You are on page 1of 9

Sensors International 1 (2020) 100028

Contents lists available at ScienceDirect

Sensors International
journal homepage: www.keaipublishing.com/en/journals/sensors-international

Detection of pathogenic bacteria with special emphasis to biosensors


integrated with AuNPs
Neelam Yadav a, b, *, Anil Kumar Chhillar b, Jogender Singh Rana a, **
a
Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonepat, Haryana, 131039, India
b
Centre for Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India

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

Keywords: Bacteria are unambiguously menacing for community healthiness globally. The common reservoirs of bacteria are
Bacterial infection contaminated foods, animals, polluted water and soil. There are several diseases causing bacteria such as Vibrio,
Nanotechnology Listeria, Yersinia, Salmonella, Shigella, Clostridium, Campylobacter and many more. The apprehension for the
AuNPs
transmission of bacterial infection has increased the incidence of outbreaks and several health issues across
Biosensors
Bacterial infections
boundaries. The key challenge for treatment of bacterial infection in early stage is the investigation of multiplexed
bacterial virulent factors. Therefore, it requisites the development of highly efficient techniques to identify,
prevent and manage growth of pathogenic bacteria for the well-being of living system. Conventional techniques
including serological tests, immuno assays, polymerase chain reaction (PCR) and isothermal microcalorimetry
(IMC) and many other techniques have been used for the testing of bacterial infection. However limitations of
aforesaid techniques have been overcome by biosensing techniques as they have offered numerous advantages
including highly simple, sensitive, specific, cost effective and quick response generating reproducible, on-site
detection and ease of portability. Present review article deciphers the role of distinct types of biosensors based
on gold nanoparticles for the analysis of bacteria from different reservoirs.

1. Introduction intestinal infections, renal disorders, central nervous disorders, arthritis,


and blindness [6]. Hence, diagnosis of bacterial pathogens has been
Foods are the natural and inevitable reservoirs of microbial popula- carried out by a number of conventional techniques including serological
tion [1]. Cooking of foods at high temperature kills the bacterial patho- tests, enzyme-linked immunosorbent assay (ELISA), PCR and IMC [7].
gens however; foods processed at mild heat contain huge population of Here we have discussed some of conventional approaches used in diag-
microbes which causes pathogenicity to human beings [1]. The chief nosis of bacterial infection.
bacterial reservoirs of food items include meat, dairy and poultry prod-
ucts. These food items have facilitated the growth of several microor- 2. Detection of bacterial pathogens by conventional methods
ganisms like Salmonella, Campylobacter, Listeria, and Escherichia coli
O157:H7 [2]. Therefore, such foodborne pathogens have increased the 2.1. Nucleic acid (NA) based methods
risk of pathogenicity. According to an estimate reported by World Health
Organization (WHO) revealed that approximately 93.8 million people These techniques detect the particular DNA/RNA segment of infect-
have been susceptible to foodborne diseases and about 155000 people ing bacteria. Zhao et al. [8] have revealed the investigation of targeted
mortality is due to foodborne pathogens annually [3–5]. The pathogenic DNA/RNA sequence by hybridizing nucleic acid sequence of specific
bacteria have caused several health complications such as repetitive bacterial pathogen with the complementary oligonucleotide segments

* Corresponding author. Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonepat, Haryana, 131039, India.
** Corresponding author. Department of Biotechnology, DCRUST, Murthal, Sonepat, India.
E-mail addresses: neelamindia12@rediffmail.com (N. Yadav), jsrana@outlook.com (J.S. Rana).

Production and hosting by Elsevier

https://doi.org/10.1016/j.sintl.2020.100028
Received 27 April 2020; Received in revised form 18 June 2020; Accepted 19 June 2020
Available online 5 August 2020
2666-3511/© 2020 The Authors. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd. This is an open access article under the CC BY-
NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
N. Yadav et al. Sensors International 1 (2020) 100028

that may be probes or primers. A number of bacteria namely Clostridium displace DNA stand during synthesis of DNA using Bst DNA polymerase at
botulinum, Vibrio cholerae, Staphylococcus aureus, and Escherichia coli high temperature (59–65  C). The time duration to complete one cycle is
(E. coli) O157 have been reported for production of toxins and thereby 60minutes. In LAMP, total 4 primers are used for the amplification of six
cause pathogenicity [9–11]. Thus, these pathogens can be detected by precise sites of target DNA. Law et al. reported the investigation of stxA2
using nucleic acid based techniques. Different types of PCR like multi- gene in E. coli O157:H7 [24]. The specificity of this technique was due to
plexed polymerase chain reaction (mPCR), real time PCR (RT-PCR), use of four distinct primers to target six different segments and conse-
nucleic acid sequence based amplification (NASBA) and loop mediated quently has provided fast multiplication, and low limit of detection than
isothermal amplification (LAMP) have been used for the investigation of PCR approach [26]. Mori and Notomi [25], have reported the bacterial
pathogenic bacteria. infection caused by distinct bacteria including Listeria, Salmonella,
Campylobacter, Legionella, and E. coli can be accessed by using commer-
2.1.1. Detection of bacterial pathogens using PCR cially available bacterial diagnostic kits [25].
PCR is widely used technique to analyse the samples with bacterial
pathogen. Velusamy et al. [12] have reported that PCR can detect single 2.1.5. Detection of bacterial pathogens using NASBA
bacteria by identifying NA sequence of target bacterial pathogen. In this This method is commonly used to amplify target RNA segment by
technique firstly bacterial NA sequence is converted into single stranded recruiting enzymatic machinery that comprises reverse transcriptase, T7
from double stranded. After that specific oligonucleotide segment i.e. RNA polymerase and RNaseH and avian myeloblastosis virus (AMV) [8].
primer binds complementary with the bacterial single stranded NA Law et al. [24] have used the application of NASBA for the detection of
sequence. The NA sequence of L. monocytogenes, E. coli O157:H7, several bacteria like S. enterica, V. cholerae, S. aureus, Campylobacter
S. aureus, Campylobacter jejuni, Salmonella spp. and Shigella spp have been jejuni, and Campylobacter coli. Nadal et al. have reported 400 CFU/mL
detected by PCR [13–15]. detection limit for the L. monocytogenes [27]. The reaction temperature of
this technique was approximately 41  C [8]. The products obtained from
2.1.2. Detection of bacterial pathogens using mPCR post-NASBA were detected by agarose gel electrophoresis ELISA. How-
This type of PCR involves multiple primers to amplify target DNA ever, these approaches were complicated, and expensive. Consequently,
sequence. The basic mechanism of amplification is similar to the simple Researchers developed an advanced approach of NASBA that is real-time
PCR [8]. Furthermore primers designing and their concentration used NASBA (RT-NASBA). This RT-NASBA requires oligonucleotides as a
during PCR reaction are very important parameters because very high probe (molecular beacons) that was fluorescently labelled probes as a
primers concentration may increase the interaction between the multiple result generated homogenous NASBA test [8]. Moreover, RT-NASBA has
oligonucleotides. mPCR has been used for the investigation of S. enteritidis, been used for the differentiation of viable cells from non-viable cells by
S. aureus, S. flexneri, L. monocytogenes, and E. coli O157:H7 [16]. Beside this, detecting the RNA of target cells and degraded the mRNA of scarified
Ryu et al. [17] have demonstrated the use of mPCR for the analysis of cells through RNase activity [28].
Listeria species namely L. monocytogenes, L. grayi, L. ivanovii, L. innocua, L.
welshimeri, and L. seeligeri [17]. They reported 7.58  104 copies detection 2.1.5.1. Disadvantages of NA based techniques. Though aforesaid NA
limit for mixed NA sequence of Listeria species. techniques were used for the sensitive detection of bacteria but these
Earlier mPCR had been employed for the investigation of only two or techniques exhibited a number of limitations such as complex procedure,
three microbes. Currently, this technique has been used for simultaneous expensive, less reliable, need large quantity of sample and lengthy
detection of more than five microorganisms. The advancement of mPCR procedure.
led to invention of another new technique that is GeXP-PCR [18]. This
GeXP-PCR was used to detect six bacteria viz. Salmonella enterica, Listeria
2.2. Detection of bacterial pathogens using culture based approaches
monocytogenes, Staphylococcus aureus, Escherichia coli O157:H7, Shigella
spp. and Campylobacter jejuni. The genome analysis by GeXP can detect
Identification of target infectious bacteria has also been monitored
several bacteria in only one reaction.
through culture based approaches. For illustration MacConkey agar
(SMAC) was used for the investigation of E. coli O157:H7 [29].
2.1.3. Detection of bacterial pathogens using qPCR
Furthermore, Tan et al. detected Y. enterocolitica and non Y. enterocolitica
In this type of PCR, the amplifying sequence can be visualized
on Cefsulodin-Irgasan-Novobiocin (CIN) agar media [30].
simultaneously during PCR reaction. The quantity of amplified PCR
product has been measured by monitoring fluorescence signals that de-
2.2.1. Disadvantages of culture based approaches
pends on concentration of PCR product [19]. The working of qPCR de-
The major disadvantages of culture based approaches are more time
pends on fluorescent systems. There are three main fluorescent systems
consuming due to slow growth of bacterial cells, less reliable as they
for qPCR include SYBR green, TaqMan probes and molecular beacons. It
sometimes give pseudo results and thus less sensitive and specific [31].
has been observed that low intensity fluorescence was emitted when
SYBR green intercalate non-specifically to major groove of
double-stranded DNA (dsDNA) [20]. Therefore, the intensity of fluores- 2.3. Detection of bacterial pathogens using ELISA
cence can be enhanced by binding of fluorescent dye in minor groove of
the DNA [21]. The other fluorescent system is TaqMan probes that are Detection of bacteria in the samples can be analysed by ELISA are
double stranded oligonucleotides consisting fluorescence emitting re- based antigen and antibody (Ab) interaction [24]. In these immunolog-
porter dye at the 50 end and quencher dye at 3'end [20]. These dyes are in ical assays primary Abs were confined on microtitre well plate followed
near vicinity to one another to inhibit the fluorescence emitted by re- by the binding of target bacteria to primary Abs. after that secondary Abs
porter [22]. Molecular beacons are oligonucleotide probes comprising bind with enzymes were bind to Ag. This binding produces coloured
sequence corresponding to specific segment and binding of this sequence signals that can be easily detected. ELISA has been reported for the
emit fluorescence [23]. This type of PCR has been used for the investi- investigation of bacteria like Vibrio parahaemolyticus [32] and Salmonella
gation of Salmonella, Vibrio parahaemolyticus and S. aureus [24]. The [33]. The detection limits of these bacteria were 103 cells and 1CFU/25 g
detection limits for these bacteria were 4 CFU/25 g, 102 CFU/mL and sample. Earlier reports suggest that sandwich ELISA can be employed for
1  104 CFU/mL respectively [24]. the investigation of a number of microorganisms responsible for causing
2.1.4. Detection of bacterial pathogens using LAMP foodborne disorders. Furthermore, ELISA test kit for instance BIOLINE
This approach was firstly employed by Notomi et al. [25] to analyse Salmonella ELISA Test was used for the evaluation of Salmonella in food
target bacteria. The working of this technique involves auto cycling to samples. The detection limit of this test kit was1CFU/25g sample with

2
N. Yadav et al. Sensors International 1 (2020) 100028

Fig. 1. Diagrammatic depiction of working and principle of a biosensor.

minimum four of the 20 food matrixes tested [33]. It has been reported features in Surface Plasmon Resonance (SPR) phenomenon as they have
that the bacterial toxins of C. perfringens α, β, and ε, s. enteroxins A, B, C, shown extraordinary absorption ability and reflecting cross sections. It
and E, botulinum toxins and E. coli enterotoxins had been detected by has been reported that AuNPs can be easily modified with immuno-
ELISA [8]. globulins, DNA/RNA probes, glycoproteins [48,49]. Nanoparticles have
been designed from Ag, ZnO, Co and many more, however these NPs
2.3.1. Disadvantages of ELISA have imparted toxicity to living cells their by restrict their use in in vivo
These immunoassays are time consuming, less sensitive and Abs are applications [50,51]. Therefore, composite NPs have gained much more
prone to degradation. attention in pharmaceutical applications. For instance, super-
However, these conventional approaches have several setbacks paramagnetic gold nanoshells have exhibited wide application applica-
including time-consuming, costly and need of highly trained person for tions in diagnostics, therapeutics, imaging and biosensing [52,53].
their operation. Therefore, it is necessary to find better alternatives to International Organization for Standardization (ISO) guidelines, the size
existing technologies that should be simple fast analysis of bacteria [34]. dimension of NPs ranges between 1 and 100 nm [50,54]. Due to various
Recently, scientific communities have shown their interest for favourable properties of AuNPs, they have been used in investigation of
nano-approaches by synthesizing nanoparticles. As these nanoparticles bacteria. For instance, bacteria namely Salmonella spp, Helicobacter pylori
have exhibited unique characteristics like small size, electronic, distance DNA and Mycobacterium tuberculosis have been detected colorimetrically
dependent colorimetric alterations and help in surface functionalization; using AuNPs [55,56]. Furthermore, biosensing devices based on modi-
and therefore employed in diverse sensing devices [35]. Consequently, fied AuNPs have analysed the bacteria by reducing their incubation time
they have been used to resolve the issues of conventional diagnostic and [57]. Use of biosensor technology has aroused the interest of scientists
therapeutic system [36,37]. In this review article we have described the because of their miniaturization, high specificity, sensitivity, ease in
characteristics features of gold (Au) nanoparticles (NPs) and their ap- transportation and automation [58], furthermore, requirement of small
plications in biosensing technology. amount of sample and reagent that make their use cost-effective [59].
Moreover, AuNPs based biosensors have also been developed for the
3. Properties of gold nanoparticles (AuNPs) on-site detection of analytes. For instance Fu et al. [60] have constructed
lateral flow biosensor for the in-field evaluation of brochilator compound
AuNPs have been widely used in research applications due to their known as clenbuterol used as an antagonist for pulmonary diseases and
flexibility in dimension as well as in configurations such as spherical, ingestion of this compound causes cardiac problems. Bu et al. [61] have
diamond, crystalline, triangular and spiral shapes [38,39]. Due to revealed the onsite detection of foodborne bacteria S. enteritidis. More-
nanosize dimension, AuNPs have exhibited unique properties of physical, over, Jiang et al. [62] reported the in-field investigation of E. coli
chemical like electrical, catalytical, optical and magnetic in comparison O157:H7 in the real samples.
to native Au. Consequently, AuNPs have been found more stable, resis- Researchers have developed AuNPs based biosensors for the investi-
tant at high temperature and can absorb and emit light just because of gation deadly diseases causing microorganisms [63]. It has been docu-
their optical properties [40]. It has been reported that AuNPs have also mented that AuNPs integrated biosensors have been fabricated for the
been used in impurity control for example air purification and respiratory investigation of microbial DNA sequences [64], proteins [65], enzymes
masks [40]. These AuNPs can bind with physiological components [66], and pathogens [67]. AuNPs have been used for the designing of
including peptides, proteins, DNA and biochemical polymers, complexes biosensing devices as they are easily soluble in water, and facile synthesis
and fusion materials that have been used in diverse medical applications procedures, compatibility in configuration, and also provide active site
[41]. AuNPs ranges from 10 to 100 nm are suitable for medical pro- due to surface functionalization [68]. Hence, biosensors have been found
spective while size larger than 10 nm are passed out via kidney and size suitable for the scrutinization of infectious bacteria [69]. For instance,
above 200 nm can be removed through body's immune system [42–44]. electrochemical nucleic acid biosensors have been reported for the
Furthermore, AuNPs have shown best optical absorbance and their investigation of bacteria/viruses found in contaminations of medical,
absorbance spectra can be fixed at precise wavelength in the near pharmaceutical and environment [70].
infrared region. Biosensing devices integrated with AuNPs have not
shown any harmful effect to living organism and help in fast detection of 4. Working and principle of biosensor
diseases causing microbes [45].
Applications of AuNPs have been reported in preparation of nano- A biosensor is a self-contained quantitative analytical approach that
medicines [46]. In the last fifty years, several reliable, reproducible and consists of biorecognition and transducing element. Significant signal
high yielding methods for their preparation have been developed [47]. generated after interaction of analytes and biorecognition element is
AuNPs have shown dimension dependent several fascinating properties detected by transducing component [71,72]. Biosensors are of different
and use of their minute concentration [48]. AuNPs have exhibited unique types depending on type of transducing components used [73,74]. The

3
N. Yadav et al. Sensors International 1 (2020) 100028

various types of biosensors include piezoelectric, optical, thermal, elec- by proportionate decline in frequency of oscillation. These analytical
trical, amperometric and potentiometric biosensors. Schematic repre- devices have been used for the investigation of several bacteria in various
sentation of working and principal of a biosensor has been depicted in food systems [79].
Fig. 1. Qiao et al. [6] have reported that complex of antimicrobial pep-
tides and AuNPs based electrochemical, optical and piezoelectric bio- 4.1.2.1. Investigation of bacterial pathogens by means of piezoelectric bio-
sensors have played a significant application to investigate several types sensors. Zheng et al. [80] designed piezoresistant biosensor based on
of foodborne pathogens. AuNPs. This biosensor was used for the investigation of pathogenic
bacteria viz. E. coli O157:H7, V. parahaemolyticus, Salmonella spp.,
4.1. Classification of biosensors S. aureus, L. monocytogenes and Shigella in foods, environment and clinical
samples. The response time of biosensor was 1 h with a limit of detection
On the basis of different types of transducers used, biosensors are of 1–9 cells/mL. Kaewphinit et al. [81] have reported the detection of
following different types: Mycobacterium tuberculosis using quartz crystal mass immobilized with
specific DNA probe for bacteria and AuNPs. The detection limit of this
4.1.1. Optical biosensors biosensor was 5 pg and exhibited 100% sensitivity. This aforesaid
These optical sensors employed for effective detection of foodborne biosensor revealed the sensitive, specific and cost-effective detection of
microorganisms [75]. The transducers used in such devices measure the M. tuberculosis. Furthermore, E. coli O157 was investigated by fabricating
analytical signals in the form of absorbance, fluorescence and reflectance. piezoelectric biosensor [62]. They developed the biosensor by inte-
Surface Plasmon Resonance (SPR) has been found most effective optical grating the Au electrode with AuNPs and single stranded DNA probe.
biosensing approach as it can investigate specific, rates of bacterial Modified Au electrode was bound to target DNA sequence of E. coli
infection detection and possible molecular interactions [76]. O157:H7 and their by helped in fast, facile, sensitive and specific
detection of bacteria. The above said piezoelectric biosensor exhibited
2.0  103 CFU/mL detection limit. Guo et al. [82] demonstrated the
4.1.1.1. Investigation of bacterial pathogens by means of optical bio-
development of piezoelectric immunosensor for the detection of E. coli
sensors. Corripio et al. [1] have detected the Salmonella bacteria by
O157:H7. They fabricate the biosensor by using quartz crystal micro-
fabricating optical biosensor integrated with fluorescently conjugated
balance immobilized with functionalized antibodies and AuNPs. They
Staphylococcal protein A at the surface of AuNPs which was further
reported the detection limit in the range of 0–1 log CFU/mL and 24 h
modified by using immunoglobulins labelled with fluorescein isothio-
response time. This biosensor was found effective, specific and quick
cyanate. The limit of detection of this biosensor was 103 and CFU/mL
detection of E. coli O157:H7 in food samples. One more study was
and 1 h response time. Moreover, Salmonella spp. was detected by con-
documented by Chen et al. [83] for the investigation of E. coli O157:H7.
structing optical biosensor using AuNPs [77]. The sensitivity of this
They fabricated AuNPs integrated circulating-flow piezoelectric
biosensor was <10 CFU/mL, response time less than 9 h, simple,
biosensor. For this investigation they used two probes; among them one
cost-effective and unique stability. Bu et al. [61] have reported the sen-
was specific for the target DNA segment of E. coli O157:H7 while other
sitive detection of S. enteritidis and E. coli O157 in water, lettuce and pork
was bound with target bacterial DNA sequence and AuNPs that acted as
samples. They investigated these two bacteria by developing AuNPs
mass enhancer. The detection limit showed by this biosensor was
based lateral flow strip. The detection limit of this sensor was about
1.2  102 CFU/mL and working range between 102 to 106 CFU/mL [83].
103 CFU/mL, sensitivity 104 CFU/mL and response time 10–15minutes.
Park et al. demonstrated the investigation of Salmonella typhimurium by
4.1.3. Colorimetric biosensors
optical ELISA [78].
Colorimetric biosensors have been employed for measuring the
quantity of coloured compounds in the evaluating sample. Investigation
4.1.2. Piezoelectric biosensors
of any analyte in the sample involves the detection of visual analytical
Piezoelectric biosensors measure the altered potential (voltage) of a
signal in the form of altering the colour of an analyte in the real sample
material which is exposed to mechanical strain. In these piezoelectric
and then analyse with the colour of standard compound [84]. Detection
biosensors, under the influence of an electrical field piezoelectric crystals
of analyte by these biosensors is simple, economic and need not required
vibrate at a specific frequency to produce electrical signal. Quartz Crystal
highly skilled person for the assessment of sample [85].
Microbalances (QCM) can sensitively detect small amounts of analytes in
the samples. Detection principal by these sensors involves the binding of
a piezoelectric sensor with a ligand like specific antibodies and followed 4.1.3.1. Investigation of bacterial pathogens by means of colorimetric bio-
by its dipping into a sample solution containing bacteria. The binding of sensors. Bu and co-workers [61] fabricated colorimetric biosensor to
any bacteria to the sensor ligand will increases the crystal mass followed investigate S. enteritidis by designing lateral flow strip integrated with

Table 1
Comparison of AuNPs integrated biosensors with highly efficient diagnostic techniques.
Name of detection method Target microorganisms Limit of Response time Reference
detection

PCR mPCR Listeria spp. 7.58  104 – [19]


copies
q-PCR S. enterica 18.6 fg/PCR 10 h [90]
L. monocytes 5 CFU/25 g <30 h [91]
LAMP Salmoneela spp. and Shigella spp. 5 CFU/10 mL <20 h [92]
NASBA E.coli, L. monocytes 40 CFU/mL 4h [93]
ELISA Salmonella 1 CFU/25 g – [33]
Optical biosensors integrated with AuNPs S.entericus, E. coli O157 103 CFU/mL 10–15minutes [61]
Piezoelectric biosensors integrated with E.coli O157:H7, V. parahaemolyticus, Salmonella spp., S. aureus, L. monocytogenes, 1-9 cells/mL 1h [80]
AuNPs Shigella
Colorimetric biosensors integrated with Shigella fexineri 80 CFU/mL 20minutes [35]
AuNPs
Electrochemical biosensors integrated with E. coli O157:H7 101CFU/mL 45minutes [94]
AuNPs

4
N. Yadav et al. Sensors International 1 (2020) 100028

Table 2
Assessment of different types of biosensor in terms of distinct analytical parameters.
Name of biosensor Electrode Limit of detection Name of microorganism Detection sample Response Name of nanomaterials Reference
used (LOD) used time

Electrochemical Pencil 30 CFU/mL Escherichia coli O157:H7 – – AuNPs [102]


(Immunosensor) graphite
electrode
Electrochemical Screen 3–9 pM Mycoplasma pneumoniae, – 30minutes AuNPs [103]
(Genosensor) printed Chlamydophila
electrode pneumoniae, Legionella
pneumophila, and
Streptococcus pneumonia
Electrochemical glassy 0.03 mM Mycobacterium tuberculosis Real samples 15minutes gold nanoparticles on the [104]
carbon and Neisseria meningitidis electropolymerized layer of
electrode poly-melamine
DNA electrochemical ITO 1.25 ng/mL Mycobacterium tuberculosis clinical sputum sample – AuNPs [105]
biosensor
Electrochemical Glassy 10.0 cfu/mL Bacillus cereus Contaminated milk 15minutes AuNPs and chitosan [106]
(Immunosensor) carbon
electrode
1
Optical (Genosensor) – 83.3 pg μL Enterococcus faecalis Detection in clinical 17minutes AuNPs [107]
samples
Electrochemical Screen 1.95  102 CFU/ Salmonella pullorum and Food products 30minutes AuNPs [65]
(Enzyme printed mL Salmonella gallinarum including eggs, chicken
immunosensor) electrode meats, chicken heart,
chicken liver, chicken
intestine
Colorimetric biosensor – 100 CFU/mL & Escherichia coli and – 1h Graphene oxide [108]
200 CFU/mL Staphylococcus aureus
Colorimetric – 1 nm mecA gene sequence of – 45 s Graphene quantum dots [109]
(Fluorescence Staphylococcus aureus (GQDs) and gold
resonance energy nanoparticles (AuNPs)
transfer (FRET)
biosensor)
Colorimetric biosensor – – E.coli O157:H7 Yellow corn samples – AuNPs [110]
Electrochemical bi- Screen- – Vibrio cholerae and Vibrio – 300s D-amino acid oxidase [111]
enzymatic biosensor printed parahaemolyticus (DAAO) and Horseradish
carbon peroxidase (HRP) enzymes
electrodes onto a MWCNTs and AuNPs
Electrochemical Glassy 3 CFU/mL Salmonella Pork samples 35minutes GO and AuNPs [89]
(Aptamer-based) carbon
electrode
impedimetric Modified Au 48 CFU/mL E. coli O157:H7 – – Self-assembled AuNPs [112]
biosensor electrodes
Colorimetric – 104 bacteria per Salmonella typhimurium – 15 min AuNPs [113]
mL
Electrochemical Glassy 0.028 nM Xanthomonas citri In real samples – AuNPs [114]
(Immunosensor) carbon
electrode

AuNPs in the drinking water, lettuce and pork samples. This biosensor AuNPs for the detection of Pseudomonas aeruginos [8]. This LFB exhibited
has exhibited 103CFU/mL low limit of detection. Colorimetric detection 10 fg and 40minutes detection limit and response time respectively. Ma
of Staphyloccocus aureus has been reported by using probes integrated et al. [89] have reported the analysis of Salmonella typhimurium in pork
with AuNPs [86]. They have reported the easy, fast, sensitive, specific samples by using AuNPs based surface enhanced raman scattering
and cost effective investigation of S. aureus in food and clinical samples. aptasensor. This aptasensor has shown 5 CFU/mL detection limit and
They reported 8.73 ng μL1 detection limit of S. aureus within 10–107CFU/mL. Comparison of various diagnostic techniques has been
10–15minutes. Niu et al. [87] have demonstrated the fabrication of depicted in Table 1.
colorimetric biosensor based on AuNPs for the analysis of Klebsiella
pneumoniae which causes nosocomial infection. They detected the 4.1.4. Electrochemical biosensors
aforesaid bacteria sensitively and specifically in clinical samples. This The transducers used in these devices measure the electrochemical
biosensor exhibited 10 fg detection limit and 25minutes response time. signal either in current or voltage [72,73]. The principal of measurement
Lateral flow biosensor (LFB) was fabricated by Wang et al. [88]. This LFB of these biosensing devices involves the specific chemical interaction
was based on AuNPs for the detection of marine sea foodborne bacteria between recognition element and binding analyte in the electrochemical
namely V. parahaemolyticus. They investigated the abovesaid pathogen in reaction system [74]. Depending on the type of electrochemical signals
clinical, food and environmental samples. This LFB has exhibited 10 fg produced, electrochemical biosensors have been classified into potenti-
detection limit with 65minutes response time. Furthermore, Feng et al. ometric, amperometric, conductometric, impedimetric systems of anal-
[35] have demonstrated the detection of food borne pathogenic bacteria ysis [95]. Amperometric biosensors measure current peaks due to
namely Shigella flexneri by fabricating aptasensor integrated with AuNPs. electrons flow during oxidation reduction reactions of working electrode
This colorimetric aptasensor was sensitive, specific and produces fast [96]. In addition to these amperometric biosensors, potentiometric bio-
response i.e. 20minutes. The aforesaid sensor has shown advantages like sensors can assess the potential/voltage on functional electrode in com-
simple, effective and ease of on-site bacterial detection. The above said parison to the auxiliary electrode [97]. Conductometric and
aptasensor has shown 80 CFU/mL detection limit and a wide working impedimetric devices during electrochemical reactions measure the
range i.e. 102 -106 CFU/mL. One more LFB has been designed using altered conductance of analytical solution or impedance of an

5
N. Yadav et al. Sensors International 1 (2020) 100028

Fig. 2. Diagrammatic depiction of challenges associated with AuNPs in clinical applications.

immobilized material. Principally the conductance or impedance of their characteristic analytical parameters has been shown in Table 2.
immobilized layer can be measured by using intermittent electric field
and thus assessing the resultant impedance magnitude for calculating the 4.1.4.2. Existing challenges associated with gold nanoparticles and their
system resistance [98]. solutions. Though, AuNPs have shown several unique characteristics that
revealed their numerous clinical applications [115]. However, there are
4.1.4.1. Investigation of bacterial pathogens by means of electrochemical several challenges that necessitate the researchers to think before using
biosensors. Electrochemical investigation of E. coli O157:H7 has been their applications for well-being of human beings. The significant issues
carried out by Wang and Alocilja [94], using AuNPs complexed with of AuNPs integrated tools and devices include reproducibility, reliability,
monoclonal antibodies which further conjugated with magnetic nano- stability and scalability during preparation [116]. In addition to these,
particles. This amperometric immunosensor exhibited 101CFU/mL limit AuNPs with specific dimensions and shapes can be prepared by using
of detection and 45minutes response time. Shoaie and co-workers [34] physical, chemical and biological methods. Physical methods of AuNPs
constructed an amperometric biosensor using screen printed electrode preparation include microwave and ultraviolet irradiation and laser
that was modified by immobilizing polyaniline and AuNPs. The signifi- ablation that have been found harmful. On the other hand, chemically
cance of this biosensor was to monitor E. coli in urine and contaminated AuNPs has been synthesis involves specific chemicals and solvents
water samples having 4 CFU detection limit. however; these chemical have caused several hazardous effects on living
Hajihosseini and co-workers [99] have revealed the differential pulse system. Beside chemicals they also need treacherous pH and temperature
voltammetry detection of Helicobacter pylori. They investigated the bac- that are not ideal [116,117]. Dimension of AuNPs has also greatly varied
teria confining graphene oxide and AuNPs onto glassy carbon electrode. with distinct clinical applications. AuNPs have shown problems of
They obtained 27.0 pM limit of detection and linear range was 60–600 species-specific differences with respect to transmission, pathophysio-
pM. Oliveira et al. [100] have impedimetrically detected the bacteria logic response, recognized as foreign by the man immune system and
namely E. coli, Serratia marcescens, Salmonella enterica and Klebsiella retention time [116,117]. Fig. 2 depicts the challenges associated with
pneumonia by identifying lipo polysaccharide (LPS). They used Au elec- AuNPs in clinical applications.
trode that was immobilized with CramoLL lectin on the PVM (poly (vinyl Therefore in view of aforesaid issues, researchers have resolved
chloride-vinylacetate maleic acid))–AuNpCy (L-cysteine-AuNPs). This certain issues like toxicity of AuNPs can be reduced by synthesizing
impedimetric biosensor detected abovesaid bacteria within 20minutes. nanoparticles from biological origins as biological methods are eco-
Zhu and co-workers [101] have constructed an electrochemical impedi- friendly in nature (Singh et al., 2018) [116,117]. Secondly, internaliza-
metric immunosensor using AuNPs. They investigated Clostridium difficile tion of AuNPs can be inhibited by the process of PEGylation (Singh et al.,
toxin A and B and obtained 0.61pg/mL and 0.60 pg/mL limit of detection 2018) [116,117]. By this process AuNPs are layered with poly-
respectively. ethyleneglycol that enhances the retention time of AuNPs in the circu-
Guner and co-workers [102] have fabricated an amperometric lation. Furthermore, future clinical research will help in better
immunosensor for the detection of E. coli O157:H7. This immunosensor understanding of the interactions of AuNPs with physiological compo-
was fabricated by functionalizing pencil graphite electrode with AuNPs, nents and hence certainly overcome the above said challenges.
multiwalled carbon nanotubes and polypyrrole. The immunosensor
exhibited detection limit 30 cfu/mL and wide linear range 5. Conclusion
3  101-3x107 CFU/mL. Paredes et al. [103] have designed genosensor
based on AuNPs immobilized onto screen printed electrode for the There are global serious concerns of human health from attack of
investigation of Mycoplasma pneumoniae, Legionella pneumophila, Chla- bacterial pathogens. Use of conventional techniques for detection of
mydophila pneumoniae and Streptococcus pneumonia. The detection limit of bacteria have some limitations like time taking, tedious, costly, need of
this genosensor for above said bacteria were 3 pM, 5 pM, 8 pM and 9 pM trained personnel, less reliable and reproducible. Hence, in this article we
respectively. The comparison between different types of biosensors with have reviewed distinct types of AuNPs biosensors for detection of

6
N. Yadav et al. Sensors International 1 (2020) 100028

bacterial pathogens. Due to fascinating properties of AuNPs, they have [18] B. Zhou, J. Xiao, S. Liu, J. Yang, Y. Wang, F. Nie, G. Zhao, Simultaneous detection
of six food-borne pathogens by multiplex PCR with a GeXP analyser, Food Contr.
been used in several clinical applications. Earlier reported publications
32 (1) (2013) 198–204.
suggest the significant applications of AuNPs based electrochemical [19] E. Omiccioli, G. Amagliani, G. Brandi, M. Magnani, A new platform for Real-Time
biosensors. These electrochemical biosensors have been found more PCR detection of Salmonella spp., Listeria monocytogenes and Escherichia coli
effective for the detection of pathogenic bacteria and also showed several O157 in milk, Food Microbiol. 26 (6) (2009) 615–622.
[20] I. Hein, A. Lehner, P. Rieck, K. Klein, E. Brandl, M. Wagner, Comparison of
other benefits including economic, highly sensitive, specific rapid and different approaches to quantify Staphylococcus aureus cells by real-time
ease of miniaturization. Moreover, optical biosensors have been used for quantitative PCR and application of this technique for examination of cheese,
the on-site detection of bacterial pathogen. In spite of having a number of Appl. Environ. Microbiol. 67 (7) (2001) 3122–3126.
[21] H. Fukushima, Y. Tsunomori, R. Seki, Duplex real-time SYBR green PCR assays for
developed biosensors for detection of food-borne pathogens, it is still a detection of 17 species of food-or waterborne pathogens in stools, J. Clin.
challenge for researchers to fabricate biosensors for the reliable and Microbiol. 41 (11) (2003) 5134–5146.
effective determination of microorganisms in real food samples. [22] R.E. Levin, The application of real-time PCR to food and agricultural systems. A
review, Food Biotechnol. 18 (1) (2004) 97–133.
[23] J.R. Patel, A.A. Bhagwat, G.C. Sanglay, M.B. Solomon, Rapid detection of
Declaration of competing interest Salmonella from hydrodynamic pressure-treated poultry using molecular beacon
real-time PCR, Food Microbiol. 23 (1) (2006) 39–46.
[24] J.W.F. Law, N.S. Ab Mutalib, K.G. Chan, L.H. Lee, Rapid methods for the detection
There is no conflict among authors of this paper. of foodborne bacterial pathogens: principles, applications, advantages and
limitations, Front. Microbiol. 5 (2015) 770.
Acknowledgement [25] Y. Mori, T. Notomi, Loop-mediated isothermal amplification (LAMP): a rapid,
accurate, and cost-effective diagnostic method for infectious diseases, J. Infect.
Chemother. 15 (2) (2009) 62–69.
Dr. Neelam thankfully acknowledges the UGC-Dr. D. S. Kothari Post- [26] Z. Xu, L. Li, J. Chu, B.M. Peters, M.L. Harris, B. Li, M.E. Shirtliff, Development and
Doctoral Fellowship (Reference id No. F.4-2/2006 (BSR)/BL/18-19/ application of loop-mediated isothermal amplification assays on rapid detection of
0107) for providing financial assistance. various types of staphylococci strains, Food Res. Int. 47 (2) (2012) 166–173.
[27] A. Nadal, A. Coll, N. Cook, M. Pla, A molecular beacon-based real time NASBA
assay for detection of Listeria monocytogenes in food products: role of target
References mRNA secondary structure on NASBA design, J. Microbiol. Methods 68 (3) (2007)
623–632.

[28] S.A. Simpkins, A.B. Chan, J. Hays, B.P. Opping, N. Cook, An RNA transcription-
[1] M.A. Rios-Corripio, L.S. Arcila-Lozano, B.E. Garcia-Perez, M.E. Jaramillo-Flores,
A.D. Hernandez-Perez, A. Carlos-Martinez, M. Rojas-L opez, Fluorescent gold based amplification technique (NASBA) for the detection of viable Salmonella
nanoparticle-based bioconjugate for the detection of Salmonella, Anal. Lett. 49 enterica, Lett. Appl. Microbiol. 30 (1) (2000) 75–79.
(12) (2016) 1862–1873. [29] J.J. Hirvonen, A. Siitonen, S.S. Kaukoranta, Usability and performance of
[2] B. Swaminathan, P. Feng, Rapid detection of food-borne pathogenic bacteria, CHROMagar STEC medium in detection of Shiga toxin-producing Escherichia coli
Annu. Rev. Microbiol. 48 (1) (1994) 401–426. strains, J. Clin. Microbiol. 50 (11) (2012) 3586–3590.
[3] M. Freitas, S. Viswanathan, H.P.A. Nouws, M.B.P.P. Oliveira, C. Delerue-Matos, [30] L.K. Tan, P.T. Ooi, E. Carniel, K.L. Thong, Evaluation of a modified cefsulodin-
Iron oxide/gold core/shell nanomagnetic probes and CdS biolabels for amplified irgasan-novobiocin agar for isolation of Yersinia spp, PloS One 9 (8) (2014).
electrochemical immunosensing of Salmonella typhimurium, Biosens. Bioelectron. [31] B. Priyanka, R.K. Patil, S. Dwarakanath, A review on detection methods used for
51 (2014) 195–200. foodborne pathogens, Indian J. Med. Res. 144 (3) (2016) 327.
[4] F. Salam, I.E. Tothill, Detection of Salmonella typhimurium using an [32] B.K. Kumar, P. Raghunath, D. Devegowda, V.K. Deekshit, M.N. Venugopal,
electrochemical immunosensor, Biosens. Bioelectron. 24 (8) (2009) 2630–2636. I. Karunasagar, I. Karunasagar, Development of monoclonal antibody based
[5] S.Y. Oh, N.S. Heo, S. Shukla, H.J. Cho, A.E. Vilian, J. Kim, Y.S. Huh, Development sandwich ELISA for the rapid detection of pathogenic Vibrio parahaemolyticus in
of gold nanoparticle-aptamer-based LSPR sensing chips for the rapid detection of seafood, Int. J. Food Microbiol. 145 (1) (2011) 244–249.
Salmonella typhimurium in pork meat, Sci. Rep. 7 (1) (2017) 1–10. [33] F.J. Bolton, E. Fritz, S. Poynton, T. Jensen, Rapid enzyme-linked immunoassay for
[6] Z. Qiao, Y. Fu, C. Lei, Y. Li, Advances in antimicrobial peptides-based biosensing detection of Salmonella in food and feed products: performance testing program,
methods for detection of foodborne pathogens: a review, Food Contr.. 107116, J. AOAC Int. 83 (2) (2000) 299–303.
112 (2020) 1-11. [34] N. Shoaie, M. Forouzandeh, K. Omidfar, Voltammetric determination of the
[7] M.S. Kumar, S. Ghosh, S. Nayak, A.P. Das, Recent advances in biosensor based Escherichia coli DNA using a screen-printed carbon electrode modified with
diagnosis of urinary tract infection, Biosens. Bioelectron. 80 (2016) 497–510. polyaniline and gold nanoparticles, Microchim. Acta 185 (4) (2018) 217.
[8] X. Zhao, C.W. Lin, J. Wang, D.H. Oh, Advances in rapid detection methods for [35] J. Feng, Q. Shen, J. Wu, Z. Dai, Y. Wang, Naked-eyes detection of Shigella flexneri
foodborne pathogens, J. Microbiol. Biotechnol. 24 (3) (2014) 297–312. in food samples based on a novel gold nanoparticle-based colorimetric aptasensor,
[9] D. Akbulut, K.A. Grant, J. McLauchlin, Development and application of real-time Food Control 98 (2019) 333–341.
PCR assays to detect fragments of the Clostridium botulinum types A, B, and E [36] T. Mocan, C. Matea, F. Tabaran, C. Iancu, R. Orasan, L. Mocan, In vitro
neurotoxin genes for investigation of human foodborne and infant botulism, administration of gold nanoparticles functionalized with MUC-1 protein fragment
Foodb. Pathog. Dis. 1 (4) (2004) 247–257. generates anticancer vaccine response via macrophage activation and polarization
[10] V. Fusco, G.M. Quero, M. Morea, G. Blaiotta, A. Visconti, Rapid and reliable mechanism, J. Canc. 6 (6) (2015) 583.
identification of Staphylococcus aureus harbouring the enterotoxin gene cluster [37] A. Llevot, D. Astruc, Applications of vectorized gold nanoparticles to the diagnosis
(egc) and quantitative detection in raw milk by real time PCR, Int. J. Food and therapy of cancer, Chem. Soc. Rev. 41 (1) (2012) 242–257.
Microbiol. 144 (3) (2011) 528–537. [38] M. Shafiei, A. Ghasemian, S.K. Mostafavi, M. Teimouri, H.R. Vardanjani,
[11] I. Son, R. Binet, A. Maounounen-Laasri, A. Lin, T.S. Hammack, J.A. Kase, Detection S.A. Mirforughi, Gold nanoparticle-based colorimetric platform technology as
of five Shiga toxin-producing Escherichia coli genes with multiplex PCR, Food rapid and efficient bacterial pathogens detection method from various sources,
Microbiol. 40 (2014) 31–40. Rev. Med. Microbiol. 30 (2) (2019) 128–132.
[12] V. Velusamy, K. Arshak, O. Korostynska, K. Oliwa, C. Adley, An overview of [39] Y. Zhang, B. Li, X. Chen, Simple and sensitive detection of dopamine in the
foodborne pathogen detection: in the perspective of biosensors, Biotechnol. Adv. presence of high concentration of ascorbic acid using gold nanoparticles as
28 (2) (2010) 232–254. colorimetric probes, Microchim. Acta 168 (1–2) (2010) 107–113.
[13] Y.K. Cheah, N.A. Salleh, L.H. Lee, S. Radu, S. Sukardi, J.H. Sim, Comparison of [40] F. Xia, X. Zuo, R. Yang, Y. Xiao, D. Kang, A. Vallee-Belisle, K.W. Plaxco,
PCR fingerprinting techniques for the discrimination of Salmonella enterica subsp. Colorimetric detection of DNA, small molecules, proteins, and ions using
enterica serovar Weltevreden isolated from indigenous vegetables in Malaysia, unmodified gold nanoparticles and conjugated polyelectrolytes, Proc. Natl. Acad.
World J. Microbiol. Biotechnol. 24 (3) (2008) 327. Sci. Unit. States Am. 107 (24) (2010) 10837–10841.
[14] L.H. Lee, Y.K. Cheah, A.S. Noorzaleha, S. Sabrina, J.H. Sim, C.H. Khoo, et al., [41] P. Gill, A.H. Alvandi, H. Abdul-Tehrani, M. Sadeghizadeh, Colorimetric detection
Analysis of Salmonella Agona and Salmonella Weltevredenin Malaysia by PCR of Helicobacter pylori DNA using isothermal helicase-dependent amplification and
finger printing and antibiotic resistance profiling, Antonie Leeuwenhoek 94 gold nanoparticle probes, Diagn. Microbiol. Infect. Dis. 62 (2) (2008) 119–124.
(2008) 377–387, https://doi.org/10.1007/s10482-008-9254-y. [42] Q. Xu, S. Du, H. Li, X.Y. Hu, Determination of acetamiprid by a colorimetric
[15] J. Alves, V.V. Marques, L.F.P. Pereira, E.Y. Hirooka, T.C.R.M. De Olivera, method based on the aggregation of gold nanoparticles, Microchim. Acta 173
Multiplex PCR for the detection of Campylobacter spp. and Salmonella spp. in (3–4) (2011) 323–329.
chicken meat, J. Food Saf. 32 (3) (2012) 345–350. [43] R.A. Sperling, P.R. Gil, F. Zhang, M. Zanella, W.J. Parak, Biological applications of
[16] J. Chen, J. Tang, J. Liu, Z. Cai, X. Bai, Development and evaluation of a multiplex gold nanoparticles, Chem. Soc. Rev. 37 (9) (2008) 1896–1908.
PCR for simultaneous detection of five foodborne pathogens, J. Appl. Microbiol. [44] K. Saha, S.S. Agasti, C. Kim, X. Li, V.M. Rotello, Gold nanoparticles in chemical
112 (4) (2012) 823–830. and biological sensing, Chem. Rev. 112 (5) (2012) 2739–2779.
[17] J. Ryu, S.H. Park, Y.S. Yeom, A. Shrivastav, S.H. Lee, Y.R. Kim, H.Y. Kim, [45] D. Pissuwan, C.H. Cortie, S.M. Valenzuela, M.B. Cortie, Functionalised gold
Simultaneous detection of Listeria species isolated from meat processed foods nanoparticles for controlling pathogenic bacteria, Trends Biotechnol. 28 (4)
using multiplex PCR, Food Contr. 32 (2) (2013) 659–664. (2010) 207–213.
[46] O.V. Salata, Applications of nanoparticles in biology and medicine,
J. Nanobiotechnol. 2 (1) (2004) 3.

7
N. Yadav et al. Sensors International 1 (2020) 100028

[47] D.A. Giljohann, D.S. Seferos, W.L. Daniel, M.D. Massich, P.C. Patel, C.A. Mirkin, [77] I. Quintela, B. de los Reyes, C.S. Lin, V.C. Wu, Simultaneous colorimetric detection
Gold nanoparticles for biology and medicine, Angew. Chem. Int. Ed. 49 (19) of a variety of Salmonella spp. in food and environmental samples by optical
(2010) 3280–3294. biosensing using oligonucleotide-gold nanoparticles, Front. Microbiol. 10 (2019)
[48] R. Wilson, The use of gold nanoparticles in diagnostics and detection, Chem. Soc. 1138.
Rev. 37 (9) (2008) 2028–2045. [78] M.K. Park, K.A. Weerakoon, J.H. Oh, B.A. Chin, The analytical comparison of
[49] S.H. Radwan, H.M. Azzazy, Gold nanoparticles for molecular diagnostics, Expert phage-based magnetoelastic biosensor with TaqMan-based quantitative PCR
Rev. Mol. Diagn 9 (5) (2009) 511–524. method to detect Salmonella Typhimurium on cantaloupes, Food Contr. 33 (2)
[50] M.A. Syed, S. Bokhari, Gold nanoparticle based microbial detection and (2013) 330–336.
identification, J. Biomed. Nanotechnol. 7 (2) (2011) 229–237. [79] L. Asensio, I. Gonzalez, T. García, R. Martín, Determination of food authenticity by
[51] K.L. Dreher, Health and environmental impact of nanotechnology: toxicological enzyme-linked immunosorbent assay (ELISA), Food Control 19 (1) (2008) 1–8.
assessment of manufactured nanoparticles, Toxicol. Sci. 77 (1) (2004) 3–5. [80] F. Zheng, P. Wang, N. Liu, Q. Du, Y. Chen, Simultaneous and ultra-sensitive
[52] Q.H. Lu, K.L. Yao, D. Xi, Z.L. Liu, X.P. Luo, Q. Ning, Synthesis and characterization detection of foodborne bacteria by gold nanoparticles-amplified microcantilever
of composite nanoparticles comprised of gold shell and magnetic core/cores, array biosensor, Front. Chem. 7 (2019) 232.
J. Magn. Magn Mater. 301 (1) (2006) 44–49. [81] T. Kaewphinit, S. Santiwatanakul, P. Jaratsing, K. Chansiri, N. Arunrut,
[53] X. Ji, R. Shao, A.M. Elliott, R.J. Stafford, E. Esparza-Coss, J.A. Bankson, C. Li, W. Kiatpathomchai, Detection of Mycobacterium tuberculosis by using loop-
Bifunctional gold nanoshells with a superparamagnetic iron oxide silica core mediated isothermal amplification combined with a lateral flow dipstick
suitable for both MR imaging and photothermal therapy, J. Phys. Chem. C 111 biosensor, in: The 4th 2011 Biomedical Engineering International Conference,
(17) (2007) 6245–6251. IEEE, 2012, January, pp. 86–88.
[54] S. Cui, S. Zhou, C. Chen, T. Qi, C. Zhang, J. Oh, A simple and rapid [82] X. Guo, C.S. Lin, S.H. Chen, R. Ye, V.C. Wu, A piezoelectric immunosensor for
immunochromatographic strip test for detecting antibody to porcine reproductive specific capture and enrichment of viable pathogens by quartz crystal
and respiratory syndrome virus, J. Virol Methods 152 (1–2) (2008) 38–42. microbalance sensor, followed by detection with antibody-functionalized gold
[55] S. Nakamura, N. Maeda, I.M. Miron, M. Yoh, K. Izutsu, C. Kataoka, Y. Hayashizaki, nanoparticles, Biosens. Bioelectron. 38 (1) (2012) 177–183.
Metagenomic diagnosis of bacterial infections, Emerg. Infect. Dis. 14 (11) (2008) [83] H.T. Chen, J. Zhang, D.H. Sun, L.N. Ma, X.T. Liu, X.P. Cai, Y.S. Liu, Development of
1784. reverse transcription loop-mediated isothermal amplification for rapid detection
[56] C. Kaittanis, S. Santra, J.M. Perez, Emerging nanotechnology-based strategies for of H9 avian influenza virus, J. Virol Methods 151 (2) (2008) 200–203.
the identification of microbial pathogenesis, Adv. Drug Deliv. Rev. 62 (4–5) [84] N. Anzar, M.R. Hasan, M. Akram, N. Yadav, J. Narang, Systematic and validated
(2010) 408–423. techniques for the detection of ovarian cancer emphasizing the electro-analytical
[57] J.W. Choi, B.K. Oh, Optical detection of pathogens using protein chip, in: approach, Process Biochem. 94 (2020) 126–135.
Advanced Environmental Monitoring, Springer, Dordrecht, 2008, pp. 348–362. [85] P. Sharma, A. Panchal, N. Yadav, J. Narang, Analytical techniques for the
[58] F. Malvano, R. Pilloton, D. Albanese, Label-free impedimetric biosensors for the detection of glycated haemoglobin underlining the sensors, Int. J. Biol. Macromol.
control of food safety–a review, Int. J. Environ. Anal. Chem. (2019) 1–24. 155 (2020) 685–696.
[59] E.B. Setterington, E.C. Alocilja, Electrochemical biosensor for rapid and sensitive [86] R. Shahbazi, M. Salouti, B. Amini, A. Jalilvand, E. Naderlou, A. Amini, A. Shams,
detection of magnetically extracted bacterial pathogens, Biosensors 2 (1) (2012) Highly selective and sensitive detection of Staphylococcus aureus with gold
15–31. nanoparticle-based core-shell nano biosensor, Mol. Cell. Probes 41 (2018) 8–13.
[60] Q. Fu, J. Liang, C. Lan, K. Zhou, C. Shi, Y. Tang, Development of a novel dual- [87] L. Niu, F. Zhao, J. Chen, J. Nong, C. Wang, J. Wang, S. Hu, Isothermal
functional lateral-flow sensor for on-site detection of small molecule analytes, amplification and rapid detection of Klebsiella pneumoniae based on the multiple
Sensor. Actuator. B Chem. 203 (2014) 683–689. cross displacement amplification (MCDA) and gold nanoparticle lateral flow
[61] T. Bu, P. Jia, J. Liu, Y. Liu, X. Sun, M. Zhang, L. Wang, Diversely positive-charged biosensor (LFB), PloS One 13 (10) (2018).
gold nanoparticles based biosensor: a label-free and sensitive tool for foodborne [88] Y. Wang, H. Li, D. Li, K. Li, Y. Wang, J. Xu, C. Ye, Multiple cross displacement
pathogen detection. Food chemistry: X, 3, 100052.thogens, J. Biol. Eng. 9 (1) amplification combined with gold nanoparticle-based lateral flow biosensor for
(2019) 16. detection of Vibrio parahaemolyticus, Front. Microbiol. 7 (2016) 2047.
[62] Y. Jiang, Z. Zhou, Y. Qian, Z. Wei, Y. Yu, S. Hu, L. Li, Plasmid-mediated quinolone [89] X. Ma, Y. Jiang, F. Jia, Y. Yu, J. Chen, Z. Wang, An aptamer-based electrochemical
resistance determinants qnr and aac (60 )-Ib-cr in extended-spectrum β-lactamase- biosensor for the detection of Salmonella, J. Microbiol. Methods 98 (2014) 94–98.
producing Escherichia coli and Klebsiella pneumoniae in China, J. Antimicrob. [90] J. Chen, L. Zhang, G.C. Paoli, C. Shi, S.I. Tu, X. Shi, A real-time PCR method for the
Chemother. 61 (5) (2008) 1003–1006. detection of Salmonella enterica from food using a target sequence identified by
[63] K. Balasubramanian, M. Burghard, Biosensors based on carbon nanotubes, Anal. comparative genomic analysis, Int. J. Food Microbiol. 137 (2–3) (2010) 168–174.
Bioanal. Chem. 385 (3) (2006) 452–468. [91] O. Ruiz-Rueda, M. Soler, L. Calv o, J.L. García-Gil, Multiplex real-time PCR for the
[64] I.K. Litos, P.C. Ioannou, T.K. Christopoulos, J. Traeger-Synodinos, E. Kanavakis, simultaneous detection of Salmonella spp. and Listeria monocytogenes in food
Multianalyte, dipstick-type, nanoparticle-based DNA biosensor for visual samples, Food Anal. Methods 4 (2) (2011) 131–138.
genotyping of single-nucleotide polymorphisms, Biosens. Bioelectron. 24 (10) [92] X. Ji, R. Shao, A.M. Elliott, R.J. Stafford, E. Esparza-Coss, J.A. Bankson, C. Li,
(2009) 3135–3139. Bifunctional gold nanoshells with a superparamagnetic iron oxide silica core
[65] C. Hu, W. Dou, G. Zhao, Enzyme immunosensor based on gold nanoparticles suitable for both MR imaging and photothermal therapy, J. Phys. Chem. C 111
electroposition and Streptavidin-biotin system for detection of S. pullorum & S. (17) (2007) 6245–6251.
gallinarum, Electrochim. Acta 117 (2014) 239–245. [93] A.J. Baeumner Min, Highly sensitive and specific detection of viable Escherichia
[66] S. Hong, I. Choi, S. Lee, Y.I. Yang, T. Kang, J. Yi, Sensitive and colorimetric coli in drinking water, Anal. Biochem. 303 (2) (2002) 186–193.
detection of the structural evolution of superoxide dismutase with gold [94] Y. Wang, E.C. Alocilja, Gold nanoparticle-labeled biosensor for rapid and sensitive
nanoparticles, Anal. Chem. 81 (4) (2009) 1378–1382. detection of bacterial pathogens 9 (2015) 1–7.
[67] Y. Liu, R. Brando, M. Cate, X. Peng, R. Stony, M. Johnson, Detection of pathogens [95] M. Wang, J. Yang, Z. Gai, S. Huo, J. Zhu, J. Li, L. Zhang, Comparison between
using luminescent CdSe/ZnS dendron nanocrystals and a porous membrane digital PCR and real-time PCR in detection of Salmonella typhimurium in milk,
immunofilter, Anal. Chem. 79 (22) (2007) 8796–8802. Int. J. Food Microbiol. 266 (2018) 251–256.
[68] P. Baptista, E. Pereira, P. Eaton, G. Doria, A. Miranda, I. Gomes, R. Franco, Gold [96] D.H. Kim, H.W. Lim, S.H. Kim, K.H. Seo, Development of a real-time PCR assay for
nanoparticles for the development of clinical diagnosis methods, Anal. Bioanal. rapid screening of acetic acid bacteria as a group in food products, Food Contr.
Chem. 391 (3) (2008) 943–950. 100 (2019) 78–82.
[69] Y. Chen, Y. Li, Y. Yang, F. Wu, J. Cao, L. Bai, A polyaniline-reduced graphene [97] I.H. Cho, J. Irudayaraj, In-situ immuno-gold nanoparticle network ELISA
oxide nanocomposite as a redox nanoprobe in a voltammetric DNA biosensor for biosensors for pathogen detection, Int. J. Food Microbiol. 164 (1) (2013) 70–75.
Mycobacterium tuberculosis, Microchim. Acta 184 (6) (2017) 1801–1808. [98] B. Pang, C. Zhao, L. Li, X. Song, K. Xu, J. Wang, X. Meng, Development of a low-
[70] M. Azimzadeh, M. Rahaie, N. Nasirizadeh, M. Daneshpour, H. Naderi-Manesh, cost paper-based ELISA method for rapid Escherichia coli O157: H7 detection,
Electrochemical miRNA biosensors: the benefits of nanotechnology, Nanomed. Anal. Biochem. 542 (2018) 58–62.
Res. J. 2 (1) (2017) 36–48. [99] S. Hajihosseini, N. Nasirizadeh, M.S. Hejazi, P. Yaghmaei, A sensitive DNA
[71] K.E. Mach, P.K. Wong, J.C. Liao, Biosensor diagnosis of urinary tract infections: a biosensor fabricated from gold nanoparticles and graphene oxide on a glassy
path to better treatment, Trends Pharmacol. Sci. 32 (6) (2011) 330–336. carbon electrode, Mater. Sci. Eng. C 61 (2016) 506–515.
[72] A.K. Chhillar Neelam, J.S. Rana, Enzyme nanoparticles and their biosensing [100] M.D. Oliveira, C.A. Andrade, M.T. Correia, L.C. Coelho, P.R. Singh, X. Zeng,
applications: a review, Anal. Biochem. 581 (2019) 113345, https://doi.org/ Impedimetric biosensor based on self-assembled hybrid cystein-gold nanoparticles
10.1016/j.ab.2019.113345. and CramoLL lectin for bacterial lipopolysaccharide recognition, J. Colloid
[73] C.S. Pundir, N. Yadav, A.K. Chhillar, Occurrence, synthesis, toxicity and detection Interface Sci. 362 (1) (2011) 194–201.
methods for acrylamide determination in processed foods with special reference to [101] Z. Zhu, L. Shi, H. Feng, H.S. Zhou, Single domain antibody coated gold
biosensors: a review, Trends Food Sci. Technol. 85 (2019) 211–225. nanoparticles as enhancer for Clostridium difficile toxin detection by
[74] N. Yadav, A.K. Chhillar, C.S. Pundir, Preparation, characterization and application electrochemical impedance immunosensors, Bioelectrochemistry 101 (2015)
of haemoglobin nanoparticles for detection of acrylamide in processed foods, Int. 153–158.
J. Biol. Macromol. 107 (2018) 1000–1013. [102] A. Güner, E. Çevik, M. Şenel, L. Alpsoy, An electrochemical immunosensor for
[75] P. Elizaquível, R. Aznar, G. Sanchez, Recent developments in the use of viability sensitive detection of Escherichia coli O157: H7 by using chitosan, MWCNT,
dyes and quantitative PCR in the food microbiology field, J.Appl. Microb. 116 (1) polypyrrole with gold nanoparticles hybrid sensing platform, Food Chem. 229
(2014) 1–13. (2017) 358–365.
[76] J. Riu, B. Giussani, Electrochemical biosensors for the detection of pathogenic [103] G. Martínez-Paredes, M.B. Gonzalez-García, A. Costa-García, Genosensor for SARS
bacteria in food, Trac. Trends Anal. Chem. 126 (2020) 115863. virus detection based on gold nanostructured screen-printed carbon electrodes,

8
N. Yadav et al. Sensors International 1 (2020) 100028

Electroanalysis: Int. J. Devoted Fund. Pract. Asp. Electroanalysis 21 (3-5) (2009) [111] M. Moreno-Guzman, L. García-Carmona, A.  Molinero-Fernandez, F. Cava,
379–385.  Gil, A. Escarpa, Bi-enzymatic biosensor for on-site, fast and reliable
M.A.L.
[104] S. Amidi, Y.H. Ardakani, M. Amiri-Aref, E. Ranjbari, Z. Sepehri, H. Bagheri, electrochemical detection of relevant D-amino acids in bacterial samples, Sensor.
Sensitive electrochemical determination of rifampicin using gold nanoparticles/ Actuator. B Chem. 242 (2017) 95–101.
poly-melamine nanocomposite, RSC Adv. 7 (64) (2017) 40111–40118. [112] D. Lin, R.G. Pillai, W.E. Lee, A.B. Jemere, An impedimetric biosensor for E. coli
[105] C. Thiruppathiraja, S. Kamatchiammal, P. Adaikkappan, D.J. Santhosh, M. Alagar, O157: H7 based on the use of self-assembled gold nanoparticles and protein G,,
Specific detection of Mycobacterium sp. genomic DNA using dual labeled gold Microchim. Acta 186 (3) (2019) 169.
nanoparticle based electrochemical biosensor, Anal. Biochem. 417 (1) (2011) [113] S. Wang, A.K. Singh, D. Senapati, A. Neely, H. Yu, P.C. Ray, Rapid colorimetric
73–79. identification and targeted photothermal lysis of Salmonella bacteria by using
[106] X. Kang, G. Pang, Q. Chen, X. Liang, Fabrication of Bacillus cereus electrochemical bioconjugated oval-shaped gold nanoparticles, Chem. Eur J. 16 (19) (2010)
immunosensor based on double-layer gold nanoparticles and chitosan, Sensor. 5600–5606.
Actuator. B Chem. 177 (2013) 1010–1016. [114] H. Haji-Hashemi, M.M. Habibi, M.R. Safarnejad, P. Norouzi, M.R. Ganjali, Label-
[107] G.H. Tondro, R.D. Vais, N. Sattarahmady, An optical genosensor for Enterococcus free electrochemical immunosensor based on electrodeposited Prussian blue and
faecalis using conjugated gold nanoparticles-rRNA oligonucleotide, Sensor. gold nanoparticles for sensitive detection of citrus bacterial canker disease, Sensor.
Actuator. B Chem. 263 (2018) 36–42. Actuator. B Chem. 275 (2018) 61–68.
[108] J. Li, L.J. Wu, S.S. Guo, H.E. Fu, G.N. Chen, H.H. Yang, Simple colorimetric [115] S. Zeng, K.T. Yong, I. Roy, X.Q. Dinh, X. Yu, F. Luan, A review on functionalized
bacterial detection and high-throughput drug screening based on a gold nanoparticles for biosensing applications, Plasmonics 6 (3) (2011) 491.
graphene–enzyme complex, Nanoscale 5 (2) (2013) 619–623. [116] P. Singh, S. Ahn, J.P. Kang, S. Veronika, Y. Huo, H. Singh, D.C. Yang, In vitro anti-
[109] J. Shi, C. Chan, Y. Pang, W. Ye, F. Tian, J. Lyu, M. Yang, A fluorescence resonance inflammatory activity of spherical silver nanoparticles and monodisperse
energy transfer (FRET) biosensor based on graphene quantum dots (GQDs) and hexagonal gold nanoparticles by fruit extract of Prunus serrulata: a green synthetic
gold nanoparticles (AuNPs) for the detection of mecA gene sequence of approach, Artif. Cell. Nanomed. Biotechnol. 46 (8) (2018) 2022–2032.
Staphylococcus aureus, Biosens. Bioelectron. 67 (2015) 595–600. [117] P. Singh, S. Pandit, V.R.S.S. Mokkapati, A. Garg, V. Ravikumar, I. Mijakovic, Gold
[110] M.A. Ali, T.A.S. Eldin, G.E. Moghazy, I.M. Tork, I.I. Omara, I. I. Detection of E. Coli nanoparticles in diagnostics and therapeutics for human cancer, Int. J. Mol. Sci. 19
O157: H7 in feed samples using gold nanoparticles sensor, Int. J. Curr. Microbiol. (7) (2018) 1979.
App. Sci 3 (6) (2014) 697–708.

You might also like