxide using proteogenomics, Sci. Rep. 7 (2017) 17073. [112] C.A. Morales, S. Porwollik, J.G. Frye, H. Kinde, M. McLelland, J. Guard-Bouldin,
[91] Z. Li, Y. Liu, J. Fu, B. Zhang, S. Cheng, M. Wu, et al., Salmonella proteomic Correlation of phenotype with the genotype of egg-contaminating Salmonella
profiling during infection distinguishes the intracellular environment of host cells, enterica serovar enteritidis, Appl. Environ. Microbiol. 71 (8) (2005) 4388–4399.
mSystems 4 (2) (2019), 00314-e00318. [113] J. Beutlich, S. Jahn, B. Malorny, E. Hauser, S. Huhn, A. Schroeter, et al.,
[92] J. Noster, T.-C. Chao, N. Sander, M. Schulte, T. Reuter, N. Hansmeier, M. Hensel, Antimicrobial resistance and virulence determinants in European Salmonella
Proteomics of intracellular Salmonella enterica reveals roles of Salmonella genomic island 1-positive Salmonella enterica isolates from different origins, Appl.
pathogenicity island 2 in metabolism and antioxidant defense, PLoS Pathog. 15 (4) Environ. Microbiol. 77 (16) (2011) 5655–5664.
(2019), e1007741. [114] F. Campioni, A.M. Moratto Bergamini, J.P. Falc~ao, Genetic diversity, virulence
[93] U.G. Barron, I. Soumpasis F. Butler, D. Prendergast, S. Duggan, G. Duffy, genes and antimicrobial resistance of Salmonella Enteritidis isolated from food and
Estimation of prevalence of Salmonella on pig carcasses and pork joints, using a humans over a 24-year period in Brazil, Food Microbiol. 32 (2) (2012) 254–264.
quantitative risk assessment model aided by meta-analysis, J. Food Protect. 72 (2) [115] R.E. Rowlands, C.A. Ristori, A.A. Ikuno, M.L. Barbosa, M. Jakabi, B.D. Franco,
(2009) 274–285. Prevalence of drug resistance and virulence features in Salmonella spp. isolated
[94] European Food Safety Authority EFSA Panel on Biological Hazards (BIOHAZ), from foods associated or not with salmonellosis in Brazil, Rev. Inst. Med. Trop. S~ao
Scientific opinion on a quantitative microbiological risk assessment of Salmonella Paulo 56 (6) (2014) 461–467.
in slaughter and breeder pigs, EFSA J. 8 (4) (2010) 1547. [116] S.D. Saroj, R. Shashidhar, M. Karani, J.R. Bandekar, Distribution of Salmonella
[95] N. Skovgaard, Risk assessments of Salmonella in eggs and broiler chickens: pathogenicity island (SPI)-8 and SPI-10 among different serotypes of Salmonella,
interpretative summary, Int. J. Food Microbiol. 91 (2) (2004) 223. J. Med. Microbiol. 57 (4) (2008) 424–427.
[96] H. Smadi, J.M. Sargeant, Quantitative risk assessment of human salmonellosis in [117] S. Chen, S. Zhao, P.F. McDermott, C.M. Schroeder, D.G. White, J. Meng, et al.,
Canadian broiler chicken breast from retail to consumption, Risk Anal. 33 (2) A DNA microarray for identification of virulence and antimicrobial resistance
(2013) 232–248. genes in Salmonella serovars and Escherichia coli, Mol. Cell. Probes 19 (3) (2005)
[97] S.M. Santillana Farakos, R. Pouillot, N. Anderson, R. Johnson, I. Son, J. van Doren, 195–201.
et al., Modeling the survival kinetics of Salmonella in tree nuts for use in risk [118] R. Biswas, R.K. Agarwal, K.N. Bielgaonkar, A. Kumar, P. Nambiar, S. Rawat,
assessment, Int. J. Food Microbiol. 227 (2016) 41–50. M. Singh, Cloning and sequencing of biofilm-associated protein (bapA) gene and
[98] S.O. Tromp, H. Rijgersberg, E. Franz, Quantitative microbial risk assessment for its occurrence in different serotypes of Salmonella, Lett. Appl. Microbiol. 52 (2)
Escherichia coli O157:H7, Salmonella enterica, and Listeria monocytogenes in leafy (2011) 138–143.

16

Das könnte Ihnen auch gefallen