Вы находитесь на странице: 1из 29

Document type: Review/Account

Award Account
Account/Review for Materials Innovation

Recent Progress on the Sensing of Pathogenic Bacteria


Using Advanced Nanostructures

Gopalan Sai-Anand,*1,2,³ Arumugam Sivanesan,2,3,³ Mercy R Benzigar,2 Gurwinder Singh,1,2


Anantha-Iyengar Gopalan,4 Arun Vijay Baskar,1,2 Hamid Ilbeygi,2 Kavitha Ramadass,1,2
Venkata Kambala,5 and Ajayan Vinu*1,2

1
Global Innovative Center for Advanced Nanomaterials, Faculty of Built Environment and Engineering,
The University of Newcastle, Callaghan 2308, New South Wales, Australia
2
Future Industries Institute, Division of Information Technology, Engineering and Environment,
University of South Australia, Mawson Lakes, South Australia 5095, Australia
3
Metrohm Australia, 56 Buffalo Road, Gladesville, NSW 2111, Australia
4
Research Institute of Advanced Energy Technology, Kyungpook National University, Daegu 41566, Korea
5
Hudson Marketing Pty Ltd, Level 2/131 Macquarie St, Sydney NSW 2000, Australia
E-mail: Ajayan.Vinu@newcastle.edu.au

Received: September 14, 2018; Accepted: October 3, 2018; Web Released: October 27, 2018

Ajayan Vinu
Prof. Ajayan Vinu is a Professor of Nanomaterials and Director at Global Innovative Centre for Advanced
Nanomaterials (GICAN), The University of Newcastle. He was working as a full professor and Australian
Research Council Future Fellow at the University of Queensland and University of South Australia. Prior
coming to Australia, he had been working as a senior researcher and research group leader at the National
Institute for Materials Science (NIMS), Tsukuba, Japan since 2006 after he successfully completed two years
of the ICYS fellowship at the same institute and a few years of research at the Technical University of
Kaiserslautern (TUK), Germany. During these 15 years of research, Prof. Vinu has made a tremendous
contribution in the field of nanoporous materials and their application in sensing, energy storage, fuel cells,
adsorption and separation, and catalysis. His contribution in the field of nanoporous materials is also clearly
reflected by his international ranking by Science Watch as one of the top 15 researchers in the field and has
led to 350 original research papers in high impact factor journals with ca. 16,600 citations and an H-index
of 67.

Gopalan Sai-Anand
Dr. Sai-Anand Gopalan is currently working as a Research Associate in Global Innovative Center for
Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle,
Callaghan Campus, New South Wales. His recent research focus is on the design and development of novel
functionalized porous materials and their widespread applications in sustainable energy technologies such
as fuel cells and supercapacitors with a special emphasis on organic optoelectronic materials and devices to
address global energy demands and environmental challenges.

Abstract tious to human beings owing to their appearance in food, water,


Ailment related to pathogenic bacteria and toxins remains a and other biological samples. Over the past several years,
significant threat to the human body. Specifically, pathogenic advanced nanomaterials-based sensing has been considered
bacteria are the main source of epidemic diseases and are infec- as an efficient and unique platform for the rapid, selective,

216 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
ultrasensitive, qualitative, and quantitative detection of single analytical methods such as polymerase chain reaction (PCR),
or multiple pathogenic bacteria. Towards this end, various colony count, enzyme linked immunosorbent assay (ELISA),
emerging nanomaterials have been purposefully designed and biosensors, electrophoresis etc. have been employed for the
developed to integrate them onto sensor systems for the recog- detection of these pathogens. The most popular methods for
nition of pathogenic bacteria. The present review describes a detection include culture and colony counting7 and PCR.8
wide range of analytical techniques such as surface-enhanced Conventional methods such as surface recognition, nucleic
Raman scattering, electrochemistry (electrochemical and elec- acid detection and enzyme mediated are quite expensive as they
tronic), a field-effect transistor, fluorescence, calorimetry and require a high level sophistication and complex sample prepa-
surface-plasmon resonance etc. which incorporate nano- ration even though they are quite sensitive. Therefore, new
biosensor technology to develop a pathogenic bacterium based methods are required to improve their performance. Under
sensor. This review also highlights the progress, trends and these circumstances there is a growing demand for the design
strategy utilized toward the identification of harmful bacteria and development of ultrasensitive, rapid, and selective patho-
by focusing on the pertinent literature available on the various gen detection methods capable of detecting very a few or a
advanced nanomaterials (such as semiconducting, magnetic, single pathogenic bacterial species in target samples (water,
noble metal and carbon-based nanomaterials) incorporating food or biological tissues). The recent advancement in the
nano-bio sensor platforms. biosensor technology promises access to highly selective,
reliable, and rapid pathogenic detection.
Keywords: Pathogenic microorganism j Biosensor j Since the invention of the first biosensor (glucose biosensor)
Nanomaterial by L.C. Clark in 1956, sensing based on biotechnology has
emerged as an alternative to expensive chromatographic meth-
ods for the analysis of a variety of analytes. This is mainly due
1. Introduction to easy operation as it involves only the conversion of a
Biocontamination and communicable ailments (generally biological response into a detectable signal. A typical biosensor
referred as infections or transmittable disease) are an existing comprises two parts, namely a bio-receptor and a physico-
global threat leading to millions of deaths every year.1­3 chemical transducer, or combined into a single sensor called
Infectious diseases are generally caused by several different a bio-transducer, a recognition-transduction component. Both
classes of pathogenic microorganisms such as viruses, bacteria, the bio-receptor or bio-recognition component and transducer
protozoa, fungi, and parasites. Among the pathogenic micro- cooperate and convert a biochemical signal into to an electrical,
organisms, bacterial infections alone account for one-third of electrochemical, optical, piezoelectric, pyroelectric, thermal,
global mortality.4 Diarrhoeal diseases caused by a variety of gravimetric or other detection signal.9 Biosensors have been
bacterial organism are the second leading cause of death in considered prominent analytical devices incorporating a bio-
children under 5 years. Typically, each year diarrhoea kills logical material (e.g., tissue, microorganism, organelle, cell
around ³520,000 children. Tuberculosis (TB), is another receptor, enzyme, etc.) or a biologically derived material (e.g.,
bacterial disease found in almost every country in the world, recombinant antibodies, engineered proteins, aptamers, etc.) or
caused by Mycobacterium tuberculosis (M. tb), a pathogenic a bio-mimic system (e.g., combinatorial ligands, and imprinted
bacterium in the family Mycobacteriaceae. TB causes ill- polymers).10 The bio-receptor usually contains an antibody or
health for nearly 10 million people each year and is recog- aptamers or enzymes/proteins capable of selectively binding
nised as one of the top ten causes of death worldwide. Some of the target molecule. The physicochemical transducer, which is
the other high mortality causing bacteria are Streptococcus in direct contact with the bio-receptor, produces a physico-
pneumoniae, Vibrio cholera, Staphylococcus aureus, Helico- chemical change when there is a biochemical interaction
bacter pylori, Salmonella typhi and Escherichia Coli (E. coli). between the bio-receptor and biomolecule. The continuous
The bacterial diseases such as anthrax, relapsing fever, bru- investigation of various aspects of biosensors has led to several
cellosis, clostridal, plague tularaemia etc. originate from the innovations which have been applied to bacterial and viral
respective causative microorganisms and are distributed world- detection and monitoring. With advancements in nanotechnol-
wide. Studies on the lung bacterial microbiome have unfolded ogy and nanoscience, advanced nanomaterial-based biosensors
the relationship between lung microbiota and acetate respi- have shown great potential in improving both sensitivity and
ratory diseases (ARD).5 Importantly, ARD is a cause of mor- selectivity.
tality, leading to the death of 4 million people every year A variety of advanced nanomaterials such as metallic nano-
globally. The distribution of ARD depends on factors such as particles (MNP), carbon based nanomaterials, magnetic nano-
bacterial infections, environmental conditions and infection particles (MP) and quantum dots (QDs) are continuously being
prevention measures.6 utilized in biosensor design and other applications due to their
There are two important aspects which need to be considered unique physical, chemical, catalytic, sensing, mechanical,
inn controlling pathogenic bacterial infection. Firstly, bacte- magnetic, and optical properties.11­24 These properties of the
rial pathogen, such as Enterrohemorrhagic Escherichia Coli nanomaterials are beneficial to enhance the sensitivity and spe-
(EHEC) can produce toxins. Secondly, a small number of the cificity of the fabricated biosensors (Scheme 1). Most impor-
pathogenic bacteria are sufficient to initiate infection in the tantly, these technological advancements provide scope for
body and can cause potential damage to the host system. tuning the properties of nanomaterials by changing the size,
Development of detection strategies for different kinds of shape, morphology, surface charge, surface area and composi-
pathogens is an important aspect of health and safety. Different tion, modifying various adsorption capacities with appropriate

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 217
ing, pathogen detection and other medicinal applications (such
as cancer cell imaging, DNA marking etc.) thus receives a great
deal of attention and has produces a large quantity of pub-
lished literature.32­35 This can be witnessed by several review
articles that present the developments of biosensors based on
the nanomaterials for various pathogens including food borne
bacterial pathogens and water borne pathogens.27,36,37 Recently,
a surge of research activity can also be witnessed through
the literature on pathogen biosensors employing advanced
nanomaterials.
The main goal of this review is to provide a comprehensive
outline of the literature from the year 2013 to 2018 (a five-year
period) for ongoing and upcoming researchers in this area as
well as new researchers who want to explore this active
research area of utilizing nanomaterials for the fabrication of
efficient biosensors toward pathogen detection. This review has
several focused subsections that highlight the utility of indi-
vidual nanomaterials (such as gold nanoparticles, quantum dots
etc.) towards the construction of biosensors for bacterial
Scheme 1. Scheme showing the detection of pathogens pathogens. It also discusses a broad range of analytical tech-
employing advanced nanomaterials by various analytical niques such as surface-enhanced Raman scattering (SERS),
techniques. electrochemistry, a field-effect transistor (FET), fluorescence,
calorimetry and surface-plasmon resonance (SPR) which are
functionalization using surface chemistry by reducing sensing being utilized in nano-biosensor technology to develop novel
time and multiplexing capability. Integration of these function- pathogenic bacteria sensors (Scheme 1).
alities in nano-biosensor devices has significantly enhanced the
sensitivity and versatility of the sensors.25,26 In a typical nano- 2. Semiconductor Nanocrystals
biosensor device, the transducer is made of a nanosized mate- Taking advantage of the developments in semiconductor
rial, i.e., nanotransducer which is then coupled to a biorecog- technology and materials chemistry of surface conjugation with
nition moiety such as an antibody or aptamer. The general pathogen detecting biomarkers, various semiconducting mate-
mechanism of optical, electrochemical and magnetic transduc- rials such as metal sulphides, metal oxides, etc. and organic
tion modes based on nanomaterials has been clearly detailed in semiconductors are used for pathogen detections. Nanostruc-
a review published in 2010.27 tured semiconductors are known to exhibit a host of nonlinear
Modern research in biosensor technology highlights a optical properties and quantum confinement effects which
constant increase in the implementation of nanomaterials consequently result in exceptional properties, like the lumines-
either into the transducer or receptor part or both to enhance cence in quantum dots (QDs). With their salient properties,
multidetection capability and sensitivity. Since the nanomate- semiconducting nanomaterials are utilized as the biolabel/
rials have a very high surface to volume ratio, they can biomarker for bacteria detection through employing various
accommodate many target-specific recognition elements and analytical techniques such as fluorescence, 3D microchan-
subsequently improve the sensitivity of the biosensor device. nels,38 photo-corrosion,39 sulfuration,40 matrix-assisted desorp-
Precisely, in comparison with traditional biosensors, nano- tion/ionization mass spectroscopy,41 colony counting,42 spec-
materials-based biosensor/bio-devices exhibit numerous trophotometry,42 flow cytometry,42 light emitting diode induced
advantages such as fast response time, high sensitivity, reduced fluorescence detection43 etc. Among the various pathogen
size and enhanced versatility. Exploiting the advantages of the detection techniques used with semiconducting nanomaterials,
nanomaterials, researchers have designed interaction between the fluorescence enabled spectroscopic technique dominates
biological elements and nanomaterial-based transducers and because it provides low detection limits and is insensitive to
utilized various kinds of nanomaterials for pathogen detection magnetic noise. However, there can be two difficulties pertain-
and related disease diagnostics. ing to the stability of the luminescence from pristine semi-
Considering the importance of pathogen detection in many conducting nanocrystals, namely, the existence of surface states
sectors such as the food industry, water management, envi- arising from non-stoichiometry and unsaturated bonds and very
ronmental quality control and biodefense and knowing the reactive nature owing to their high surface-area-to-volume
varieties of pathogens, a number of detection strategies have ratio. Therefore, controlling the surface properties of the semi-
been reviewed over the past 5 years focusing individually on conducting nanocrystals is highly important for achieving high
multi-drug resistant pathogens,28 food borne pathogens,29 water luminescence with adequate stability to be used for pathogen
borne pathogens30 and whole cell bacteria.31 In conjunction detection. In the forthcoming sections, the use of individual
with the advancement of nanomaterials synthesis and utiliza- semiconducting materials toward pathogen detection will be
tion for various applications, the field of bacterial biodetection discussed.
also flourishes with the variety of bio-sensing strategies. The 2.1 Quantum Dots (QDs). QDs represent the II­VI, III­V,
advanced technology of utilizing nanomaterials toward sens- and IV­VI group semiconductor materials, their binary, alloyed

218 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
Figure 1. Formation of Fe@Au functionalized nanoparticles conjugated with anti-Salmonella and the determination of Salmonella
typhimurium.51 (Adapted with permission from Ref. 51, Copyright 2014 Elsevier)

and core-shell systems with a confined size on the nanoscale biological applications such as labeling cellular structures,
(2­20 nm) in three dimensions. QDs are photostable, fluores- tracking the path and fate of an individual cell and as contrast
cent semiconductor nanoparticles and their emission wave- agents for imaging purposes have been discussed in many
lengths are tunable with particle size and composition.44,45 The reviews.44,50 However, the specific use of CdS for pathogen
physicochemical properties of QDs, specifically focusing on detection has not been reviewed yet.
the cellular toxicity of cadmium-containing semiconductor Sulfate reducing bacteria (SRB) are a group of microorgan-
QDs are detailed.46 A review on the importance of function- isms widely distributed in anoxic environments and are a
alization of QDs towards bio related applications is available.47 source of serious industrial and environmental concerns owing
It has been identified that toxicity is dependent on the QDs to their toxicity and corrosiveness. Freitas et al. reported the
surface properties (including a shell, ligand, and modifica- use of functionalized iron/gold core/shell nanoparticles
tions), aspect ratio and exposure time. The main advantages of (Fe@Au) with different self-assembled monolayers (SAM)
QDs in photoluminescence (PL) applications include size- and CdS nanocrystal (NC) for the amplified electrochemical
tunable PL (full-width-at-half-maximum, 25­35 nm), high immuno-sensing on a screen printed carbon electrode of
quantum yield (>20%) and long PL decay times (>10 ns).48 Salmonella typhimurium in food products (milk).51 The elec-
As compared to conventional fluorescent dyes, QDs have trochemical immunoassay was developed by the immobiliza-
superior properties like high photo-stability, dual colour tion of anti-Salmonella antibodies and CdS nanocrystals and is
detection, good selectivity, and biocompatibility. Thus, QDs represented in Figure 1. From this method, bacteria detection in
can enhance the existing fluorescent detection schemes or a milk sample can be made in very low concentration with an
mechanisms. Besides, QDs can be functionalized with a broad analysis time of less than 1 hour.
range of pathogen recognition elements.49 Notably, one of the Qi et al. developed a novel technique based on the microbial
unique advantages of QDs over conventional fluorophores is biosynthesis of CdS nanoparticles (NPs) and used it as a power-
that QDs can be utilized for multiplexed detection or simu- ful fluorescent label for SRB detection.52 Since the CdS NPs
ltaneous detection of two or more bacteria. Due to these are synthesized both intracellularly and extracellularly, they are
advantages, the use of bioconjugated semiconductor QDs in expected to be present and distributed near the bacterial walls.
bacteria detection has been emerging over the last few years. The detection rate of SRB, Desulforibrio caledoiens is found to
The problems associated with QDs, such as surface traps and be in the range of 1*102 to 1*107 cfu/mL. With this proposed
defects, could be overcome by growing epitaxial layers of method, the authors claimed that they were able to shorten the
inorganic material over the QD core material to obtain a core/ detection time to 2 days over the widely used most probable
shell QD structure, which can provide a substantial improve- number (MPN) technique which takes about 15 days to com-
ment in the PL efficiency. Cadmium selenide (CdSe) cores plete the process. CdS NP with suitable surface modification
covered with a layer of zinc sulfide (ZnS) is considered the best has also been used for the detection of pathogens. Abdelhamid
available QD fluorophore owing to exceptional performance. In et.al. designed a biosensing platform with novel chitosan
these nanostructures, the ZnS layer passivates the core surface, capped QDs (CdS@CTS) for the detection of pathogenic
protects it from oxidation, prevents leaching of the CdSe into bacteria (Pseudomonas aeruginosa and Staphylococcus aure-
the surrounding medium and in turn produces a remarkable us)53 utilizing their affinity interactions between the chitosan
enhancement in the PL yield. The development of core-shell molecules and bacterial membranes (CdS@CTS-bacteria).
QD structures has become vital for pathogen detection. Further, non-covalent interactions in CdS@CTS-bacteria were
2.1.1 Cadmium Sulfide (CdS): CdS is an essential II­VI probed through thermodynamic studies (negative and posi-
group semiconductor with a wide bandgap of 2.42 eV. Its tive enthalpy), and matrix-assisted laser desorption/ion mass

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 219
spectrometry (MALDI-MS). Transmission electron microscopy 2.1.2 Cadmium Selenide (CdSe): CdSe has a narrow band
(TEM) images confirmed the affinity and interactions of gap of 1.74 eV and finds a broad range of potential applications
CdS@CTS with bacteria, P. aeruginosa (Figure 2(A)) and in the fabrication of nanoscale electronic, photonic, electro-
S. aureus (Figure 2(B)). Two pathogenic bacteria, S. aureus mechanical and biomedical devices.54­56 Xiong et al. reported
and P. aeruginosa, exhibited a wide linear dynamic range and a the biosynthesis of CdSe QDs intracellularly57 and employed
low detection limit of 150 and 200 cfu/mL, respectively in a them as nanoprobes for the detection of viruses and bacteria.
short period (³1 min). The bacteria detection involves a space-time-coupling strategy
for converting the bacteria (Staphylococcus aureus) into fluo-
rescent cells (cellular beacons). The pictorial representation of
nanobioprobe fabrication and the bacterial detection scheme is
depicted in Figure 3. Typically, a low detection limit of 8.94
ng/ml was reported. The proposed scheme has many advan-
tages like fluorescent probes being monodispersed with uni-
form size, high luminance with outstanding photostability.
Also, the probes are highly accurate, reliable, and repeatable.
There are only a few reports of the metal-accumulating bacte-
ria, Lysinibacillus fusiformis. By the suitable selection of the
antibody conjugation, this kind of new bioprobe can be extend-
ed towards the sensitive detection of various other bacterial
pathogens including pseudorabies virus, baculovirus, Salmo-
nella typhimurium bacteria, and SKBR-3 cells. CdSe based
core-shell nanomaterials were also effectively utilized for the
detection of pathogens and this will be separately presented in a
forthcoming section.
2.1.3 Cadmium Telluride (CdTe): CdTe QDs are widely
used for effectively capturing and detecting pathogenic bacte-
ria. For selective capturing of Serratia marcescens, CdTe QDs
with an average size of 4­5 nm, conjugated with Concanavalin
(CdTe QDs-Con A) were used.58 The PL intensity of CdTe
QDs­Con A showed a linear graph in the range from 10 to
Figure 2. TEM images of (A) P. aeruginosa and (B) 90 f/ml. Similarly, there was a linear relationship between PL
S. aureus after incubation with (a) control, (b) CdS@MPA intensity and logarithm of the cell population of S.Marcescens
and (c) CdS@CTS.53 (Reprinted with permission from in the range from 1 © 10 to 1 © 106 (colony forming units)
Ref. 53, Copyright 2013 The Royal Society of Chemistry) cfu/mL. Shanehsaz et al. reported the detection of Helicobacter

Figure 3. Pictorial representation of (a) nanobioprobe fabrication and (b) bacterial detection scheme using fluorescent-bio targeting
bifunctional cells as bioprobes.57 (Reprinted with permission from Ref. 57, Copyright 2014 American Chemical Society.)

220 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
pylori using CdTe QDs.59 The detection is based on fluores-
cence resonance energy transfer (FRET) where it is used in
combination with a polymerase chain reaction (PCR) system.
Similarly, Morarka et al. fabricated a 3D microchannel with
CdTe QDs with diameter 3.5 nm of uniform size distribu-
tion within the polydimethylsiloxane (PDMS) elastomer for
the rapid detection of E. coli using fluorescence technique.38
Fluorescence image of E. Coli concentration ranging from 102
to 108 cells/l was determined. Nitrocellulose based dot assay
and FRET assay are used for the detection of pathogenic bacte-
ria Staphylococcus aureus using CdTe QDs as a biolabel.60 The
detection limits of dot assay and FRET assay were 3 ng/mL and
0.5 ng/mL, respectively.
CdTe QDs with a typical size range of 2­3 nm were synthe-
sized by Beibei Wang et al. for the detection of Salmonella
Enteritidis in egg samples.61 The low detection limit that was Figure 4. Pictorial representation of the GaAs/AlGaAs
achieved was 1*102 cfu/mL. This was achieved by a simple biochip functionalized with a Bio-PEG/HDT/Neutravidin/
bio-conjugation of anti Salmonella Enteritidis with QDs. It was antibody architecture.39 (Adapted with permission from
found that the detection time can be as low as 2 hours in egg Ref. 39, Copyright 2016)
which is effective in real time analysis of food samples. On the
other hand, a sensing device based on the CdTe was able to
simultaneously detect different bacterium such as Salmonella
Enteritidis, Staphylococcus aureus, and Escherichia coli.62
This was possible by tuning the emission wavelengths of these
QDs located at 504 nm, 557 nm, and 604 nm. For the effective
use of this developed technology, real food samples (such as
soda, mineral water, apple juice, milk, tomato sauce, chicken
sauce etc.) were used for bacteria detection.
Functionalized CdTe QDs have also been utilized for
pathogen detection. Kurt et al. reported the use of carboxylic
acid functionalized CdTe QD for bacteria detection.63 Aptamer-
functionalized CdTe QDs and upconverting nanoparticles were
used for specific detection of two food pathogens, namely,
Salmonella typhimurium and Staphylococcus aureus. The Figure 5. Scheme depicting the stages involved in DNA
minimum detectable concentrations of 16, and 28 cfu/mL were bielectrode fabrication.74 (Adapted with permission from
found for Staphylococcus aureus and Salmonella typhimurium, Ref. 74, Copyright 2014 Elsevier.)
respectively. Similarly, Agarwal et al. demonstrated the multi-
plexed detection of waterborne pathogens in a circular micro-
fluidics, Escherichia Coli and Salmonella typhimurium using corrosion is dependent on the concentration of the electric
CdTe QDs.64 In that report, green and red fluorescent nano- charge of the surface. A detection limit of 103 cfu/mL is
materials were synthesized by changing the size of the CdTe achieved through this technique. In a similar way, Aziziyan
nanomaterials. The detection limits of Escherichia Coli and et al. used a GaAs/AlGaAs QDs biochip functionalized with
Salmonella typhimurium are found to be as low as 103 and a Bio-PEG/HDT/Neutravidin/antibody architecture for the
107 cfu/mL. detection of Legionella pneumophilia (Figure 4).39 Bacteria
2.1.4 Gallium Arsenide (GaAs): GaAs is a compound detection is found to be in the order of 104 bacteria/ml when
semiconductor which has a high electron mobility and a direct the bacteria is suspended in phosphate buffer saline (PBS)
band gap structure which result in improved PL signal in the solution. The biosensing of bacteria using photo-corrosion can
NIR range.65 The intrinsic luminescence of GaAs either alone control the etch depth resolution in the order of nanometres.
or in a heterostructure with other compounds such as aluminum 2.1.5 Zinc Oxide (ZnO): ZnO is a II­VI type inorganic
gallium arsenide (AlGaAs) is sensitive to the physical and semiconductor exhibiting direct wide band gap (3.3 eV), a high
chemical states of its surface and is exploited for PL based excitonic binding energy (60 meV), high refractive index, ther-
optical biosensing application. GaAs or GaAs/AlGaAs hetero- mal conductivity and many other outstanding physico-chemical
structures can be combined with a molecular recognition ele- and fascinating photo-physical properties.67­73 ZnO and nano-
ment and form promising candidates toward designing optical sized ZnO are also identified as antibacterial agents. These
biosensors for pathogen detection. Nazemi et al. reported the unique characteristics enable ZnO to be used in a broad range
synthesis of GaAs/AlGaAs heterostructures and demonstrated of applications including pathogen detection.
that their photo corrosion effect can be effectively used for the Low cost hydrothermal technique was used for the synthesis
detection of bacteria.66 By using PL technique, the detection of of flower-like ZnO nanostructures (ZnF) which were then
E. Coli was possible. It was reported that the rate of photo- immobilized with DNA molecules for the effective detection of

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 221
N. Meningitides.74 Figure 5 shows the stages involved in the a particular species.75 ZnO NPs assisted MALDI-MS is used
preparation of DNA bioelectrode fabrication. The nanostruc- as a sensitive technique for bacteria detection. This report
tures and the higher loading of DNA were found to be respon- demonstrated that ants could carry pathogenic bacteria such as
sible for the highly selective detection of this bacteria. The Staphylococcus aureus where a detection limit of 103 cfu/mL
detection limit was found to be 5 ng uL¹1. The fabricated was found.
bioelectrode exhibited a high sensitivity of about 168.64 ng and Chen et al. designed peptide-based ZnO QDs and developed
a wide range of 5 to 240 ng. Figure 6 presents the SEM images a fluorescent nano-probe called ZnO@BSA-PEP-MPA (BSA:
of (a­c) ZNF/Pt/Si and (d) DNA/ZNF/Pt/Si bioelectrodes at stabilizing agent, MPA: near infra-red dye) with low cytotox-
high and low magnifications before and after immobilization. icity for the detection of in vivo bacteria diagnosis through
Gopal et al. also reported the synthesis of ZnO NPs for the in vivo early bacteria imaging.76 The structure and stages
detection of Staphylococcus aureus from the surface of ants of involved in the preparation procedure are shown in Figure 7.
The developed strategy could achieve a detection level of
1.6 © 104 and potentially find prospects in anti-multidrug resis-
tance applications. Pan et al. reported the synthesis of ZnO,
titanium dioxide (TiO2) and silicon dioxide (SiO2) NPs for the
detection of four pathogenic bacteria (Staphylococcus epider-
midis, Enterococcus faecalis, and Escherichia coli).42 Three
different methods are used for the quantification of bacteria
such as colony counting to determine colony forming unit
(CFUs), spectrophotometer method of optical density and flow
cytometry (FCM). Among the three different methods, CFU
counting is found to be time-consuming and less accurate. The
spectrophotometry technique is also found to be the most
unreliable while FCM is considered to be an appropriate tech-
nique owing to its rapid, sensitive and simultaneous detection
of pathogen.
Like bacteria detection in a food sample, detection of the
same in a water sample is also challenging. Gedda et al. report-
ed the synthesis of ZnO NPs for bacteria detection in water.41
Figure 6. SEM images acquired at low and high magnifi- Dispersive liquid­liquid microextraction was used to detect
cations before and after immobilization. (a­c) ZNF/Pt/Si bacteria using ZnO NPs modified with polymethyl methacry-
and (d) DNA/ZNF/Pt/Si bioelectrodes.74 (Reprinted with late (ZnO@PMMA). The chemical structure and the pictorial
permission from Ref. 74, Copyright 2012 Elsevier) representation of ZnO@PMMA coupled with MALDI-MS for

Figure 7. Synthetic route for the preparation of ZnO@BSA-PEP-MPA.76 (Reprinted with permission from Ref. 76, Copyright 2015
Elsevier)

222 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
In another report, the stability of the sensors were signifi-
cantly enhanced by using oligonucleotide microarray combined
with CdSe/ZnS QDs as fluorescent labels.78 The bacteria was
identified with PerkinElmer Gx microarray scanner displaying
sensitivity of 10 CFU/mL. Fluorescence detection of E. Coli
0157: H7 bacteria was obtained by using CdSe/ZnS QDs with
carboxylic functional groups.79 The 72-mer aptamer is used as
the probe and sensing element where detection limit of 102
cfu/ml could be achieved. Drugs are susceptible to the infec-
tion by bacteria and other contaminations. Therefore, it is
essential to detect bacteria in drug samples. Wu et al. used
ZnS/CdSe QDs conjugated with the rabbit anti goat antibody
for the detection of E. Coli in drugs.80 The detection process
took less than four hours to complete which is acceptable for
ensuring the rapid detection of bacteria. Detection of Salmo-
nella bacteria from chicken breast samples using CdSe/ZnS
Figure 8. Illustration of ZnO@PMMA-DLLME towards QD was reported by Kim et al.81 A microfluidic nanosensor
extraction of pathogenic bacteria.41 (Adapted with permis- based on CdSe/ZnS QD immobilized with anti-salmonella
sion from Ref. 41, Copyright 2014 The Royal Society of polyclonal antibodies was used for the detection and showed
Chemistry) the detection limit of 103 cfu/mL of borate buffer and food
extract. In another report, the same group employed streptavi-
the extraction of bacteria is shown Figure 8. A minimum fluo- din functionalized CdSe/ZnS core-shell QDs for the detection
rescence detection level of 9.7 © 103 and 1.7 © 104 cfu/mL for of Salmonella.82 The detection limits were found to be 1.4*103
S. aureus and P. aeruginosa was reported. A high surface area cfu/mL in bovine serum albumin (PBS­BSA) and 4*103 in
of ZnO@PMMA NPs was found to be the primary reason for food extracts. Light emitting diode induced fluorescence detec-
the high sensitivity of MALDI-MS spectra. tion (LIF) can also be used for bacteria detection. Wang et al.
2.1.6 Core-Shell Quantum Dots: It has been well demon- reported the synthesis of CdSe/ZnS QDs for the detection of
strated through several research studies that a fraction of sur- Salmonella typhirium.43 The detection was based on a micro-
face atoms in QD nanocrystals are passivated by the stabilizing fluidic chip and a LIF microsystem. The detection limit was
organic ligands. Hence, these passivated core QD nanocrystals found to be 37 cfu/mL. The simultaneous detection of E. Coli
exhibit surface trap/defect states which can act as fast non- and Salmonella in ground beef was also successfully achieved
radiative channels for photogenerated charge carriers and by Wang et al. using ZnS/CdSe QDs.83 The detection limit was
decrease the fluorescence quantum efficiency. One of the found to be 10 cfu/g after 24-hour enrichment. It used a bead
strategies to alleviate the problems in QD nanocrystals is to free technique for the detection and detection time was less
form a shell layer of a second semiconductor, resulting in core/ than 3 hours without enrichment.
shell QDs. Core/shell QD-based nanomaterials with both core Qi et al. reported the synthesis of ZnO for the determination
and shell made of semiconducting materials are commonly of SRB. From ZnO, firstly an intermediate such as ZnO/ZnS
synthesized and used as biomarkers for the sensitive detection is formed during sulfuration and then later ZnS arrays are
of various bacteria. Various semiconducting nanomaterials formed. The proposed method takes only 4 days versus 15 days
such as cadmium sulfide (CdS), CdS0.5Se0.5, CdSe/ZnS@SiO2, using the conventional most probable number (MPN) tech-
cadmium telluride (CdTe), CdTe/Cd(OH)2, GaAs/AlGaAs, nique. ZnS nanoparticles were also utilized for the bacteria
PbS, ZnO, ZnS, ZnS/CdSe, ZnS:Mn were utilized for bacteria detection towards SRB detection.40 Using this method, the
detection by exploiting the unique physicochemical character- population of SRB can be monitored selectively and can be
istics of the nanomaterials through different kinds of analytical employed without any biological markers or sophisticated
methods/techniques. instruments. Importantly, the time taken for detection is only
2.1.6.1 Zinc Sulfide (ZnS)/Cadmium Selenide (CdSe); 3 days and is much less than MPN method.
ZnS/CdSe is the most commonly used core/shell semiconduct- 2.2 Inorganic Metal Oxides. Bahadoran et al. reported the
ing nanomaterial for the detection of bacteria using fluores- use of SiO2-TiO2 hybrid for the detection of the Salmonella
cence techniques. Arshad et al. reported the use of CdSe/ZnS bacteria in human blood.84 Here, the SiO2-TiO2 nanomaterial
QDs for the detection of Vibriyo harveyi in solution and in was used for making a double ring add-drop filter micro ring
animal cells.77 Techniques such as fluorescence microscopy, resonator which in turn was used to detect bacteria. The scatter-
elastase assay, polyacrylamide gel electrophoresis (PAGE), and ing matrix method was utilized for the detection of the output
comet assay are used to evaluate the interactions of QDs with signal by inductive calculation. The achieved sensitivity is
Vibriyo harveyi. The appearance of bright yellow fluorescence 95500 nm/refractive index unit (RIU), and the detection limit
when different concentrations of QDs were applied confirms is 1 © 10¹8 RIU.
the perfect attachment of the QDs to the bacteria. It was also
demonstrated that the toxicity level of CdSe/ZnS QDs is 3. Magnetic Nanoparticle Based Bacteria Detection
genetically and cytotoxically safe for labelling the bacteria Crystal violet or gentian violet (CV; 4,4¤,4¤¤-dimethylamino-
allowing live imaging and tracking of the microorganisms. triphenylmethane) is a triarylmethane based deep purple dye

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 223
particularly used in Gram’s method for classifying bacteria.85 multiplex PCR methodology for the sensitive and concurrent
Budin et.al. modified CV functionalized with trans-cyclooctene recognition of the three food pathogens, viz. Salmonella
(CV-TCO) for the recognition of Gram-positive bacteria and enterica, Listeria monocytogenes, and Escherichia coli. Silica
evaluated three samples Staphylococcus aureus (S. aureus), magnetic nanoparticles have been utilized as carrier platform
Escherichia coli (E.coli) and the combination of two species.86 for immobilization along with various electrochemical report-
Interestingly, they observed that CV-TCO could act as a host ers (anti-Fluorescein­HRP, Streptavidin­HRP and anti-Digox-
anchor to immobilize tetrazine (Tz)-modified magnetic nano- igenin­HRP) specific to each pathogen. This study was aimed
particles. Gram stain, being the most standard technique for at detecting low concentrations of Salmonella enterica, Listeria
differentiating bacteria was utilized for the sensitive detection of monocytogenes and Escherichia coli (Figure 10). It was report-
Gram-positive pathogens using micro nuclear magnetic reso- ed that this method could successfully detect bacteria in less
nance (μNMR). It was observed that orthogonal CV is useful in than 50 min with LOD ranging from 12­46 pg/¯L.89 The
distinguishing biological samples. In addition, the implemented authors envisage that this versatile sensing strategy could find
standard Gram strain method along with labeling of magneto- applications in microfluidic-based devices.
fluorescent nanoparticles modified with tetrazine (MFNP-Tz) Xia et.al. investigated bio-accessibility of seven types of
for strain bacteria can enhance the detection and character- soil with various amounts of As, Cd, and Pb. They showed that
ization of bacteria both by μNMR and optical imaging.86 The presence of Pb in the soil was the main control parameter for
proposed strategy can be further extended to attach small
molecule ligands for the specific recognition of other bacterial
samples.
Chan et.al. reported biofunctional polystyrene magnetic
beads (particles) for ultra-sensitive detection of E. Coli
O157:H7 through conjugating magnetic bead concentration
on a nanoporous alumina membrane-based electrochemical
immunosensor. The beads were modified by specific antibody
to enhance sensitivity. Figure 9 presents the functionalization
process for nanoporous alumina membrane. By utilizing an
external magnetic field, E. Coli O157:H7 cells could be easily
localized in a confined area on the nanoporous membrane to
form a sandwich architecture of antibody-modified magnetic
beads. Impedance measurements were used to evaluate the effi-
cacy of the conjugated and non-conjugated E. Coli O157:H7
by applying a magnetic field. By using this conjugation-
concentration strategy, the authors could attain the detection
limit of 10 CFU/mL (Figure 9).87
Among the different strategies available for simultaneous
detection (such as time-resolved fluorescence (TRF), biosen- Figure 9. Pictorial representation for biofunctional mag-
sors, multiplex quantitative polymerase chain reaction (PCR), netic bead concentration of E. Coli O157:H7 in the bio-
etc.) electrochemical genosensors have been most actively chip. (a) Polystyrene beads were conjugated with E. Coli
employed for identifying pathogens, offering a futuristic pros- O157:H7; (b) Concentrated in a confined region under the
pect for simultaneous detection, owing to their high sensitivity, influence of an magnetic field and (c) Removal of magnetic
specificity and cost effectiveness.88 Brandao et al. combined field.87 (Adapted with permission from Ref. 87, Copyright
electrochemical magneto-sensing together with triple-tagging 2013 Elsevier)

Figure 10. Electrochemical approach of triple-tagging magneto genosensing on silica magnetic particles by varying the quantity of
electrochemical reporter (from 1 to 30 ¯g/assay).89 (Reprinted with permission from Ref. 89, Copyright 2015 Elsevier)

224 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
Figure 11. Combinatorial strategy using Apt-MB and AuNCs@Van for the quantification of SA.92 (Adapted with permission from
Ref. 92, Copyright 2016 American Chemical Society.)

bio-accessibility of Pb. Further studies for a simulated human netic nanoparticles (MNPs) and TiO2 nanocrystals (TNs) as
digestive system revealed no impact of As or Cd on Pb bio- optical nanoprobes to capture Salmonella bacteria in a buffer
accessibility.90 It was proved that proximity ligation assay solution or milk through optical sensing (UV-vis spectroscopic
(PLA) could be utilized using magnetic particle based substrate- detection) (Figure 12). The UV-visible spectrum of the ob-
free sensing, without the aid of thermocyclic or optical tained complex revealed the high sensitivity of the assay
equipment.91 toward low concentration of Salmonella bacteria. The detection
Cheng et.al. devised a strategy to combine inexpensive and scheme is illustrated in Figure 12. The proposed measurement
widely available aptamer and antibiotic-based dual recognition process was found to be much quicker, cost efficient and less
units with magnetic enrichment for the sensitive detection of hazardous over conventional methods due to the absence of a
Staphylococcus aureus (SA) in the presence of other bacteria cell culturing process with a detection limit of over 100 cfu/mL
with higher contents. To capture SA, magnetic beads modified for Salmonella bacteria in milk.94
aptamer (Apt-MB) were utilized. A single step process has Kim et.al. also proposed a detection platform for the rapid
been applied for vancomycin-stabilized fluorescent gold nano- and selective capture of pathogenic Salmonella, based on a
cluster (AuNCs@Van) synthesis. Apt-MB and AuNCs@Van theoretically optimized model. They have reported the prepa-
were further used for the quantification of SA in milk and ration of water-soluble superparamagnetic Fe3O4 magnetic
human serum samples with a high recovery ratio. It has been clusters coated with hydrophobic ligand (oleic acid) to provide
shown that around 70 cfu/mL of SA in the mixture can be conjugation sites for antibody immobilization followed by
detected via this technique (Figure 11). The as-developed evaporation-induced self-assembly (EISA) strategy to modify
approach can be used as a dual recognition technique for the the magnetic properties. Two different antibodies: H- and O-
specific and sensitive detection of SA in diagnosing infectious antigens were immobilized on the cluster surface to enhance
diseases.92 the selective binding. Typically, the results suggested that O-
3.1 Iron Oxide Nanomaterials. Kwon et.al. devised a antibody-coated polysorbate 80-coated magnetic nanoclusters
facile method for detection of Salmonella bacteria in milk (PCMNCs) exhibited the highest capture efficiency (³99%)
using magnetophoretic chromatography. To accomplish this, for Salmonella,95 while the capture efficiency for H-antibody
gold-coated Fe3O4 magnetic nanoparticles (Au/MNCs) were modified magnetic nanoparticles was relatively lower (³57%).
suitably surface functionalized with amine-reactive N-hydroxy- Shukla et.al. demonstrated a method for rapid detection of
succinimide (NHS) linkers to immobilize the antibody. After Cronobacter Sakazakii (C.sakazakii) in both culture and infant
10 min of magnetophoretic chromatography, 100 cfu/mL of formula. They have coated immunoglobulin G (lg G) onto
Salmonella bacteria could be detected with the naked eye. carboxyl Fe2O3 magnetic nanoparticles. The proposed assay
The proposed method sensitivity is limited to 100 cfu/mL, exhibited a detection sensitivity of 3.3 © 103 CFU/mL of
which has been validated using absorption spectroscopy. It was C.sakazakii in pure culture within 2 h 30 min, while an indirect
believed that the reported sensitivity could be further enhanced non-competitive enzyme-linked immunosorbent assay exhib-
by improving impregnation of MNCs with fluorescent tags. ited a detecting ability of 6.2 © 105 cfu/mL of C.sakazakii in
This technique can also be applied to rapid on-site detection pure culture after 17 h and 103 cfu/mL of C.sakazakii was
as it can be assembled with a permanent magnet and precision observed in infant formula. However, the developed method
pipette for food safety monitoring.93 suffered from the limitation of multiplexing efficiency.96
In another study, Jin et.al. described a similar magnetic Tian et.al. utilized two kinds of magnetic particles, micron-
separation technique via anchoring antibody onto Fe3O4 mag- sized and nano-sized particles as capture and detection parti-

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 225
Figure 12. The schematic representation using optical nanoprobe (left) and (right) TEM images acquired for Salmonella bacterium
after binding to (a) bare MNPs, (b, c) antibody-immobilized MNPs and antibody-immobilized TiO2 and (d) A magnified portion of
the square region from (c).94 (Reprinted with permission from Ref. 94, Copyright 2012 The Royal Society of Chemistry.)

cles, respectively through a novel and cost-efficient Blu-ray DNTB were identified at 1582 cm¹1 and 1333 cm¹1 that corre-
optomagnetic technique. The proposed method was able to sponds to the S. typhimurium and S. aureus, respectively.
detect Salmonella bacteria as low as 8 © 104 CFU/mL within Under optimal conditions, this simple approach presented the
3 h of process time. The observed sensitivity limit was 20 times LOD of 35 CFU mL¹1 for S.aureus and 15 CFU mL¹1 for
lower by using the as-developed technique compared to volu- S.typhimurium that is high when compared to the existing
metric magnetic stray field detection device immunoassay. The methods.100 This method is straightforward and rapid, results
newly developed method exhibited high compatibility to dif- in high sensitivity and specificity, and can be used for the
ferent types of pathogens. Moreover, no visible cross-reaction detection of practical samples.
has been observed with other bacteria.97 Wang et.al. introduced a technique for developing porta-
Xu et.al. proposed a detection technique using ble and sensitive pathogen detection using a personal glucose
immunosensor-based on a screen printed interdigitated micro- meter (PGM). Sequential steps were followed for the preparat-
electrode (SP-IDME) coupled with 130 nm iron oxide (Fe3O4) ion of glucoamylase-quaternized magnetic nanoparticles (GA-
magnetic beads (MBs) and glucose oxidase (GOx) antibody QMNPS) through the chemical modification of Fe3O4 with
labeling for the capture of foodborne and iatrogenic pathogens SiO2 (Fe3O4@ SiO2). Fe3O4@SiO2 is further surface modified
(Escherichia coli O157:H7 and Salmonella Typhimurium). It with N-trimethoxysilypropyl-N, N, N-trimethylammonium
was reported that as developed approach was capable of detect- chloride to obtain QMNPS. Implemented glucoamylase-
ing the E. Coli O157:H7 and Salmonella Typhimurium with quaternized magnetic nanoparticles (Fe3O4) (GA-QMNPS)
LOD of 2.05 © 103 CFU g¹1 and 1.04 © 103 CFU g¹1, respec- interacted with the pathogenic bacteria and released GA as a
tively.98 Yang et.al. utilized dual recognition agent to increase result. The presence of free GA, after magnetic separation,
the specificity of the Staphylococcus aureus (S. aureus), by could catalyze the hydrolysis of amylose. The proposed
applying vancomycin, as a strong antibacterial agent function- approach was very sensitive to low concentration down to
alized with magnetic beads and alkaline phosphate (ALP)- 20 cell mL¹1 suitable for clinical application. However, there is
tagged rabbit immunoglobulin G(ALP-lg G). It has been ob- no result on detection ability of their approach towards various
served that the S. aureus detection was 12­1.2 ©106 CFU mL¹1 microbes and pathogenic bacteria.101
with LOD as low as 3.3 CFU mL¹1, during 75 min of process A gold-coated nanoparticle-labelled electrochemical bio-
time. Interestingly, the presence of other bacteria did not have sensor has been developed by Wang and Alocilja for E. Coli
any effect on S. aureus detection. However, utilizing vanco- O157:H7 detection in broth samples. Magnetic gold nano-
mycin in the process produces a drug-resistant pathogen that particles (AuNPs) were conjugated with polyclonal antibodies
may require further sterilization.99 to segregate E. Coli O157:H7 from broth samples. The amount
S. Typhimurium and S. aureus are considered to be the root of E. Coli bacteria was quantified by measuring the content of
cause of many food borne diseases. In order to address and AuNPs coated on a screen-printed carbon electrode via an elec-
provide a feasible solution, Zhang et al. developed a Raman- trochemical method (Differential pulse voltammetry, DPV).
based biosensing platform for the concurrent detection of The dynamic linear range of 10­106 CFU mL¹1 has been
pathogens, S. typhimurium and S. aureus. For this purpose, reported within 1 h of detection and extraction.102
gold nanoparticles (GNPs) have been suitably incorporated Wang et.al. developed a low-cost magnetic assisted SERS
with Raman molecules (mercaptobenzoic acid (MBA) and biosensor for single-cell detection of S. aureus based on apta-
Dithiobis(2-nitrobenzoic acid)) (DNTB)). GNPs and aptamer mer recognition. The as-developed biosensor contained two
have been applied as the signal probe for S. typhimurium and main parts including a SERS substrate (Ag-coated magnetic
S. aureus, correspondingly. The SERS intensity of MBA, NPs) and SERS tag (AuNP-DTNB@Ag-DTNB core-shell plas-

226 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
Figure 13. Colorimetric immunoassay for rapid detection of S. pullorum based on blue-SiNPs and MNPs.104 (Adapted with
permission from Ref. 104, Copyright 2015 American Chemical Society.)

monic NPs or DTNB-labelled inside-and-outside plasmonic


NPs, DioPNPs). DioPNPs exhibited 10 fold higher Raman
intensity compared to AuNR-DTNB and observed good linear
range from 10 to 105 cells/mL for S.aureus detection.103
A colorimetric immunoassay antibody-coated magnetic
nanoparticle (IgG-MNPs) has been developed by Sun et al.
for quantitative Samonella pullorum (S. pollorum) detection.
Blue-SiNPs (Tetraethyl orthosilicate as Si source) surface-
modified with anti-Salmonella pullorum (S. pollorum) has been
utilized as detection probes. Magnetic nanoparticles (MNPs) Figure 14. Rapid capture of selected pathogen through
has been applied to anchor polyclonal antibodies against gold-coated iron oxide nanoparticle treatment.107 (Adapted
(S. pollorum) considered as a capture probe (Figure 13). The with permission from Ref. 107. Copyright 2015 Elsevier.)
application range for the as-prepared assay was 4.4 © 102
CFU mL¹1 to 4.4 © 107 CFU mL¹1 with LOD of 4.4 © 101 treatment of QCM surface with NaOH solution could regen-
CFUmL¹1 for S. pollorum. Interestingly, in this assay, no erate the aptamer-sensor allowing each crystal to be reused.106
enzyme catalysis was required and exhibited a high selectivity Niemirowicz et.al. reported the effect of gold-coated iron
toward S. pollorum among other pathogenic bacterium.104 oxide nanoparticles and their derivatives on restricting various
Rahman et.al. studied the impact of various nanoparticles on pathogens growth and assisting in capturing them (Figure 14).
multiplex polymerase chain reaction (PCR) method for recog- The results revealed a substantial reduction of E. Coli,
nition and strain typing of Salmonella enterica serotype typhi S. aureus; P. aeruginosa and sandida albicans growth after
(S. Typhi) (targeting Variable Number of Tandem Repeats exposing them to MNPs for 24 h. However, the phenomena
[VNTR]) by employing different nanomaterials such as citrate depend on MNPs concentrations, pathogens strains, surface
capped gold nanoparticles, rhamnolipid protected gold and chemistry and the surrounding environment.107
silver and magnetic iron oxide nanoparticles. It was observed Antibody/enzyme-conjugated MNPs have been utilized for
that appropriate incorporation of nanoparticles significantly S. typhimurium detection by Mun et al. using 100 nm-antibody
reduced non-specificity of PCR. The results demonstrated the and horseradish peroxidase-conjugated magnetic MNPs (Ab-
enhancement of target DNA template. It is worth mention, the HRP-MNPs). The response range of this technique was 104
improvement relied heavily on the nature of applied metal CFU/mL with LOD of 10 CFU/mL. An 80 nm iron oxide core
nanoparticles. However, there is no clear discussion on the exact and 10 nm silica and APTES EDC/NHS treated MNPs also
mechanism of diagnosis using different metal nanoparticles.105 have been used.108
Ozlap et.al. utilized a quartz crystal microbalance sensor Mezger et al. demonstrated a rapid and sensitive molecular
(QCM) for the recognition of Salmonella enterica serovar detection approach for detection of bacteria causing urinary
Typhimurium cells in milk samples. The results demonstrated tract infection using padlock probe, and on-bead isothermal
that the integration of aptamer-magnetic and QCM-sensor tech- rolling cycle amplification (RCA) integrated with on-disk auto-
nique provided a quick and sensitive detection of Salmonella in mated aptomagnetic nanoparticles. Interestingly, the change
food contaminants with a detection range from 100 to 4 © 104 of temperature and laborious DNA preparation were not
CFU/mL cell and LOD of 100 cells in less than 10 min. Further required. Optomagnetic read out has been utilized for ampli-

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 227
fication product detection and demonstrated highly sensitive
and multiplexing capability. Implementing RCA monomers 4. Noble Metal Based Nanostructures
enhanced the sensitivity by 10 fold, along 3 fold improvement for Bacteria Detection
for magnetic incubation. Therefore, the proposed approach Noble metal nanoparticles such as platinum, gold, and silver
exhibited 10 times higher sensitivity than the sample prepara- are fascinating materials with unique electrical, optical, mag-
tion amplification and detection process was more rapid (4 h netic, surface, and chemical properties depending on their size
including preparation time) compared to 7 days for other and morphology. Due to their unique physico-chemical prop-
methods.109 erties at the nanoscale, noble metal nanoparticles have been
Liu et al. prepared a magnetic nanoparticle-labeled immu- continuously explored in the evolution of new kind of sensors.
nochromatographic test strip (MNP/ICTS) for the food-borne Furthermore, nanoparticle-based biosensors are particularly
pathogen, vibrio parahaemolyticus (v. parahaemolyticus). The attractive because the synthetic process of nanoparticles is
specific antibody has been immobilized onto the superpara- straightforward and does not require advanced fabrication
magnetic nanoparticles. The observed sensitivity was 1.58 © methodologies. They also offer very high surface areas due to
102 CFU/mL for v. parahaemolyticus in about 4.5 h.100 Lin and their minuscule size and are typically used as suspensions in
Hamme developed an assay for quantitative and qualitative solutions, where they effectively interact with the analytes.
detection of Salmonella DT104 by utilizing antibody affinity Because of their large surface area, excellent biocompatibility
binding; popcorn-shaped gold nanoparticle (GNPoPs) labeling, and ease of functionalization, various noble metal nanomate-
surface enhanced Raman spectroscopy (SERS), and inductively rials, especially gold/silver nanoparticles, have been used as
coupled plasma mass spectrometry (ICP-MS). The LOD of excellent carriers for loading different bio-recognition moieties
100 CFU/mL has been noticed within 40 min (Figure 4).110 such as enzymes, antibodies, aptamers, organic ligands, and
Although the developed assay is very sensitive in detecting antimicrobial agents which can dramatically amplify and trans-
Salmonella DT104, a higher concentration of bacteria causes duce the signals in a biosensor device. In the forthcoming
less accurate detection due to bacteria binding to antibody- sections, we will focus on the noble metal nanomaterial such as
conjugated GNPoPs. gold silver, platinum, and bimetallic nanomaterials, used for
Lee et al. incorporated a 3D-printed helical microchannel the quantification of pathogenic bacteria using various ana-
and magnetic nanoparticle clusters to detect E. Coli bacteria lytical techniques.
in milk samples. The process includes dispersing free MNCs 4.1 Gold Nanostructures for Pathogen Detection.
and MNCs-EC components into buffer mixture, followed by Fleischmann et al. discovered the phenomenon of surface-
injecting the resultant solution into a helical microchannel enhanced Raman spectroscopy (SERS) which is considered to
device. The LOD was reported around 10 CFU/mL in buffer be among the most popular ultrasensitive analytical techniques
solution and 100 CFU/mL in milk sample.111 A colorimetric due to its high detection capability and sensitivity.115 Noble
technique has been developed using Pt-coated magnetic nano- metal colloidal particles and nanostructured surfaces have
particles and magnetophoretic chromatography for pathogenic become the most commonly used platform for SERS. The
bacteria detection. The Pt/MNPs were coated by monoclonal SERS phenomenon can be generally explained by a combina-
E. Coli O157:H7 antibodies to anchor E. Coli bacteria in tion of an electromagnetic and a chemical enhancement and
milk. After separating Pt/MNPs-EC from free Pt/MNPs via a out of which electromagnetic enhancement predominantly con-
magnetic field, tetramethylbenzidine (TMB) solution has been tributes to SERS. Electromagnetic enhancement exploits the
added to the separated part. Catalytic oxidation of TMB by Pt Raman scattering from molecules in close neighbourhood to a
led to colour changes of the solution visible to the naked eye. nanostructured surface due to the coupling effect of surface
The sensitivity observed was 10 CFU/mL in 30 min.112 plasmon (gold) via the oscillating electric field of the incident/
Hardiansyah et al. developed a multifunctional hybrid core- scattered radiation. The possibility of observing Raman signals
shell nanostructure composed of iron platinum (FePt), silica with enhancements in the order (106­1014) is normally weak
(SiO2) decorated by gold nanoparticles (AuNPs) (Au-FePt@ and the ability to obtain molecular recognition of target bacte-
SiO2-N NPs) for label-free surface-enhanced Raman scattering ria at very low concentrations allow SERS to be unique for
(SERS) sensing of biomolecules (adenine) and gram positive ultrasensitive pathogen sensing. High selectivity, sensitivity
pathogenic bacteria (S. aureus). Remarkable intensity has been and informative spectral characteristics derived from Raman
observed in SERS spectra for anchored adenine and S. aureus spectroscopy render SERS-based methods to be a feasible
on Au-FePt@SiO2-N NPs. The proposed SERS platform alternative for bacterial detection than traditionally used optical
displayed the potential application for simultaneous magnetic sensing methods. In this part of the review, we will discuss
separation and bio-sensing.113 Horikawa et al. demonstrated the SERS-based detection of pathogenic bacteria using the
bacteria detection in stagnant liquids using magnetoelastic advanced plasmonic nanomaterials including gold and silver.
(ME) bisentinels. ME bisentinels are made of pristine ME The plasmonic nanomaterials include both solution based
resonator grafted with an engineered phage-based molecular nanoparticles and nanomaterials prepared in the form of solid
recognition to bind with pathogenic bacteria of interest. Mag- platform. On reviewing this sub-topic, our observation is that
netic properties of biosentinels enable them to move through the majority of the plasmonic nanomaterials use SERS tech-
liquid via external magnetic force. After forming a chemical nique to identify pathogenic bacteria.
bond between bisentinels and pathogen bacteria, the presence Papadopoulou et al. reported the SERS-based label-free
of pathogen was identified by noticing the changes in the sensitive detection of unmodified single and double stranded
biosentinel resonant frequency.114 DNA (ssDNA and dsDNA) down to nanomolar concentration

228 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
by adsorbing Au and Ag colloids. Herein, magnesium sulfate core-gold plasmonic nanoparticle shell for the label-free SERS-
(MgSO4) was used as an aggregating agent mainly because of based detection, magnetic separation, imaging and selective
the low-affinity of sulfate ions for metal (Au and Ag) colloids photothermal destruction of MDR Salmonella DT104, a multi-
and the tendency to aggregate even at a higher concentration drug-resistant bacteria (MDRB), by utilizing an excitation
without displacing the adsorbed DNAs. Five DNA sequences source of 670 nm light.120 They have modified the surface of
that correspond to different strains of E. coli bacterium were the prepared core-shell nanoparticle with an antibody which is
used.116 The authors were able to differentiate SERS spectra of highly specific for the detection of MDR Salmonella DT104.
five sequences corresponding to various E.coli strains without The experimental results demonstrate promising results with a
using advanced multivariate data analysis techniques. On the limit of detection as low as 102 CFU/mL for MDR Salmonella
other hand, Zhang et al. demonstrated the rapid concentration DT104 using the hybrid nanostructures.
and SERS-based detection and identification of bacteria cells With an aim of enhancing the Raman signal, Chae et al.
at a concentration range of 105 CFU/mL using magnetic­ reported the SERS detection of infectious bacteria, E. Coli
plasmonic iron oxide-Au core-shell (Au-MNPs) nanoparticles (gram negative) and listeria monocytogenes (gram positive)
via applying external point magnetic field.117 The authors through the electrodeposition of AuNPs onto indium tin oxide
coated a known quantity (10 uL of 105 CFU/mL) of prepared substrate (ITO)121 and verified the successful formation through
Au-MNPs on the silica substrate to enable sensitive SERS- appropriate spectroscopic and microscopic techniques. The
active substrate. The authors could effectively differentiate antibody against E. Coli was immobilized on AuNPs to bind
three different Gram-negative bacterial strains (Acinetobacter the bacteria on the SERS substrate selectively through anti-
calcoaceticus, E. coli K12, and Pseudomonas aeruginosa) body-antigen interaction. Like any other SERS based nano-
through principal component analysis (PCA). With beneficial biosensors, the observed shift in Raman peak after E. Coli
plasmonic and magnetic properties and the existence of an binding was used for the detection of bacteria. Concentration
external magnetic field, Au-MNPs could be condensed to form range from 101 to 105 were detected with R2 = 0.99.
a highly compact dot (60 times more concentrated) in less than Xu et al. developed unique quasi-3D (Q3D) plasmonic
5 min. The proposed Au-MNP sensor platform offers many nanostructure arrays of plasmonic nanoholes via electron beam
advantages over the existing sample preparation method lithography (EBL) and used as a sensitive and reproducible
including low-cost, easy operation and rapid detection and SERS substrate to rapidly differentiate seven different strains
concentration of bacterial samples. The authors also envisage (50, 3355, 605, 551, 846, 12310 and 3256) of the marine
the potential applications in various field applications such as pathogen Vibrio parahaemolyticus.122 The fabricated Q3D was
environmental monitoring, food safety etc. patterned on the ITO/glass (³140 nm) substrate through EBL
Lin et al. also prepared highly sensitive SERS filter-like followed by metallization. The main advantage of this tech-
substrate using AuNPs encapsulated mesoporous silica nique is that herein in-situ acquisition of SERS spectra and
(AuNPs@MS) through a convenient one-pot method with a barcoding were utilized towards the direct identification of
high specific surface area and it utilized towards SERS appli- bacterial sample, Vibrio parahaemolyticus. The authors envis-
cation.118 This simple surfactant free SERS substrate was aged the potential usage of this technique in broad sectors such
prepared by dispersing Au NPs into the mesoporous silica as in environmental monitoring, homeland security etc. Chen
matrix and subsequent thermal treatment at 600 °C for 6 h to et al. also demonstrated a hybridization chain reaction-SERS
remove the organic protecting agents. S.aureus can be effec- coupled nano-biosensing platform via target DNA (tDNA)
tively localized on the prepared AuNPs@MS substrates within triggered self-assembly of Au nanoparticles (Au NPs) probes
a few seconds. The resultant SERS spectra exhibited character- on DNA nanowires for the signal amplification of Bacillus
istics peaks at 1158 and 1520 cm¹1 attributing to the cell wall thuringiensis DNA sequence and sensitive detection ranging
of S.aureus. SERS intensity increased significantly (over 900 from 50 pM to 500 pM with good linearity (S/N = 3).123 The
fold) for AuNPs@MS with Au wt% = 16 when compared with proposed SERS sensing platform exhibited a high sensitivity
that of the pure mesoporous silica without nanoparticle loading. and specificity for the detection of tDNA and many advantages
Keeping in the mind the low pathogenic concentration such as simple operation, and room-temperature implementa-
(<100 CFU/mL) in the cases like food poisoning and other tion without the assistance of an enzyme over the standard
infectious diseases, Drake et al. employed two different nano- enzyme reaction and RCA method.
particles labels, iron-oxide nanoparticles for magnetic trap- Lin et al. reported a highly sensitive SERS filter-like sub-
ping and 4-mercaptobenzoic acid (MBA) coated SERS active strate prepared via a one-step method by embedding AuNPs
AuNPs (MBA-AuNPs), for the quantification of pathogenic within mesoporous silica (AuNPs@MS). The AuNPs@MS acts
Staphylococcus aureus (S. aureus).119 MBA-AuNPs exhibited as a simultaneous matrix for the rapid filtration of the bacte-
signature peaks at 1077 and 1584 cm¹1 assigned to the aromatic ria in a few seconds and subsequent detection by SERS. This
ring vibrations, respectively.119 These kinds of nanoparticles bifunctional substrate was able to generate 900 fold greater
were conjugated with antibodies that can accurately bind to SERS signal by concentrating the Staphylococcus aureus
protein A on the S. aureus which led to an ultrasensitive SERS bacteria (Figure 15) than the typical SERS method.124
detection of 1 S. aureus cell/mL within 10 min. This work Lin et al. reported an integrated SERS nanoprobe and a
encouraged other researchers to work on the mixed pathogenic microfluidic dielectrophoretic (DEP) device for the faster
multiplexed system to enable simultaneous detection of several multiplex detection of multiple pathogens, Salmonella enterica
bacterial species. On the other hand, Fan et al. designed and serotype Choleraesuis and Neisseria lactamica. These probes
developed the multifunctional popcorn-shaped iron magnetic were purposefully prepared through the immobilizing antibody

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 229
Figure 15. One-step method for the preparation of AuNPs@MS to serve as SERS filter like substrate.124 (Adapted with permission
from Ref. 124. Copyright 2014 Elsevier.)

Figure 16. (a) Preparation sequence of NAEBs. (b) TEM images, and (c) absorption spectra of AuNPs and various Raman-reporter-
labelled NAEBs.125 (Reprinted with permission from Ref. 125. Copyright 2014 Wiley-VCH.)

onto nanoaggregate-embedded bead (NAEBs) surfaces and suit- ing a similar strategy, reported the SERS-based detection and
ably coated with silica and AuNPs. (Figure 16). Each NAEB killing of E. coli using an antibody coupled gold nanopopcorns
provides highly enhanced Raman signals due to the existence attached to SWCNTs.127
of well-defined plasmonic hot spots at the interface between Capture and detection technique has been gaining momen-
AuNPs enabling efficient single bacterium detection.125 tum recently owing to their selective sensing capability. Najafi
Lin and Hamme developed a SERS active nanohybrid and co-worker established a combined nano-immunomagnetic
system by coupling antibody conjugated Raman reporter separation capture and SERS detection method for E. coli
labeled gold nanoparticles to single-walled carbon nanotubes O157 in a liquid sample (apple juice). Antibody-functionalized
(SWCNT). The nanohybrid system was used to selectively iron oxide-gold bimetallic nanoparticles were employed for the
detect and eradicate multiple drug resistant Salmonella separation, concentration, and detection of E. coli with a detec-
(MDRS) typhimurium DT104 bacteria. 670 nm light source tion limit of 102 CFU/mL.128 On the other hand, Szymborski
was used for the photothermal killing of bacteria by creating and co-worker prepared a new class of SERS substrate by
irreparable damage to more than 99% Salmonella DT104 at the sputter coating 90 nm gold on top of an electrospun polymer
concentration of 105 CFU/mL.126 The same group, by follow- mat. The substrate showed a good SERS enhancement factor in

230 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
fluorescence microscopy measurements. The SERS measure-
ments were efficient enough for the detection of a single bacte-
rium. A limit of detection of 210 CFU mL¹1 using a portable
Raman system was obtained within a rapid DEP capture time
of ³50 s.60
A Graphene-AuNPs nanohybrid system was successfully
developed by Mevold et al. and subsequently demonstrated the
SERS-based sensitive detection of bacteria. In their approach,
the authors first prepared poly (diallyldimethylammonium
chloride) (PDDA) functionalized graphene nanosheets. Citrate
protected AuNPs were adsorbed onto the surface of graphene-
PDDA sheets facilitated by electrostatic interaction. The char-
acterization results showed that the particle size of AuNPs was
in the range of 15­20 nm immobilized on the graphene-PDDA
Figure 17. Preparation procedure of QSY 21 derivatized sheets, while the zeta potential value of AuNPs/graphene-
IO­Au NOVs as nanotags toward SERS measurements and PDDA ratio was found to be 7.7­38.4 mV. Finally, AuNPs/
their attachment to E. coli bacterial cells.60 (Adapted with graphene-PDDA nanohybrid was successfully used for the
permission from Ref. 60. Copyright 2015 The Royal sensitive detection of S. aureus.102
Society of Chemistry.) 4.2 Silver Nanostructures for Pathogen Detection.
Nanostructures based on silver (Ag) have been given special
the order of 106. The bacteria from the contaminated sample is emphasis and find extensive applications in pharmaceutical,
adsorbed on gold coated polymer mat by filtering the solution food, health, energy sectors etc. owing to their intriguing prop-
through the SERS mat using a filter funnel, and subsequently, erties as compared to other noble metals. This attracted a para-
SERS measurement was carried out. This new class of SERS mount importance in the field of sensing. Silver nanoparticles
substrate was able to selectively detect E. Coli and S. aureus in (Ag NPs) prepared through numerous methods including
contaminated water and blood samples.129 chemical, physical and biological procedures have been suc-
Cao and co-workers also successfully demonstrated the pre- cessfully utilized for human pathogens. Liu et al. used silver
paration of large-area SERS active substrates using porous nanoparticles grown on porous anodic alumina templates as a
polymer monolithic layers. A porous polymer layer was highly reproducible SERS platform and successfully demon-
initially formed on the glass surface through a simple photo- strated the label-free and rapid detection of Staphylococcus
initiated polymerization process using glycidyl methacrylate Aureus, Klebsiella Pneumoniae, and Mycobacterium Smegma-
and ethylene dimethacrylate as the starting material. Followed tis.131 Multivariate data analysis techniques such as PCA, linear
by, the thiol groups were introduced onto the surface of the discriminant analysis (LDA), clustering analysis, and SVM
porous polymer using cysteine molecules, which were then were used to differentiate the various bacterial species. Negri
exploited to uniformly adsorb the preformed gold nanoparti- et al. developed novel Ag nanorod uniform arrays by oblique
cles which eventually resulted in a sensitive and reproducible angle vapour deposition (OAD) and used as a robust platform
large area SERS substrate. The resultant strong SERS signal of for the rapid SERS detection of various pathogenic bacterial
E. Coli demonstrated the sensitivity of prepared large area species such as E. coli, E. coli O157: H7, E. coli DH 5¡,
SERS substrate.130 S. aureus, S. epidermidis, and S. typhimurium 1925-1, a poul-
In 2014, Madiyar et al. reported a synergistic approach for try isolate.132 This multivariate statistical method was em-
the concentration, detection and kinetic monitoring of E. coli ployed to differentiate the various bacterial species. Sundaram
using an integrated system which is the combination of nano- et al. demonstrated the SERS detection of S. Typhimurium,
structured dielectrophoresis (DEP), microfluidics and nanotag- E. coli, S. aureus and L. innocua from chicken wash using
labelled SERS. First, a microfluidic chip with nanoelectrode biopolymer encapsulated AgNPS deposited on a mica sheet as
array was developed using vertically aligned carbon nanofibers a SERS platform.133 Principal component analysis (PCA)
(VACNFs). SERS nanotags were prepared by functionalizing model was utilized to classify the foodborne bacteria types.
the iron oxide-gold (IO-Au) core-shell nanoovals (NOVs) of Xiao et al. reported an interesting single step self-referencing
³50 nm size with QSY21 Raman reporter and an antibody method for the SERS-based detection of pathogenic bacteria
specific towards E. coli DHα5 cells (Figure 17). Then, SERS using silver nanocubes as Raman enhancer.134 In the reported
tags and the E. coli DHα5 cells containing solution was mixed self-referencing scheme, SERS signal after target binding was
together in the microfluidic chip while applying an AC voltage referenced against blank Raman tag with no washing/sepa-
between VACNFs and the transparent ITO top electrode. Here, ration required to a sensitivity of 102 CFU/mL. Cheng et al.
the ITO surface acts as a cover slip for the microfluidic chip demonstrated a novel and simple dielectrophoretic approach to
and as an electrode material to complete a DEP device. The assembling microparticles to form nanogaps which are then
applied potential concentrates the bacterial cells on top of the used to trap the 20 nm silver nanoparticles rapidly.135 This
VACNFs which are in turn bound by the SERS nanotags due approach was successfully utilized in the bead-based SERS
to selective immunochemistry reaction. The SERS signal was for the rapid identification of bacteria from diluted blood.135
recorded on a confocal Raman microscope and a portable Cho et al. demonstrated a simple, fast and sensitive tech-
Raman probe while DEP capturing and was confirmed with nique (10 CFU/mL) for the detection of E. Coli O157:H7 strain

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 231
Figure 18. Plausible mechanism showing Ag-SP and Ag-SP-S contact with hydrophilic and hydrophobic microbes.137 (Reprinted
with permission from Ref. 137. Copyright 2014 American Chemical Society.)

using membrane filtration and silver intensification steps along Initially, silver ions were adsorbed onto the cell wall of bacteria
with SERS. Prior to SERS measurement, the target bacteria followed by reduction with hydroxylamine hydrochloride to
were captured and separated by two different antibody bear- form AgNPs on the cell wall surface. The SERS signal of
ing magnetic nanoparticles and Raman reported labeled gold bacteria varied with respect to the zeta potential of the bacterial
nanoparticles.136 cell wall. When the cell surface became more negative, the
Ho et al. reported a tricomponent nanohybrid system com- SERS signal increased which is attributed to the adsorption of
posed of exfoliated nanoscale silica platelets (SP) stabilized more silver ions on the cell surface. With this approach, the
by silver nanoparticles (Ag) and subsequently encapsulated Raman signal of bacteria was enhanced 30-fold over that of
by non-ionic surfactant poly (oxyethylene) alkyl ether. This mixed colloid-bacterial suspension. The assay time and reac-
hydrophobic nanohybrid system was then used to selectively tants volume needed to quantify bacteria in a real environment
detect hydrophobic bacteria such as E. coli and Mycobacterium was found to be 10 min and 1 mL, respectively. Besides, a
smegmatis via SERS technique (Figure 18).137 droplet of 3 ¯L sample is more than sufficient for every SERS
Betancourt et al. reported a layer-by-layer polyelectrolyte measurement. Furthermore, the authors used this approach
encapsulation of Mycoplasma pneumoniae cells within Ag NPs combined with hierarchy cluster analysis to differentiate 3
in a matrix of poly (allylamine hydrochloride) and poly strains of E. coli and 1 strain of S. epidermidis.130
(styrene sulfonate) and subsequently detected the bacteria by In a similar fashion, Chen et al. reported the formation of
SERS technique. Multivariate distribution of the Raman spec- AgNPs on the surface of bacterial cell wall using the com-
tra distinguished the 3 strains of M. pneumoniae with high spe- bination of Triton X-100, cell membrane disruption reagent,
cificity.138 Sivanesan and co-worker developed a silver­gold and sodium borohydride. In a typical experiment, the bacterial
bimetallic SERS platform by electro-deposition method and cells were mixed with 0.1% Triton X-100 for 5 min and then
subsequently coated with the antibiotic vancomycin to selec- incubated with sodium borohydride for about 5 minutes before
tively capture bacterial species from a blood sample. This kind the addition of silver nitrate solution. The pre-treatment of
of novel antibody functionalized bimetallic SERS substrate bacterial cells with Triton X-100 resulted in the disruption of
successfully differentiated four different pathogenic bacteria, the cell wall and released the intracellular matrix allowing the
namely E. coli, S. enterica, S. epidermidis, and B. megaterium successful discrimination of common disease-causing bacteria
from blood samples.139 Xiaomeng and co-workers developed a including E. coli, P. aeruginosa, Methicillin-resistant S. aur-
sensitive SERS substrate by growing silver nanorod arrays on eus, L. monocytogenes and L. innocua. By using L. innocua as
glass substrates. The substrates were fabricated by the oblique a model sample, the limit of detection of the assay was found to
angle deposition technique using an electron beam evapora- be 103 CFU/mL. The proposed analytical method proves to be
tion method. The as-prepared SERS substrate was used to a rapid tool for selective and label-free identification of
detect one of the major biomarkers of P. aeruginosa, pyocyanin pathogenic bacterium.141
at the very low concentration of 5 ppb or 2.38 © 10¹8 mol/L Mycoplasma pneumoniae is a cell wall-less bacterial patho-
in both aqueous solutions and spiked clinical sputum samples. gen of the human respiratory tract accounting to more than 20%
Most importantly, this technique would allow the dynamic of all community-acquired pneumonia (CAP). A silver nanorod
monitoring of pyocyanin excretion during the growth of array based SERS platform was developed for the detection of
P. aeruginosa.140 M. pneumoniae with high specificity and sensitivity in simu-
Ag NPs were also effectively utilized for the detection of lated and clinical throat swab samples. The methodology was
bacteria from drinking water. Zhou and co-workers demon- able to differentiate the two major genotypes of M. pneumo-
strated the SERS-based detection of living bacteria in drinking niae. The lowest possible detection limit of 5.3 « 1 cells/¯L
water by synthesizing AgNPs on the cell wall of bacteria. was also achieved.142

232 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
Figure 19. Pictorial representation of step by step sensor chip development.144 (Reprinted with permission from Ref. 144.
Copyright 2014 The Royal Society of Chemistry.)

Kumar and co-workers reported a method to fabricate highly the surface of silicon wafer facilitates the reduction of silver
sensitive Ag nanorod (AgNRs) array based SERS substrates ions and subsequently leads to the formation of AgNPs. The
atop a poly(dimethylsiloxane) (PDMS) substrate for the sensi- SERS substrate was then functionalized with 4-mercaptophen-
tive detection of P. aeruginosa. To do this, a suspension was ylboronic acid (4-MPBA), where the boronic acid group acts
coated onto the AgNRs and grown on a PDMS substrate. This as a bacteria binding moiety by reversibly binding to the wall
AgNR-PDMS buckled system potentially increase the number of bacteria. Further, the benzene ring of the MPBA serves as
of active spots thereby providing improved entrapment of a marker band whose SERS intensity increases upon bacte-
P. aeruginosa. It is to be noted that this buckled AgNR-PDMS ria binding. The primary advantages of the fabricated SERS
array system showed 11-fold enhancement in the Raman signal chip include outstanding reproducibility (standard deviation
as compared to that of AgNR-PDMS film. This remarkable <11.0%), bacterial-capture efficiency (60%) at low concentra-
enhancement in SERS can be assigned to the formation of tions (500­2000 CFU/mL), a low detection capability (1.0 ©
dense active hotspots surrounding the AgNRs and improved 102 cells/mL), and high antibacterial activity (97%).145
bacterial interaction with the metal surface. These modified Wu et al. developed a combined SERS and chemometric
AgNR-PDMS array substrates can potentially be used in analysis method for the differentiation and classification of
chemical and biological sensing applications.143 bacteria. A sensitive SERS substrate was prepared by fabricat-
In 2015, Srivastava et al. reported a highly sensitive SERS ing silver nanorods (AgNR) on glass substrates by oblique
nanobiosensor chip for the specific and quantitative detection angle deposition (OAD) in a custom-designed electron beam
of E. Coli using T-4 bacteriophages as the target capturing evaporation (e-beam) system. The AgNR substrates were
agent (Figure 19). The authors employed glancing angle depo- coated with vancomycin (VAN) and subsequently used for
sition (GLAD) to prepare plasmon enhanced and SERS active the SERS analysis of 27 different isolates from 12 species. The
nanosculptured thin films (nSTFs) of silver on a silicon (Si) VAN AgNR substrates produced reproducible SERS spectra for
wafer. Then, T-4 bacteriophages specific towards E. Coli were the bacteria. By taking advantage of variation in the surface
covalently immobilized on the chip surface. It was revealed protein of each bacteria, the differentiation of bacterial species
that the fabricated sensor was found to be accurate detection was demonstrated by using chemometric analyses of the ob-
of E. coli with ultra-small concentrations of bacteria down to tained SERS spectra. Further, the authors performed an analyti-
the level of a single bacterium. Further, the sensor was very cal step within the species cluster to be able to differentiate
accurate and selective towards E. Coli in the presence of other serotypes and strains. The proposed method was also able to
bacterial species such as Chromobacterium violaceum, Para- discriminate gram-positive and gram-negative bacteria with
coccus denitrificans and Pseudomonas aeruginosa.144 This high sensitivity and specificity and accuracy for the prediction
was possible because of utilization of GLAD and enhanced of actual test samples.146
plasmonic nSTFs of silver coated on Si towards the realization Zhou et al. reported a label-free in-situ method for the
of Raman bands. identification of live and dead bacteria by SERS. These new
In 2015, Wang et al. developed a new multifunctional SERS nanostructures were prepared by coating AgNPs on the surface
chip capable of capturing, discriminating, and deactivating the of bacteria (Bacteria@AgNPs) and their utility for rapid count-
pathogenic bacteria. The AgNPs decorated SERS chip was ing of live and dead bacteria by SERS. Interestingly, the
grown on a silicon wafer by etching the wafer in hydrofluoric AgNPs bind only to the live bacteria and not to the dead bacte-
acid followed by AgNPs deposition. It was found that Si-H on ria. Thus, the authors were able to selectively detect the gram-

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 233
negative bacteria in a suspension of live and dead bacteria.147 The concentration of S. typhimurium with a limit of detection
Duan et al. reported a SERS-based aptasensor for quantitative of 15 cfu/mL can be achieved with this technique. The meth-
detection of pathogenic bacteria.148 In this approach, Au@Ag odology was also extended to detect S. typhimurium species in
core/shell nanoparticles (NPs) functionalized with target bac- food samples.
teria specific aptamer 1 (apt 1) were employed for both the 4.3 Carbon Nanomaterials for Bacteria Detection. 4.3.1
SERS substrate and also the capturing agent. A second type of Carbon QDs: Nanostructured biosensors based on carbon
aptamer (apt 2), functionalized with X-Rhodamine (ROX) as based nanomaterials (such as carbon nanotubes, graphene, ful-
Raman reporter molecule, which is also specific towards the lerenes) have been constantly utilized toward bacterial detec-
target bacteria and simultaneously acts as a recognition element tion due to their remarkable properties including aspect ratio
was also used. When both the Au@Ag-apt 1 and apt 2-ROX and surface modification. Table 1 summarizes the details of the
encounter the target S. typhimurium, all three form a complex carbon nanomaterials based detection of bacteria with their
structure where the bacteria is sandwiched between both the linear concentration range and limit of detection. A novel
aptamers. Thus, the proximity of Au@Ag NPs and ROX results hybrid detection strategy, utilizing carbon dots magnetized with
in an improved SERS signal which is directly proportional to iron oxide (Fe3O4) nanoparticles and functionalized using
the concentration of bacterial species present in the mixture. nitrogen rich chitosan was developed to detect S. aureus and

Table 1. Summary of carbon nanomaterials based detection of bacteria with their linear concentration range and limit of detection.

Target Nanomaterials Detection Linear range LOD


Reference
pathogen utilized strategy (CFU/mL) (CFU/mL)
E. coli O157:H7 Holey reduced graphene oxide FM, SEM and AFM ® 803 18

(hRGO)
Salmonella Paratyphi A SWCNT-DNAzyme assembly FM 103­107 103 167

E. coli K-12 SWCNT ­ immobilised EC 102­105 102 168

specific antibodies
Heat killed O157-H7 Chitosan-MWNT-SiO2/ EC 4.12 © 102­ 250 169

THI nanocomposite 4.12 © 105


Neisseria gonorrhoeae PANI-nFe3O4-CNT DPV 1 © 10¹19 M­ 1 © 10¹19 M 170

nanocomposite 1 © 10¹6 M
S. aureus ZrO2 and graphene DPV 1 © 10¹13­ 3.23 © 10¹14 M 159

nanocomposite 1 © 10¹6 M
Heat killed E.coli Au-SiO2-CHI-SH/Fc/C60 CV 3.2 © 101­ 15 171

O157:H7 nanocomposite 3.2 © 106


E. coli O157:H7 and SWCNT-aptamer assembly Electromotive force 0.2 © 10¹6 0.2 172

S.aureus
V. parahaemolyticus,
Soot CNP ­ dye loaded Confocal laser scanning
S. aureus and 102­106 25­50 173
aptamers microscopy
S. Typhimurium
Uropathogenic E. Coli GCE/f-MWCNT-Chit@Th CV 102­109 50 174

V. parahaemolyticus and Quantum dot-CNP-aptamer FM 50­106 25, 35 175

S. typhimurium
Aeromonas MWCNT­Chi­Bi-PbNP- DPV 0.3 © 10¹12 M­ 1 © 10¹14 M 176

aptamer 0.01 © 10¹12 M


E. coli mAb-AuNP@f3-SWCNT SERS 102­106 100 127

B. anthracis ssDNa/AuNp/Gr/GCE DPV 10 ­10¹7 M


¹12
2 © 10¹13 M 177

Enterobacter sakazakii AuNP/ERGO/SPCE-HRPab CV 103­109 119 178

S. aureus Graphene-modified Piezoelectric frequency 4.1 © 101­ 41 162

IDE-SPQC-aptamer shift 4.1 © 105


Listeria monocytogenes QD­GO nanoplatform FM 102 to 106 fg/¯L 100 fg/¯L 163

Shewanella oneidensis Ab/AuNPs/BSA/GO Anodic stripping 70 © 107 to 12 164

voltammetry 7 © 107
E. coli Gr-Pt hybrid NP-aptamer DPV 1.5 © 10¹7 and 1.12 © 10¹9 M 165

2.25 © 10¹6 M
M. tuberculosis rGO-AuNP-aptamer CV 1.0 © 10¹15 and ® 179

1.0 © 10¹9 M
FM ­ fluorescence microscopy, SEM ­ scanning electron microscopy, AFM ­ atomic force microscopy, CV ­ cyclic voltammetry,
DPV ­ differential pulse voltammetry, EC ­ electric current.

234 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
E. coli. Fluorescent spectroscopy, and MALDI-MS were used coated graphene nanosensor was shown to have stronger
for bacterial detection, and results show a detection limit of affinity/bonding for the single bacterium as compared to the
3 © 102 and 3.5 © 102 for both bacteria respectively.149 Natural only graphene nanosensor.155
biomass was used to develop carbon dots which can act as A highly sensitive thermally reduced monolayer graphene
fluorescent probes for bacterial detection. Punicagranatum, oxide (TRMGO) FET immunosensing device built by ultra-
when subjected to HTC in an autoclave, led to the formation of sonic incorporation of graphene oxide monolayer sheets on
carbon dots which were then used for fluorescent imaging of FET functionalised gold electrodes can detect E. Coli between
bacterium Pseudomonas aeruginosa. This method is very concentration ranges of 10 CFU per mL to 103 CFU/mL. Scan-
inexpensive as the final material is used without any chemical ning electron microscopy (SEM) and atomic force microscopy
or magnetic treatment and the process also signifies the (AFM) data suggest the semiconducting role of TRMGO sheets
importance of natural biomass for the development of suitable which get plugged into the electrode gaps. The device senses
materials for bacterial detection.150 Another interesting and the response from specific binding of E.coli cells to anti E.coli
recent study on bacterial detection was based on amphiphilic antibodies coated/immobilised on its surface, and its sensitiv-
carbon dots. The detection of different bacteria relies on the ity of detection was shown to be more when tested for E.coli
affinity of these dots towards the surface of the bacterium cell. O157:H7 (7.3%) as compared to E.coli DH5α (1.4%) and
When tested for Escherichia coli MG1655 wild type, Salmo- Dickeya dadantii 3937 (1.3%).156
nella typhimurium (strain ATCC14028), Pseudomonas aerugi- An antimicrobial peptide, magainin I functionalised holey
nosa PAO1 wild type, Bacillus cereus, and PET28a-FtsA-GFP reduced graphene oxide (hRGO) based FET device was report-
strains, the intensity and spectral positions in fluorescence ed for gram negative bacteria with a detection limit of 803
spectra were different for all bacteria. This indicates the ability CFU mL¹1 for E. Coli O157:H7. The presence of abundant
of such carbon dots to distinguish among the various bacterial oxygen functionalities along with rich electronic properties
strains.151 A combination of the microwave-synthesized carbon accounted for its superior bacterial detection when compared
dots and antibiotic vancomycin provides a pathway for fluores- with three other similar carbon nanomaterials, reduced GO,
cent detection of Staphylococcus aureus showing a linear range pristine SWNTs and oxidized SWCNTs. Fluorescence micros-
relation between 3.18 © 105 and 1.59 © 108 CFU mL¹1 and copy, SEM, and AFM were used to visualize the bonding
detection limit of 9.40 © 104 CFU mL¹1. This probe shows between FET device and bacterium. From AFM images, it is
higher affinity for gram positive bacteria which is due to the quite clear that bacterium is well attached to the surface of
ligand receptor interactions between the vancomycin and hRGO.18
bacterial cell walls. Recovery experiments with orange juice A selective fluorescent aptamer-monolayer GO combined
demonstrate the capability of this sensing approach in real- bacterium sensor was developed with a detection limit of 100
world detection of Salmonella aureus.152 CFU mL¹1 and a range of 103 to 108 CFU mL¹1 for Salmonella
4.3.2 Graphene: An aptamer free sensing platform was typhimurium. Systematic evolution of ligands by exponential
developed by incorporating chitosan in reduced graphene enrichment (SELEX) procedure was used to select single strand
sheets and then electrodepositing a film on tin oxide electrode DNA specific to S. typhimurium which was then dissolved in
that can detect the charge transfer resistance produced by the phosphate buffer and GO to make a standard testing solution.
presence of sulfate reducing marine bacteria (SRB). The The addition of target bacteria to fluorophore 5-carboxyfluores-
impedimetric measurements using electrochemical impedance cein (FAM) ­aptamer ­GO sensor solution results in fluores-
spectroscopy (EIS) demonstrates the capability of the sensor cence quenching of FAM-aptamer by GO which is detected by
within a linear range of 1.0 © 104 CFU mL¹1 to 1.0 © 108 the spectrophotometer.157
CFU mL¹1. This device is quite selective, and the further A polymethyl methacrylate (PMMA) fabricated graphene
enhancement is possible by immobilizing aptamers or anti- device when functionalized with a linker molecule immobilizes
bodies on the surface.153 the E. coli antibodies on the surface can successfully iden-
A label-free SERS probe built by attaching nanopopcorn to tify the bacterium E. coli at a very low concentration of 10
graphene oxide can detect methicillin-resistant Staphylococcus CFU mL¹1. One end of the linker molecule (1-pyrenebutanoic
aureus (MRSA) at a low concentration of 10 CFU mL¹1. An acid succinimidyl ester) forms π-π interactions with graphene,
enhancement in SERS signal was observed due to the assembly and the other one is connected to the antibody. Conductance
of graphene to nanopopcorn which is attributed to the large measurements show a linear relationship to E. coli concen-
surface area with high aspect ratio of graphene and lighten- tration within a range of 0­105 CFU mL¹1 and as the antibody
ing rod effect and surface plasmon excitations produced in on the FET device is specific to E. Coli, it shows no conduc-
the nanopopcorn. During the detection process using aptamer tance response for P. aeruginosa in the same concentration
modified SERS probe, it was found that the MRSA bacteria range. This FET device also responds very well to real-time
tends to form aggregates inside the graphene oxide sheets.154 detection of glucose metabolism induced by bacteria which
A wireless graphene silk nanosensor was synthesized and highlights its potential application for real sample analysis.
transferred onto tooth enamel using water-soluble silk. It was E. Coli aided glucose metabolism studies showed that the
reported that the dissolution of silk is faster on muscle than device picked up the response only when glucose and bacteria
tooth enamel. This thin film nanosensor on tooth enamel, when are present together and not with glucose alone.158
functionalized with antimicrobial peptides (AMP) biorecogni- 4.3.2.1 Graphene-Modified Electrode; Graphene-coated
tion molecules and fitted with inductive coil becomes ex- carbon electrodes, when electrodeposited with zirconium
tremely sensitive and can even detect a single bacterium. AMP- dioxide (ZrO2), can effectively immobilize a single strand

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 235
DNA probe sequence on the surface for rapid and sensitive
detection of S. aureus DNA sequence with a detection limit
of 3.23 © 10¹14 M and offers a detection range of 1 © 10¹13 to
1 © 10¹6 M. The high surface area along with conductive
environment provided by graphene and high affinity of ZrO2
for DNA sequence together makes a perfect combination for
efficient immobilization of DNA sequences on the surface.159
A metal hybrid nanocomposite constructed using electro-
deposition of platinum nanoclusters (nPt) on Pt/Ir electrode
with a surface coated with GO can easily incorporate In1A
aptamers on its surface, and the assembly can be utilized for
successful detection of L. innocua using cyclic voltammetric Figure 20. (a) rGO nanocomposite membrane (b) incorpo-
and impedimetric measurements. High surface area, conduc- ration of the AuNP film, (c) immobilisation (d) combina-
tivity and electrocatalytic properties of the biosensor make it tion, and (e) detection of target DNA.166 (Adapted with
possible to detect bacterium at a low detection limit of 100 permission from Ref. 166. Copyright 2014 The Royal
CFU mL¹1 with an amplified signal.160 Society of Chemistry.)
Anti- Vibrio parahaemolyticus (VP)/N-(4-aminobutyl)-
N-ethylisoluminol (ABEI) immobilized on the surface of a of 7 © 106 CFU mL¹1 was measured. S. oneidensis gave a very
nano Fe3O4 - graphene oxide (nano@Fe3O4@GO) combination high response as compared to the other two bacteria demon-
and then coated onto a glassy carbon electrode offers a stable, strating the specificity of the sensor. The analysis results for
selective, highly sensitive and rapid sensing capability for the river water spiked with the bacterium provided a linear corre-
detection of Vibrio parahaemolyticus in seawater and seafood lation within a range of 2.31 © 103 to 2.43 © 107 CFU mL¹1
samples. The detection ability of this assembly was recorded and can potentially be used for such practical samples.164
using electrochemiluminescence of ABEI. The presence of a An electrochemical genosensor prepared by modifying
two-dimensional structure and high conductivity of nano Fe3O4 glassy carbon paste electrode (GCPE) with graphene-platinum
- GO increases the electrochemiluminophorisity of ABEI and hybrid nanoparticle (Gr-Pt hybrid NP) and then immobilized
anti-VP, thereby enhancing the detection sensitivity of the with ssDNA on the surface can detect guanine oxidation
sensor. The sensor is capable of detecting VP within a range signals from E. coli oligonucleotides before and after hybrid-
of 10­108 CFU mL¹1 with a detection limit of 5 CFU mL¹1 and ization at the surface of the assembly. Analytical signals from
5 CFU g¹1 for seawater and seafood respectively.161 differential pulse voltammetric exhibited a linear range of
Another aptamer-based framework for detection of S. aureus detection between 1.5 © 10¹7 and 2.25 © 10¹6 M and a LOD
is constructed by cross-linking graphene to interdigital Au of 1.12 © 10¹9 M.165
electrodes connected to a series electrode piezoelectric quartz An electrochemical sensing platform developed for the
crystal (SPQC). The attachment of S. aureus aptamer to the specific DNA insertion sequence IS6110 of M. tuberculosis
surface of graphene offers binding sites for the corresponding involving components such as aptamer, rGO and AuNPs can
antigens of the bacterium. Frequency shifts in the oscillation of detect the bacterium in a linear range of 1.0 © 10¹15 and
SPQC produced due to antibody-antigen reaction were found to 1.0 © 10¹9 M (Figure 20). The combination, when coated on
be linear in the range of 4.1 © 101­4.1 © 105 CFU mL¹1 with GCE, could enhance the effective surface area, conductivity,
the lowest shift detected at 41 CFU mL¹1. The bacterial detec- and electron transfer capability of the electrode and was con-
tion limit for a real sample of milk (41 CFU mL¹1) is in 100% firmed by the increase of peak current in cyclic voltammetry
agreement with the plate count value indicating the potential measurements. Further, the attachment of capture probes and
application of such a sensor in a real world scenario.162 then target probes led to decrease in peak current indicating the
A fluorescence sensing platform generated by a combination success of the detection process. The sensitivity of the sensor
of GO-QD with surface immobilized specific ssDNA possesses was amplified using an Au-PANI nanocomposite which dis-
excellent ability to detect multiple genes of Listeria mono- plays good biocompatibility and outstanding electrochemical
cytogenes. High amplification using linear-after-the-exponen- activity.166
tial (LATE) ­ polymerase chain reaction (PCR), quenching A combination of a glass microfluidic system and aptamer
effect of GO, and high quantum yield of QDs make it possible functionalized GO offers a straightforward and rapid method
to detect bacterium DNA at a low concentration of 100 fg/¯L for simultaneous detection of Staphylococcus aureus and
in 1.5 hours within a range of 102 to106 fg/¯L.163 Salmonella enteric with a linear detection range from 104­
Bovine serum albumin (BSA)-modified GO when loaded 106 CFU mL¹1 and 42.2­675 CFU mL¹1 for each bacterium,
with AuNP’s can immobilize antibodies on the surface for respectively (Figure 21). PDMS was used to set up a top layer
highly sensitive electrochemical detection of Shewanella with- with microchannels for reagent delivery and middle layer with
in a range of 70 © 107 to 7 © 107 and a detection limit of microwells was used for incubation and detection. The chro-
12 CFU mL¹1. This conjugate can reduce silver ion to metallic matographic paper was introduced in microwells for adsorption
silver which can be quantified using anode stripping voltam- of aptamer-GO assembly, and the whole assembly sits on a
metry. To validate the specificity of the sensing platform glass bottom layer. A detection limit of 11 CFU mL¹1 and
for S. oneidensis, stripping response of two other bacteria linear range of 9.4­150 CFU mL¹1 was detected for L. Acid-
B. subtilis, E. coli and S. oneidensis in a similar concentration ophilus using fluorescence measurements.180

236 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
with a detection limit of 10 CFU mL¹1. This assembly is stable
due to the linkage of ssDNA with rGO using ³-³ stacking and
AuNP using Au-S bonding. EIS signals for this system were
significantly enhanced due to the high electronic and surface
properties of rGO and AuNPs, respectively. When tested for
three different bacteria, E.coli, Salmonella and S. aureus, the
probe showed excellent specificity towards S. aureus with ¦Ret
value of nearly 200 ³ as compared to only 20 ³ for other two
bacteria.123
DNA sequence of Klebsiella pneumonia carbapenemase
(KPC) can be precisely detected by an electrochemical bio-
sensor constructed using ssDNA, graphene, GCE and Au-NP
(ssDNA/Au-NP/Gr/GCE). The synergistic effect produced by
combining Gr/GCE with Au-NP led to higher conductivity,
electrochemical and enhanced surface properties which help in
detecting bacteria within a linear range of 10¹12­10¹7 M at a
detection limit of 2 © 10¹13 M using differential pulse voltam-
metry.177 E. sakazakii, a foodborne bacterium can be detected
Figure 21. Scheme showing PDMS/paper hybrid micro- using a screen-printed carbon electrode (SPCE) modified with
fluidic system using aptamer-functionalized GO bio- ERGO-AuNP and [BMIM]PF6-HRP-anti-E. Sakazakii on the
sensors. (a) layout of microfluidic biochip, (b) and (c) surface. SEM images showed the formation of 100 nm size
principle of the one-step ‘turn-on’ detection approach.180 AuNP on the surface of ERGO while using AFM it was con-
(Adapted with permission from Ref. 180. Copyright 2013 firmed that HRP antibody had been successfully immobilized
The Royal Society of Chemistry.) on the AuNP/ERGO system. This sensor platform provides a
high surface area, conductivity and electrocatalytic function
A magnetic nanohybrid biosensor (GMCS) developed by which helps to detect bacterium within a range of 103 to 109
combining GO and magnetic chitosan can detect pathogenic CFU mL¹1 reaching the lowest detection limit of 119 CFU mL¹1
bacteria Pseudomonas aeruginosa and Staphylococcus aureus as shown using cyclic voltammetric measurements.178
using matrix-assisted laser desorption/ionization (MALDI-MS) Europium loaded graphene quantum dots (Eu-GQD) of size
and fluorescence spectroscopy. GMCS interacts with bacteria 3 and 10 nm with the enhanced surface to volume ratio can
via hydrogen bonding, hydrophobic and electrostatic interac- adequately bind Bacillus anthracis (B. anthracis) at very low
tions, acid­base, π-π, and polar functional groups interactions concentrations of 10 pM which are six orders of magnitude less
which make it highly sensitive for fluorescent analysis of blood than the infectious dose of the bacterium at 60 ¯M. Fluores-
samples.181 cence measurements showed that the smaller Eu-QD (3 nm)
A potentiometric aptamer ­ GO/RGO biosensor was con- with larger surface to volume ratio is 1.5 times faster (5.2 s) in
structed for single cell detection of Staphylococcus aureus. detecting B. anthracis when compared to 10 nm size dots (8 s).
When coated on GCE, GO and RGO can be covalently or Such a GQD platform has potential for the effective detection
non-covalently functionalized with corresponding S. aureus of pathogenic Bacillus Anthrax.184
aptamers. The results proved that the covalently functionalized 4.3.3 Carbon Nanotubes: An electrochemical impedance
sensors provide the best results with a detection limit of 800 immunosensing platform of AuNPs and poly(amidoamine)-
CFU mL¹1 as compared to non-covalent functionalised sam- multiwalled carbon nanotubes-chitosan nanocomposite film
ples which gave a poor 107 detection limit. Also, the device is modified GCE (AuNPs/PAMAM-MWCNT-Chi/GCE) pro-
very selective towards S. aureus, and this is reflected by no vides a sensitive mechanism for detecting Salmonella typhi-
potentiometric response for E. coli, a gram negative bacteria murium in a linear range of 1.0 © 103 to 1.0 © 107 CFU mL¹1
and L. casei, a gram positive bacteria testing.182 with a LOD of 5.0 © 102 CFU mL¹1. The interaction of bacte-
An impedance measuring device for detection of Salmonella rium with immobilized antibodies on the sensing probe causes
was built using a combination of reduced GO and COOH- change in the electron transfer resistance which can be mea-
modified MWNTs coated on a GCE. The electrode surface was sured using electrochemical impedance spectroscopy. The real
further modified with covalently bonded amide linkage to an sample testing of the sensor was done using fat-free milk, and
anti-salmonella aptamer. The rGO-MWCNT nanocomposite the recoveries of the bacteria were within a range of 94.5­
is proposed to be connected through ³-³ interactions and 106.6% indicating the potential use of this sensor in prac-
provides excellent electronic properties, a high surface area tical utilities. Drastic increase in electron transfer resistance
and binding sites for the accommodation of anti-salmonella (806 ohms) with S. typhimurium as compared to E.coli and
aptamer. The sensor is able to detect spiked in chicken within S. aureus (100 ohm) demonstrated the specificity of the sensing
a range of 75­7.5 © 105 CFU mL¹1 with a lower detection limit platform.185
of 25 CFU mL¹1.183 The binding of aptamer with SWCNTs in a covalent fashion
A sensing device constructed using a combination of rGO offers enhanced detection of S. aureus as compared to the non-
and AuNP and ssDNA linked to this nanocomposite is capable covalent interaction between the two. In the covalently bonded
of measuring S. aureus in the range of 10­106 CFU mL¹1 and system, a detection range of 8 © 102 to 108 CFU mL¹1 and a

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 237
Table 2. Comparison of virtual and real sample analysis for carbon based nanomaterials toward detection of pathogen.

Plate Count Analysis Real Sample Analysis


Target Linear detection Limit of
pathogen Real Linear range LOD
range detection Ref.
sample (CFU/mL) (CFU/mL)
(CFU/mL) (CFU/mL)
Salmonella Paratyphi A 103­107 103 City water 104­108 104 167

Heat killed O157-H7 4.12 © 10 ­4.12 © 10


2 5
250 Water 3.06 © 103­3.46 © 105 ® 169

Heat killed E.coli O157:H7 3.2 © 10 ­3.2 © 10


1 6
15 Synthetic stool 3.2 © 102­2.3 © 106 ® 171

E. coli O157:H7 0.2 © 10¹6 0.2 Milk/fruit juice 4­104 4 172

S. aureus 0.2 © 10¹6 0.2 Pig skin 8 © 102­108 800 172

V. parahaemolyticus, 102­106 25­50 Milk Salmon 1 © 103­0.9 © 105 ® 173

S. aureus and 0.9 © 103­1 © 105


S. Typhimurium
V. parahaemolyticus and 50­106 25, 35 Shrimp 1.1 © 103­1.2 © 105 1.1 © 103 175

S. typhimurium Chicken 0.9 © 103­1.0 © 105 0.9 © 103


Aeromonas 0.3 © 10¹12 M­ 1 © 10¹14 M Tap water 101­106 100 176

0.01 © 10¹12 M
S. aureus 4.1 © 101­4.1 © 105 41 Milk 4.1 © 101­4.1 © 105 41 162

Shewanella oneidensis 70 © 107 to 7 © 107 12 River water 2.31 © 103 to 2.43 © 107 ® 164

LOD of 8 © 102 CFU mL¹1 was observed, while the non- SWNT with PEI makes it possible to detect E. Coli K-12
covalently functionalized sensor responded to a bacterium only precisely. The reaction between antibodies and the bacteria was
at a high concentration of 107 CFU mL¹1. The number of avail- recorded as changes in the electric current which resulted in a
able binding sites on the surface is a contributing factor for linear range of 102­105 CFU mL¹1 with a detection limit of
different responses of the sensor towards the bacterium.186 102 CFU mL¹1. There is a negligible change in current when
A sensor based on the Clavanin A immobilized CNT that measured for S. aureus (7.25 nA) as compared to E. Coli
can differentiate gram positive and gram negative bacteria (65.76 nA) indicating the specificity of this sensor to E. Coli.168
was developed by Andrade et al.187 Electrochemical responses A layer by layer (LBL) methodology to construct an
using EIS showed the ability of the sensor to differentiate immunosensor for detecting heat killed Escherichia coli
Klebsiella pneumoniae, Enterococcus faecalis, Escherichia coli O157:H7 was successfully achieved using a chitosan-
and Bacillus subtilis within a range of 102 to 106 CFU mL¹1. MWNT-SiO2/THI nanocomposite and GNP multilayer films.
The development of SWNT fluorescent probes in combination ELISA was used to observe the covalent linkage of E. Coli
with M13 virus also offers a much safer, cheaper, and non- O157:H7 antibody to the surface of GNP monolayer. Current
invasive method for fluorescent optical imaging of bacterial measured by using a cyclic voltammetric method records a
infections. M13-SWNT probe can distinguish between the F- linear range of 4.12 © 102­4.12 © 105 CFU mL¹1 with a detec-
positive and F-negative strains and can be suitably tuned to tion limit of 250 CFU mL¹1. There is a significant degree of
sense specific detection of F-negative strain. Staphylococcus agreement between measurements when the sensor is tested
aureus intramuscular infections in mice were successfully for real samples of milk and water. The qualitative data using
detected with this probe using FIT. Such biologically function- milk showed 94.4% similarity, and it was also found that the
alized probes without any chemical treatment/modification quantitative analysis of water is also in good agreement with
could prove to a be a great potential in the field of bacterial the plate count data.169
detection in the future.188 A combination of PANI-nFe3O4-CNT nanocomposite and
Salmonella Paratyphi A bacteria could also be detected biotinylated nucleic acid sequence of 20 bases immobilized on
using a non-covalent assembly of SWNT, and a DNAzyme its surface using biotin-avidin coupling offers a very sensitive
labeled aptamer with a detection limit of 103 CFU mL¹1 and genosensor for the precise detection of Neisseria gonorrhoeae.
linear range of 103 to 107 CFU mL¹1. Systematic evolution of The facile electrochemical synthesis of the nanocomposite
ligands by exponential enrichment (SELEX) procedure was involves the fabrication of polyaniline (PANI)-iron oxide nano-
used to derive the single strand DNA aptamers form oligonu- particles and MWCNT on an indium tin oxide (ITO) coated
cleotides which were shown to have high affinity and specific- glass tube which was then characterized using TEM, FTIR,
ity against Salmonella Paratyphi A using fluorescent assays. XRD, and SEM. Differential pulse voltammetric measurement
When applied to the real-world situation of a water sample, it results indicate a detection limit of 1 © 10¹19 M and a detection
was found that the bacterial detection limit was 104 CFU mL¹1 concentration range of 1 © 10¹19 to 1 © 10¹6 M (Table 2).170
with a linear range of 104 to 108 CFU mL¹1. This indicates the Potentiometric aptasensors using SWCNT and specific aptam-
potential use of such materials for bacterial detection in real ers were also developed for successful detection of both gram
samples (Table 2).167 The fabrication of immobilized anti- positive (S. aureus) and gram negative (S. typhi and E. coli)
bodies streptavidin and biotin in the crossbar junction of bacteria in real samples. The assembly includes carboxylated

238 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
SWCNT deposited on a GCE and then covalently linked to different fluorescence dyes (fluorescein amidite, cyanine, and
­ (CH2)5NH2 3¤ end of aptamers on the surface establishing 6-carboxy-X-rhodamine) were attached to 5¤ end of three
stable CO-NH bonding. SWCNT provides a transducing effect aptamers, and the combination was then attached to soot carbon
which minimizes instrument noise and allows low concen- nanoparticles through ³-³ stacking interactions. Confocal laser
tration of bacteria to be detected, whereas the actual sensing scanning microscopy provided fluorescence images of the
function is performed by the specific aptamer and is recorded bacterial binding to the specific aptamers and TEM images
as changes in the electromotive force for each individual showing the soot nanoparticles. The aptasensors gave con-
bacterium. When tested for liquid samples such as semi- sistent results when the plate count was compared with a real
skimmed milk or apple juice and a solid sample of pig skin, it sample analysis using milk and salmon.173 Gold nanoparticles
was found that the aptasensors are more selective than sensitive when combined with the selective RNA sequences can further
towards bacterial detection (Table 2).172 enhance the capability of E. Coli detection by 189% as com-
It was also demonstrated that uropathogenic Escherichia coli pared to without nanoparticles. The whole assembly is sensitive
(UPEC) can be detected within a wide linear range of 102­ to electrochemical signals relying on the conductive behavior
109 CFU mL¹1 and LOD of 50 CFU mL¹1 using an immuno- of CNTs and high surface area of gold nanoparticles for
sensor constructed by immobilizing thionines dye on function- bacterial detection.192
alized MWNT-chitosan composite coated on GCE. The peak 4.3.5 Fullerenes: Fullerenes (C60) are stable caged mole-
current in the cyclic voltammetric measurement increases with cules with sp2 carbon atoms arranged in the form of hexagons
the increase in UPEC cells in the range 102­106 CFU mL¹1 and pentagons. C60 are considered promising members of the
with a determined current sensitivity of 7.16 © 0.04 ¯A. This carbon family owing to their exceptional properties such as
electrochemical immunosensor can also measure the current stable redox properties, high thermal/mechanical stability, ease
from peroxide reduction in a simulated urine sample inoculated of functionalization, conductivity etc.193 In recent years, C60,
with 109 cells of E. coli, thereby showing its applicability in derivatized C60, and its porous counterparts have been utilized
real sample analysis (Table 2).174 in energy and environmental applications.194,195 Besides, read-
A sensitive dendritic nanotip with a LOD of 103 CFU mL¹1 ers are strongly encouraged to refer to some of the articles/
was also constructed by using SWCNTs coated Si nanowires reviews based on advanced fullerene nanostructures highlight-
that was designed with the help of UV lithography and etching. ing their state-of-the-art trends and developments.196­202 In
The bacterial detection capability of the system is evident from particular, their biocompatibility and unique features (such as
the reduction in electric current when target bacteria BCG signal amplification) enable the specific application of C60 in
combine with fluorescence antibodies at the dendritic tip. Both biosensing based platforms. Based on the available literature,
fluorescent and electrical measurements showed the efficient C60 derivatives are powerful antioxidants capable of scaveng-
binding of the bacteria onto the tip.189 A biosensor construct- ing and detoxifying free radicals (reactive oxygen/nitrogen
ed by a combination of MWCNT-chitosan-bismuth complex species). They are very good photosensitizers and thus can be
with lead nanoparticles and surface attached DNA probes can utilized in photodynamic therapy to heal cancer and eradicate
also rapidly detect Aeromonas at a very low concentration of microorganisms. In one specific study, heat killed E. Coli
1 © 10¹14 M. The interaction of target bacterium DNA and O157:H7 can be detected within a concentration range of 3.2 ©
probe DNA is recorded using differential pulse voltammetry 101­3.2 © 106 CFU mL¹1 with a detection limit of 15 CFU mL¹1
signals. The probe is capable of detecting Aeromonas within a by employing an efficient sensitive and biocompatible electro-
range of 101­106 and a detection limit of 101 in real water chemical immunosensor through the immobilization of biotin
samples spiked with the bacterium.190 using C60 based biocompatible platform and enzyme function-
A rapid and selective detection of E. Coli was also possible alized Pt nanochains tracing tag. The immunosensing platform
using a sensor based on immobilization of monoclonal E. coli comprises multi-components that include C60, ferrocene (Fc),
antibodies on the surface of gold nanoparticles combined with and thiol functionalized chitosan (CHI-SH) nanocomposites
thiol terminated SWCNT’s. This sensor is capable of detect- (CHI-SH/Fc/C60). In addition, Au embedded SiO2 was also
ing bacterium in a range of 102­106 CFU mL¹1 with a lower utilized to assemble Au-SiO2-CHI/Fc/C60 nanocomposite.171 It
detection limit of 102 CFU mL¹1. The whole process used for is interesting to note the concentration value reported in this
constructing the device offers a high conductivity and plenty of work is well below the threshold limit and the proposed sys-
binding sites for monoclonal antibodies. These immobilized tem has been successfully implemented in analyzing synthetic
antibodies coated on nanohybrid structure detect E. coli and stool samples. The values obtained for this immunosensor is
form microbial clusters on the surface, and this leads to very well comparable to the conventional plate count method
enhancement in SERS.191 The strong electromagnetic and indicating they can be used in practical utilities.171
chemical effects attributable to the components of the hybrid
nanostructure significantly increase SERS-active enhancements 5. Conclusion and Future Prospects
and hence enable detection and quantification of E. coli in The field of pathogenic bacteria detection using advanced
water for environmental monitoring. nanomaterials in biosensors is continuously evolving which is
4.3.4 Carbon Nanoparticles: A multiplexed FRET system evident from significant research emphasis focussed on the
from fluorescence dyes and carbon soot nanoparticles was development and utilization of more efficient materials for the
designed for simultaneous detection of V. parahaemolyticus, fabrication of next level sensing devices which will possess a
S. aureus and S. Typhimurium with a linear range of 102­ rapid and controlled sensitivity, higher magnitudes of detection
106 CFU mL¹1 and a detection limit of 25­50 CFU mL¹1. Three levels, longer life, and relative ease of operational use. With

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 239
respect to this, advanced nanomaterials have gained paramount minimised and or identified by employing sophisticated tech-
importance in the recent past due to their unique physicochem- niques that allow only a specific and time bound nanoparticle
ical properties such as small size, high surface to volume ratio, interaction with target pathogens without any side interac-
tuneable semiconducting band gap, fast reaction rate, magnetic tions. Not only these, more research also needs to be done on
activity, optical activity, and antibacterial surface. More impor- improving the reproducibility of nanomaterial based biosensing
tantly, these materials can be suitably functionalized with devices. An integration of such advanced research in the field
relative ease for the detection of specific target pathogenic of advanced nanomaterials based biosensing should respond to
bacteria. The amalgamation of nanotechnology into the field of these obstacles/challenges which will allow for the fabrication
biosensing is based on the underlying mechanism of initial of sensing devices and further offer more specific and environ-
binding/reaction of the nanoparticles with the specific bio- mentally friendly methods with higher accuracy and precision
logical component of the pathogenic bacteria which is then to detect bacterial pathogens. Although the research and devel-
subsequently and rapidly converted into a detectable signal. opment on coupling of nanotechnology with biotechnology for
This approach is quite user friendly and the fact that nano- designing advanced bio-sensing devices for detecting patho-
technology allows the fabrication of devices with much smaller genic bacteria is gaining momentum, much work should be
sizes makes it easier for handling which is an added advantage. done in integrating the recent advancement in the nano-
In this review article, we have reviewed and summarized technology with emerging bio-nanotechnological approaches.
the recent literature reported on the detection of pathogenic For example, work on 1D, 2D and 3D nanosheets with different
bacteria by using biosensor devices fabricated using advanced elemental frameworks have been receiving a lot of attention
nanomaterials such as metal/metal oxides, magnetic core-shell these days but the applications of these nanostructures with or
nanostructures, metal selenides, metal tellurides, carbon dots, without the functionalization of biomolecules are quite limited.
quantum dots, magnetic nanostructures, graphene, etc. Graph- Focusing on these areas would bring next generation devices
ene based nanoparticles were the focus of the research inves- for the selective sensing and the detection of pathogenic
tigations in the field of biosensing for bacterial detection for the bacteria.
past few years. However, several other promising nanomate- Thus, rapid pursuance of advanced nanotechnology for
rials such as metal/metal oxides nanoparticles, quantum dots, pathogenic bacterial detection would certainly benefit from
magnetic nanoparticles and other carbon-based nanomaterials future research undertaken for the fabrication of advanced
have recently attracted great attention due to their superior biosensing devices which could be scalable for large oper-
characteristics. Therefore, more exploratory research needs to ations. The most important aspect also lies in the application
be carried out for the design of next level of biosensors based of such nano biosensing devices for analysis of pathogenic
on these nanomaterials for the selective detection of pathogens. bacteria in real samples for rapid translation into the commer-
Each class of nanomaterials discussed in this review has its cial world.
own peculiar advantages which helps in improving the qual-
itative and quantitative sensitivities for the pathogenic bacterial References
detection. For example, semiconductor based NPs exhibit ³ These authors contributed equally for this review.
unique optical properties while magnetic NPs offer the ease of 1 A. Zumla, M. Rao, R. S. Wallis, S. H. E. Kaufmann, R.
detection using NMR. On the other hand, noble metal NPs Rustomjee, P. Mwaba, C. Vilaplana, D. Yeboah-Manu, J. Chakaya,
possess high surface to volume ratio whereas carbon based NPs G. Ippolito, E. Azhar, M. Hoelscher, M. Maeurer, Lancet Infect.
show a large linear detection range etc. The unique properties Dis. 2016, 16, e47.
of such nanoparticle based devices provide a platform for an 2 R. A. Stein, Int. J. Clin. Pract. 2015, 69, 41.
easier and quicker detection of the pathogenic bacteria as 3 M. G. Head, J. R. Fitchett, M. K. Cooke, F. B. Wurie, A. C.
compared to the conventional microbial culture and growth Hayward, R. Atun, Lancet Infect. Dis. 2013, 13, 55.
based techniques which can take up to six days for the overall 4 C. L. Ventola, P&T 2015, 40, 277.
detection process to complete.176 There is no doubt that the 5 R. P. Dickson, J. R. Erb-Downward, G. B. Huffnagle,
combination of nanotechnology and biosensing offers an attrac- Expert Rev. Respir. Med. 2013, 7, 245.
tive platform for the development of sophisticated devices, 6 C. Fraser, S. Riley, R. M. Anderson, N. M. Ferguson, Proc.
however, there are still several associated challenges that need Natl. Acad. Sci. U.S.A. 2004, 101, 6146.
to be addressed for enhancing the overall performance of these 7 E. Leoni, P. P. Legnani, J. Appl. Microbiol. 2001, 90, 27.
8 A. K. Bej, M. H. Mahbubani, J. L. Dicesare, R. M. Atlas,
devices. By far the most important criteria that should be con-
Appl. Environ. Microbiol. 1991, 57, 3529.
sidered is to lower the cost involved in the fabrication of such
9 O. Lazcka, F. J. D. Campo, F. X. Muñoz, Biosens.
devices. For example, nanomaterials derived from semicon-
Bioelectron. 2007, 22, 1205.
ductors (CdS, CdTe and GaAs), metal/metal oxides (Au, Ag 10 G. Anantha-Iyengar, K. Shanmugasundaram, M. Nallal,
and ZnO, SiO2), magnetic materials (Fe2O3) provide highly K.-P. Lee, M. J. Whitcombe, D. Lakshmi, G. Sai-Anand, Prog.
promising results at the lab scale but these could prove too Polym. Sci. 2018, doi:10.1016/j.progpolymsci.2018.08.001.
expensive when applied on a commercial scale such as clinical 11 G. P. Mane, S. N. Talapaneni, C. Anand, S. Varghese, H.
labs. Besides this, the interference generated due to the pres- Iwai, Q. Ji, K. Ariga, T. Mori, A. Vinu, Adv. Funct. Mater. 2012,
ence of nanoparticles could also pose an issue during the 22, 3596.
analysis of the real samples which might be constituted of other 12 G. Lawrence, P. Kalimuthu, M. Benzigar, K. J. Shelat, K. S.
components along with the pathogenic bacteria. The potential Lakhi, D.-H. Park, Q. Ji, K. Ariga, P. V. Bernhardt, A. Vinu, Adv.
toxicity arising due to the presence of nanoparticles must be Mater. 2017, 29, 1702295.

240 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
13 Q. Ji, I. Honma, S.-M. Paek, M. Akada, J. P. Hill, A. Vinu, 43 R. Wang, Y. Ni, Y. Xu, Y. Jiang, C. Dong, N. Chuan, Anal.
K. Ariga, Angew. Chem., Int. Ed. 2010, 49, 9737. Chim. Acta 2015, 853, 710.
14 G. P. Mane, S. N. Talapaneni, K. S. Lakhi, H. Ilbeygi, U. 44 P. Alivisatos, Nat. Biotechnol. 2004, 22, 47.
Ravon, K. Al-Bahily, T. Mori, D.-H. Park, A. Vinu, Angew. Chem., 45 T. Jamieson, R. Bakhshi, D. Petrova, R. Pocock, M. Imani,
Int. Ed. 2017, 56, 8481. A. M. Seifalian, Biomaterials 2007, 28, 4717.
15 L. Jia, G. P. Mane, C. Anand, D. S. Dhawale, Q. Ji, K. 46 E. Oh, R. Liu, A. Nel, K. B. Gemill, M. Bilal, Y. Cohen,
Ariga, A. Vinu, Chem. Commun. 2012, 48, 9029. I. L. Medintz, Nat. Nanotechnol. 2016, 11, 479.
16 L. Jia, G. Lawrence, V. V. Balasubramanian, G. Choi, J.-H. 47 A. Kumar, V. Kumar, Chem. Rev. 2014, 114, 7044.
Choy, A. M. Abdullah, A. Elzatahry, K. Ariga, A. Vinu, Chem.® 48 X. Michalet, F. F. Pinaud, L. A. Bentolila, J. M. Tsay, S.
Eur. J. 2015, 21, 697. Doose, J. J. Li, G. Sundaresan, A. M. Wu, S. S. Gambhir, S. Weiss,
17 G. P. Mane, D. S. Dhawale, C. Anand, K. Ariga, Q. Ji, Science 2005, 307, 538.
M. A. Wahab, T. Mori, A. Vinu, J. Mater. Chem. A 2013, 1, 2913. 49 C. Kaittanis, S. Santra, J. M. Perez, Adv. Drug Delivery Rev.
18 Y. Chen, Z. P. Michael, G. P. Kotchey, Y. Zhao, A. Star, 2010, 62, 408.
ACS Appl. Mater. Interfaces 2014, 6, 3805. 50 T. M. Jovin, Nat. Biotechnol. 2003, 21, 32.
19 M. A. Chari, D. Shobha, E.-R. Kenawy, S. S. Al-Deyab, 51 M. Freitas, S. Viswanathan, H. P. A. Nouws, M. B. P. P.
B. S. Reddy, A. Vinu, Tetrahedron Lett. 2010, 51, 5195. Oliveira, C. Delerue-Matos, Biosens. Bioelectron. 2014, 51, 195.
20 M. Karthik, A. Tripathi, N. Gupta, A. Vinu, M. Hartmann, 52 P. Qi, D. Zhang, Y. Zeng, Y. Wan, Talanta 2016, 147, 142.
M. Palanichamy, V. Murugesan, Appl. Catal., A 2004, 268, 139. 53 H. N. Abdelhamid, H.-F. Wu, J. Mater. Chem. B 2013, 1,
21 S. Mandal, M. V. Lee, J. P. Hill, A. Vinu, K. Ariga, 6094.
J. Nanosci. Nanotechnol. 2010, 10, 21. 54 M. Amelia, C. Lincheneau, S. Silvi, A. Credi, Chem. Soc.
22 T. Wagner, S. Haffer, C. Weinberger, D. Klaus, M. Rev. 2012, 41, 5728.
Tiemann, Chem. Soc. Rev. 2013, 42, 4036. 55 L. Li, H. Lu, K. Deng, J. Mater. Chem. A 2013, 1, 2089.
23 P. Srinivasu, S. Alam, V. V. Balasubramanian, S. Velmathi, 56 Z. Li, X. Peng, J. Am. Chem. Soc. 2011, 133, 6578.
D. P. Sawant, W. Böhlmann, S. P. Mirajkar, K. Ariga, S. B. 57 L.-H. Xiong, R. Cui, Z.-L. Zhang, X. Yu, Z. Xie, Y.-B. Shi,
Halligudi, A. Vinu, Adv. Funct. Mater. 2008, 18, 640. D.-W. Pang, ACS Nano 2014, 8, 5116.
24 S. N. Talapaneni, G. P. Mane, A. Mano, C. Anand, D. S. 58 S. Ebrahim, M. Reda, A. Hussien, D. Zayed, Spectrochim.
Dhawale, T. Mori, A. Vinu, ChemSusChem 2012, 5, 700. Acta, Part A 2015, 150, 212.
25 B. Wu, G. Rong, J. Zhao, S. Zhang, Y. Zhu, B. He, Nano 59 M. Shanehsaz, A. Mohsenifar, S. Hasannia, N. Pirooznia,
2012, 7, 1250049. Y. Samaei, M. Shamsipur, Microchim. Acta 2013, 180, 195.
26 M. Punzet, D. Baurecht, F. Varga, H. Karlic, C. Heitzinger, 60 F. R. Madiyar, S. Bhana, L. Z. Swisher, C. T. Culbertson,
Nanoscale 2012, 4, 2431. X. Huang, J. Li, Nanoscale 2015, 7, 3726.
27 P. J. Vikesland, K. R. Wigginton, Environ. Sci. Technol. 61 B. Wang, X. Huang, M. Ma, Q. Shi, Z. Cai, Food Control
2010, 44, 3656. 2014, 35, 26.
28 N. Tawil, E. Sacher, R. Mandeville, M. Meunier, Analyst 62 B. Wang, Q. Wang, Z. Cai, M. Ma, LWT®Food Sci.
2014, 139, 1224. Technol. 2015, 61, 368.
29 A. Roda, M. Mirasoli, B. Roda, F. Bonvicini, C. Colliva, P. 63 H. Kurt, M. Yüce, B. Hussain, H. Budak, Biosens.
Reschiglian, Microchim. Acta 2012, 178, 7. Bioelectron. 2016, 81, 280.
30 J. T. Connelly, A. J. Baeumner, Anal. Bioanal. Chem. 2012, 64 S. Agrawal, A. Morarka, D. Bodas, K. M. Paknikar, Appl.
402, 117. Biochem. Biotechnol. 2012, 167, 1668.
31 A. Ahmed, J. V. Rushworth, N. A. Hirst, P. A. Millner, 65 J. Yoon, S. Jo, I. S. Chun, I. Jung, H.-S. Kim, M. Meitl, E.
Clin. Microbiol. Rev. 2014, 27, 631. Menard, X. Li, J. J. Coleman, U. Paik, J. A. Rogers, Nature 2010,
32 S. Acharya, J. P. Hill, K. Ariga, Adv. Mater. 2009, 21, 2959. 465, 329.
33 A. Rananaware, A. N. Abraham, D. D. La, V. Mistry, R. 66 E. Nazemi, S. Aithal, W. M. Hassen, E. H. Frost, J. J.
Shukla, S. V. Bhosale, Aust. J. Chem. 2017, 70, 652. Dubowski, Sens. Actuators, B 2015, 207, Part A, 556.
34 M. Al Kobaisi, S. V. Bhosale, K. Latham, A. M. Raynor, 67 J.-S. Lee, B.-H. Kang, S.-H. Kim, J.-W. Lee, S.-W. Lee,
S. V. Bhosale, Chem. Rev. 2016, 116, 11685. S.-W. Kim, S.-A. Gopalan, J.-B. Kwon, J.-H. Bae, E.-S. Kim,
35 A. Rananaware, R. S. Bhosale, H. Patil, M. Al Kobaisi, A. D.-H. Kwon, S.-W. Kang, Org. Electron. 2017, 42, 393.
Abraham, R. Shukla, S. V. Bhosale, S. V. Bhosale, RSC Adv. 2014, 68 S.-W. Lee, K.-J. Choi, B.-H. Kang, J.-S. Lee, S.-W. Kim,
4, 59078. J.-B. Kwon, S.-A. Gopalan, J.-H. Bae, E.-S. Kim, D.-H. Kwon,
36 B. Stephen Inbaraj, B. H. Chen, J. Food Drug Anal. 2016, S.-W. Kang, Org. Electron. 2016, 39, 250.
24, 15. 69 B. Xu, G. Sai-Anand, A.-I. Gopalan, Q. Qiao, S.-W. Kang,
37 H. Bridle, D. Balharry, B. Gaiser, H. Johnston, Environ. Polymers 2018, 10, 121.
Sci. Technol. 2015, 49, 10762. 70 B.-H. Kang, J.-S. Lee, S.-W. Lee, S.-W. Kim, J.-W. Lee,
38 A. Morarka, S. Agrawal, S. Kale, A. Kale, S. Ogale, K. S.-A. Gopalan, J.-S. Park, D.-H. Kwon, J.-H. Bae, H.-R. Kim,
Paknikar, D. Bodas, Biosens. Bioelectron. 2011, 26, 3050. S.-W. Kang, Sci. Rep. 2016, 6, 34659.
39 M. R. Aziziyan, W. M. Hassen, D. Morris, E. H. Frost, J. J. 71 B. Xu, S.-A. Gopalan, A.-I. Gopalan, N. Muthuchamy,
Dubowski, Biointerphases 2016, 11, 019301. K.-P. Lee, J.-S. Lee, Y. Jiang, S.-W. Lee, S.-W. Kim, J.-S. Kim,
40 P. Qi, D. Zhang, Y. Wan, Sens. Actuators, B 2013, 181, 274. H.-M. Jeong, J.-B. Kwon, J.-H. Bae, S.-W. Kang, Sci. Rep. 2017,
41 G. Gedda, H. N. Abdelhamid, M. S. Khan, H.-F. Wu, RSC 7, 45079.
Adv. 2014, 4, 45973. 72 S.-W. Lee, S.-H. Cha, K.-J. Choi, B.-H. Kang, J.-S. Lee,
42 H. Pan, Y. Zhang, G.-X. He, N. Katagori, H. Chen, BMC S.-W. Kim, J.-S. Kim, H.-M. Jeong, S.-A. Gopalan, D.-H. Kwon,
Microbiol. 2014, 14, 222. S.-W. Kang, Sensors 2016, 16, 74.

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 241
73 G. Sai-Anand, A. Dubey, A.-I. Gopalan, S. Venkatesan, S. R.-J. Jeng, J.-K. Wang, Y.-L. Wang, Nanoscale Res. Lett. 2015, 10,
Ruban, K. M. Reza, J. Choi, K. S. Lakhi, B. Xu, Q. Qiao, A. Vinu, 397.
Sol. Energy Mater. Sol. Cells 2018, 182, 246. 103 J. Wang, X. Wu, C. Wang, N. Shao, P. Dong, R. Xiao, S.
74 M. Tak, V. Gupta, M. Tomar, Biosens. Bioelectron. 2014, Wang, ACS Appl. Mater. Interfaces 2015, 7, 20919.
59, 200. 104 Q. Sun, G. Zhao, W. Dou, Anal. Methods 2015, 7, 8647.
75 J. Gopal, H.-F. Wu, C.-H. Lee, M. Manikandan, Analyst 105 A. Rehman, Y. Sarwar, Z. A. Raza, S. Z. Hussain, T.
2012, 137, 357. Mustafa, W. S. Khan, M. A. Ghauri, A. Haque, I. Hussain, Analyst
76 H. Chen, M. Zhang, B. Li, D. Chen, X. Dong, Y. Wang, Y. 2015, 140, 7366.
Gu, Biomaterials 2015, 53, 532. 106 V. C. Ozalp, G. Bayramoglu, Z. Erdem, M. Y. Arica, Anal.
77 E. Arshad, A. Anas, A. Asok, C. Jasmin, S. S. Pai, I. S. Chim. Acta 2015, 853, 533.
Bright Singh, A. Mohandas, V. Biju, RSC Adv. 2016, 6, 15686. 107 K. Niemirowicz, I. Swiecicka, A. Z. Wilczewska, K. H.
78 A. Huang, Z. Qiu, M. Jin, Z. Shen, Z. Chen, X. Wang, J.-W. Markiewicz, U. Surel, A. Kułakowska, Z. Namiot, B. Szynaka, R.
Li, Int. J. Food Microbiol. 2014, 185, 27. Bucki, H. Car, Colloids Surf., B 2015, 131, 29.
79 D. O. Demirkol, S. Timur, Int. J. Polym. Mater. Polym. 108 S. Mun, S.-J. Choi, BioChip J. 2015, 9, 10.
Biomater. 2016, 65, 85. 109 A. Mezger, J. Fock, P. Antunes, F. W. Østerberg, A. Boisen,
80 S.-M. Wu, J. Chen, X.-X. Ai, Z.-Y. Yan, J. Pharm. Biomed. M. Nilsson, M. F. Hansen, A. Ahlford, M. Donolato, ACS Nano
Anal. 2013, 78­79, 9. 2015, 9, 7374.
81 G. Kim, J.-H. Moon, C.-Y. Moh, J. Lim, Biosens. 110 Y. Lin, A. T. Hamme, II, J. Mater. Chem. B 2015, 3, 3573.
Bioelectron. 2015, 67, 243. 111 W. Lee, D. Kwon, W. Choi, G. Y. Jung, S. Jeon, Sci. Rep.
82 G. Kim, S. B. Park, J.-H. Moon, S. Lee, Anal. Methods 2015, 5, 7717.
2013, 5, 5717. 112 D. Kwon, S. Lee, M. M. Ahn, I. S. Kang, K.-H. Park, S.
83 L. Wang, C.-S. Wu, X. Fan, A. Mustapha, Int. J. Food Jeon, Anal. Chim. Acta 2015, 883, 61.
Microbiol. 2012, 156, 83. 113 A. Hardiansyah, A.-Y. Chen, H.-L. Liao, M.-C. Yang, T.-Y.
84 M. Bahadoran, A. F. A. Noorden, K. Chaudhary, F. S. Liu, T.-Y. Chan, H.-M. Tsou, C.-Y. Kuo, J.-K. Wang, Y.-L. Wang,
Mohajer, M. S. Aziz, S. Hashim, J. Ali, P. Yupapin, Sensors 2014, Nanoscale Res. Lett. 2015, 10, 412.
14, 12885. 114 S. Horikawa, Y. Chai, H. C. Wikle, J. Dai, J. Hu, S.-J. Suh,
85 D.-M. Kong, J.-H. Guo, W. Yang, Y.-E. Ma, H.-X. Shen, V. Vodyanoy, B. A. Chin, Proc. SPIE, 2015, 9488, 94880C.
Biosens. Bioelectron. 2009, 25, 88. 115 M. Fleischmann, P. J. Hendra, A. J. McQuillan, Chem.
86 G. Budin, H. J. Chung, H. Lee, R. Weissleder, Angew. Phys. Lett. 1974, 26, 163.
Chem., Int. Ed. 2012, 51, 7752. 116 E. Papadopoulou, S. E. J. Bell, Chem.®Eur. J. 2012, 18,
87 K. Y. Chan, W. W. Ye, Y. Zhang, L. D. Xiao, P. H. M. 5394.
Leung, Y. Li, M. Yang, Biosens. Bioelectron. 2013, 41, 532. 117 L. Zhang, J. Xu, L. Mi, H. Gong, S. Jiang, Q. Yu, Biosens.
88 F. Lucarelli, S. Tombelli, M. Minunni, G. Marrazza, M. Bioelectron. 2012, 31, 130.
Mascini, Anal. Chim. Acta 2008, 609, 139. 118 C. C. Lin, Proc. SPIE, 2012, 8351, 83512V.
89 D. Brandão, S. Liébana, S. Campoy, M. P. Cortés, S. 119 P. Drake, P.-S. Jiang, H.-W. Chang, S.-C. Su, J. Tanha,
Alegret, M. I. Pividori, Biosens. Bioelectron. 2015, 74, 652. L.-L. Tay, P. Chen, Y.-J. Lin, Anal. Methods 2013, 5, 4152.
90 Q. Xia, C. Peng, D. Lamb, M. Kader, M. Mallavarapu, R. 120 Z. Fan, D. Senapati, S. A. Khan, A. K. Singh, A. Hamme,
Naidu, J. C. Ng, Chemosphere 2016, 154, 343. B. Yust, D. Sardar, P. C. Ray, Chem.®Eur. J. 2013, 19, 2839.
91 T. Z. Gómez de la Torre, R. Ke, A. Mezger, P. Svedlindh, 121 E.-J. Chae, J.-H. Lee, B.-K. Oh, J.-W. Choi, J. Biomed.
M. Strømme, M. Nilsson, Small 2012, 8, 2174. Nanotechnol. 2013, 9, 659.
92 D. Cheng, M. Yu, F. Fu, W. Han, G. Li, J. Xie, Y. Song, 122 J. Xu, J. W. Turner, M. Idso, S. V. Biryukov, L. Rognstad,
M. T. Swihart, E. Song, Anal. Chem. 2016, 88, 820. H. Gong, V. L. Trainer, M. L. Wells, M. S. Strom, Q. Yu, Anal.
93 D. Kwon, J. Joo, J. Lee, K.-H. Park, S. Jeon, Anal. Chem. Chem. 2013, 85, 2630.
2013, 85, 7594. 123 K. Chen, L. Wu, X. Jiang, Z. Lu, H. Han, Biosens.
94 J. Joo, C. Yim, D. Kwon, J. Lee, H. H. Shin, H. J. Cha, S. Bioelectron. 2014, 62, 196.
Jeon, Analyst 2012, 137, 3609. 124 C.-C. Lin, Y.-M. Yang, P.-H. Liao, D.-W. Chen, H.-P. Lin,
95 Y.-T. Kim, K.-H. Kim, E. S. Kang, G. Jo, S. Y. Ahn, S. H. H. C. Chang, Biosens. Bioelectron. 2014, 53, 519.
Park, S. I. Kim, S. Mun, K. Baek, B. Kim, K. Lee, W. S. Yun, Y. H. 125 H.-Y. Lin, C.-H. Huang, W.-H. Hsieh, L.-H. Liu, Y.-C. Lin,
Kim, Bioconjugate Chem. 2016, 27, 59. C.-C. Chu, S.-T. Wang, I.-T. Kuo, L.-K. Chau, C.-Y. Yang, Small
96 S. Shukla, G. Lee, X. Song, S. Park, M. Kim, Biosens. 2014, 10, 4700.
Bioelectron. 2016, 77, 986. 126 Y. Lin, A. T. Hamme, II, Analyst 2014, 139, 3702.
97 B. Tian, R. S. Bejhed, P. Svedlindh, M. Strömberg, Biosens. 127 T. J. Ondera, A. T. Hamme, II, J. Mater. Chem. B 2014, 2,
Bioelectron. 2016, 77, 32. 7534.
98 M. Xu, R. Wang, Y. Li, Talanta 2016, 148, 200. 128 R. Najafi, S. Mukherjee, J. Hudson, A. Sharma, P. Banerjee,
99 S. Yang, H. Ouyang, X. Su, H. Gao, W. Kong, M. Wang, Q. Int. J. Food Microbiol. 2014, 189, 89.
Shu, Z. Fu, Biosens. Bioelectron. 2016, 78, 174. 129 T. Szymborski, E. Witkowska, W. Adamkiewicz, J. Waluk,
100 Y. Liu, Z. Zhang, Y. Wang, Y. Zhao, Y. Lu, X. Xu, J. Yan, A. Kamińska, Analyst 2014, 139, 5061.
Y. Pan, Int. J. Food Microbiol. 2015, 211, 109. 130 H. Zhou, D. Yang, N. P. Ivleva, N. E. Mircescu, R.
101 Z. Wang, Z. Chen, N. Gao, J. Ren, X. Qu, Small 2015, 11, Niessner, C. Haisch, Anal. Chem. 2014, 86, 1525.
4970. 131 T.-Y. Liu, Y. Chen, H.-H. Wang, Y.-L. Huang, Y.-C. Chao,
102 A. H. H. Mevold, W.-W. Hsu, A. Hardiansyah, L.-Y. K.-T. Tsai, W.-C. Cheng, C.-Y. Chuang, Y.-H. Tsai, C.-Y. Huang,
Huang, M.-C. Yang, T.-Y. Liu, T.-Y. Chan, K.-S. Wang, Y.-A. Su, D.-W. Wang, C.-H. Lin, J.-K. Wang, Y.-L. Wang, J. Nanosci.

242 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan
Nanotechnol. 2012, 12, 5004. hybrid structures, ed., ed. by Editor, City, 2015, Vol. 9487, Chap.
132 P. Negri, R. A. Dluhy, J. Biophotonics 2013, 6, 20. Chapter.
133 J. Sundaram, B. Park, Y. Kwon, K. C. Lawrence, Int. J. 161 Y. Sha, X. Zhang, W. Li, W. Wu, S. Wang, Z. Guo, J. Zhou,
Food Microbiol. 2013, 167, 67. X. Su, Talanta 2016, 147, 220.
134 N. Xiao, C. Wang, C. Yu, Biosensors 2013, 3, 312. 162 Y. Lian, F. He, H. Wang, F. Tong, Biosens. Bioelectron.
135 I.-F. Cheng, T.-Y. Chen, R.-J. Lu, H.-W. Wu, Nanoscale 2015, 65, 314.
Res. Lett. 2014, 9, 324. 163 Y. Liao, X. Zhou, D. Xing, ACS Appl. Mater. Interfaces
136 I.-H. Cho, P. Bhandari, P. Patel, J. Irudayaraj, Biosens. 2014, 6, 9988.
Bioelectron. 2015, 64, 171. 164 J. Wen, S. Zhou, Y. Yuan, Biosens. Bioelectron. 2014, 52,
137 J.-Y. Ho, T.-Y. Liu, J.-C. Wei, J. K. Wang, Y.-L. Wang, J. J. 44.
Lin, ACS Appl. Mater. Interfaces 2014, 6, 1541. 165 S. C. Sultan, Ü. Anik, Talanta 2014, 129, 523.
138 O. E. Rivera-Betancourt, E. S. Sheppard, D. C. Krause, 166 C. Liu, D. Jiang, G. Xiang, L. Liu, F. Liu, X. Pu, Analyst
R. A. Dluhy, Analyst 2014, 139, 4287. 2014, 139, 5460.
139 A. Sivanesan, E. Witkowska, W. Adamkiewicz, Ł. Dziewit, 167 M. Yang, Z. Peng, Y. Ning, Y. Chen, Q. Zhou, L. Deng,
A. Kamińska, J. Waluk, Analyst 2014, 139, 1037. Sensors (Basel) 2013, 13, 6865.
140 X. Wu, J. Chen, X. Li, Y. Zhao, S. M. Zughaier, 168 K. Yamada, C. T. Kim, J. H. Kim, J. H. Chung, H. G. Lee,
Nanomedicine 2014, 10, 1863. S. Jun, PLoS One 2014, 9, e105767.
141 L. Chen, N. Mungroo, L. Daikuara, S. Neethirajan, 169 Y. Li, P. Cheng, J. Gong, L. Fang, J. Deng, W. Liang, J.
J. Nanobiotechnology 2015, 13, 45. Zheng, Anal. Biochem. 2012, 421, 227.
142 K. C. Henderson, A. J. Benitez, A. E. Ratliff, D. M. Crabb, 170 R. Singh, R. Verma, G. Sumana, A. K. Srivastava, S. Sood,
E. S. Sheppard, J. M. Winchell, R. A. Dluhy, K. B. Waites, T. P. R. K. Gupta, B. D. Malhotra, Bioelectrochemistry 2012, 86, 30.
Atkinson, D. C. Krause, PLoS One 2015, 10, e0131831. 171 Y. Li, L. Fang, P. Cheng, J. Deng, L. Jiang, H. Huang, J.
143 S. Kumar, D. K. Lodhi, P. Goel, Neeti, P. Mishra, J. P. Zheng, Biosens. Bioelectron. 2013, 49, 485.
Singh, Chem. Commun. 2015, 51, 12411. 172 G. A. Zelada-Guillén, P. Blondeau, F. X. Rius, J. Riu,
144 S. K. Srivastava, H. B. Hamo, A. Kushmaro, R. S. Marks, Methods 2013, 63, 233.
C. Grüner, B. Rauschenbach, I. Abdulhalim, Analyst 2015, 140, 173 N. Duan, W. Gong, Z. Wang, S. Wu, Anal. Methods 2016,
3201. 8, 1390.
145 H. Wang, Y. Zhou, X. Jiang, B. Sun, Y. Zhu, H. Wang, Y. 174 C. H. Gayathri, P. Mayuri, K. Sankaran, A. S. Kumar,
Su, Y. He, Angew. Chem., Int. Ed. 2015, 54, 5132. Biosens. Bioelectron. 2016, 82, 71.
146 X. Wu, Y. W. Huang, B. Park, R. A. Tripp, Y. Zhao, Talanta 175 N. Duan, S. Wu, S. Dai, T. Miao, J. Chen, Z. Wang,
2015, 139, 96. Microchim. Acta 2015, 182, 917.
147 H. Zhou, D. Yang, N. P. Ivleva, N. E. Mircescu, S. 176 A. M. Fernandes, M. H. Abdalhai, J. Ji, B.-W. Xi, J. Xie, J.
Schubert, R. Niessner, A. Wieser, C. Haisch, Anal. Chem. 2015, Sun, R. Noeline, B. H. Lee, X. Sun, Biosens. Bioelectron. 2015,
87, 6553. 63, 399.
148 N. Duan, B. Chang, H. Zhang, Z. Wang, S. Wu, Int. J. Food 177 H. Z. Pan, H. W. Yu, N. Wang, Z. Zhang, G. C. Wan, H.
Microbiol. 2016, 218, 38. Liu, X. Guan, D. Chang, J. Biotechnol. 2015, 214, 133.
149 M. L. Bhaisare, G. Gedda, M. S. Khan, H.-F. Wu, Anal. 178 X. Hu, W. Dou, G. Zhao, J. Electroanal. Chem. 2015, 756,
Chim. Acta 2016, Ahead of Print. 43.
150 B. S. B. Kasibabu, S. L. D’souza, S. Jha, R. K. Singhal, H. 179 J. Shi, C. Chan, Y. Pang, W. Ye, F. Tian, J. Lyu, Y. Zhang,
Basu, S. K. Kailasa, Anal. Methods 2015, 7, 2373. M. Yang, Biosens. Bioelectron. 2014, 67, 595.
151 S. Nandi, M. Ritenberg, R. Jelinek, Analyst (Cambridge, 180 P. Zuo, X. Li, D. C. Dominguez, B. C. Ye, Lab Chip 2013,
U. K.) 2015, 140, 4232. 13, 3921.
152 D. Zhong, Y. Zhuo, Y. Feng, X. Yang, Biosens. Bioelectron. 181 H. N. Abdelhamid, H.-F. Wu, J. Mater. Chem. B 2013, 1,
2015, 74, 546. 3950.
153 P. Qi, Y. Wan, D. Zhang, Biosens. Bioelectron. 2013, 39, 182 R. Hernandez, C. Valles, A. M. Benito, W. K. Maser, F.
282. Xavier Rius, J. Riu, Biosens. Bioelectron. 2014, 54, 553.
154 Z. Fan, R. Kanchanapally, P. C. Ray, J. Phys. Chem. Lett. 183 I. P.-J. Lai, S. G. Harroun, S.-Y. Chen, B. Unnikrishnan,
2013, 4, 3813. Y.-J. Li, C.-C. Huang, Sens. Actuators, B 2016, 228, 465.
155 M. S. Mannoor, H. Tao, J. D. Clayton, A. Sengupta, D. L. 184 J. Ryu, E. Lee, K. Lee, J. Jang, J. Mater. Chem. B 2015, 3,
Kaplan, R. R. Naik, N. Verma, F. G. Omenetto, M. C. McAlpine, 4865.
Nat. Commun. 2012, 3, 763. 185 J. Dong, H. Zhao, M. Xu, Q. Maa, S. Ai, Food Chem. 2013,
156 J. Chang, S. Mao, Y. Zhang, S. Cui, G. Zhou, X. Wu, C. H. 141, 1980.
Yang, J. Chen, Nanoscale 2013, 5, 3620. 186 G. A. Zelada-Guillén, J. L. Sebastián-Avila, P. Blondeau, J.
157 Y. F. Duan, Y. Ning, Y. Song, L. Deng, Microchim. Acta Riu, F. X. Rius, Biosens. Bioelectron. 2012, 31, 226.
2014, 181, 647. 187 C. A. S. Andrade, J. M. Nascimento, I. S. Oliveira, C. V. J.
158 Y. Huang, X. Dong, Y. Liu, L. J. Li, P. Chen, J. Mater. de Oliveira, C. P. de Melo, O. L. Franco, M. D. L. Oliveira,
Chem. 2011, 21, 12358. Colloids Surf., B 2015, 135, 833.
159 W. Sun, X. Wang, W. Wang, Y. Lu, J. Xi, W. Zheng, F. Wu, 188 N. M. Bardhan, D. Ghosh, A. M. Belcher, Nat. Commun.
H. Ao, G. Li, J. Solid State Electrochem. 2015, 19, 2431. 2014, 5, 4918.
160 D. C. Vanegas, Y. Rong, N. Schwalb, K. D. Hills, C. 189 J. H. Kim, M. Hiraiwa, H. B. Lee, K. H. Lee, G. A.
Gomes, E. S. McLamore, in Book Rapid detection of listeria spp. Cangelosi, J. H. Chung, RSC Adv. 2013, 3, 4281.
using an internalin a aptasensor based on carbon-metal nano- 190 A. M. Fernandes, M. H. Abdalhai, J. Ji, B. W. Xi, J. Xie, J.

Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan | 243
Sun, R. Noeline, B. H. Lee, X. Sun, Biosens. Bioelectron. 2015, Acharya, L. K. Shrestha, Phys. Chem. Chem. Phys. 2018, 20,
63, 399. 18873.
191 T. J. Ondera, A. T. Hamme, II, J. Mater. Chem. B 2014, 2, 197 P. Bairi, T. Tsuruoka, S. Acharya, Q. Ji, J. P. Hill, K. Ariga,
7534. Y. Yamauchi, L. K. Shrestha, Mater. Horiz. 2018, 5, 285.
192 E. I. Maurer, K. K. Comfort, S. M. Hussain, J. J. Schlager, 198 F.-Y. Hsieh, L. K. Shrestha, K. Ariga, S.-h. Hsu, Chem.
S. M. Mukhopadhyay, Sensors (Switzerland) 2012, 12, 8135. Commun. 2017, 53, 11024.
193 M. R. Benzigar, S. N. Talapaneni, S. Joseph, K. Ramadass, 199 P. Bairi, K. Minami, W. Nakanishi, J. P. Hill, K. Ariga,
G. Singh, J. Scaranto, U. Ravon, K. Al-Bahily, A. Vinu, Chem. L. K. Shrestha, ACS Nano 2016, 10, 6631.
Soc. Rev. 2018, 47, 2680. 200 P. Bairi, K. Minami, J. P. Hill, W. Nakanishi, L. K.
194 M. R. Benzigar, S. Joseph, A. V. Baskar, D.-H. Park, G. Shrestha, C. Liu, K. Harano, E. Nakamura, K. Ariga, ACS Nano
Chandra, S. Umapathy, S. N. Talapaneni, A. Vinu, Adv. Funct. 2016, 10, 8796.
Mater. 2018, 28, 1803701. 201 P. Bairi, K. Minami, J. P. Hill, K. Ariga, L. K. Shrestha,
195 M. R. Benzigar, S. Joseph, H. Ilbeygi, D.-H. Park, S. ACS Nano 2017, 11, 7790.
Sarkar, G. Chandra, S. Umapathy, S. Srinivasan, S. N. Talapaneni, 202 Q. Tang, P. Bairi, R. G. Shrestha, J. P. Hill, K. Ariga, H.
A. Vinu, Angew. Chem. 2018, 130, 578. Zeng, Q. Ji, L. K. Shrestha, ACS Appl. Mater. Interfaces 2017, 9,
196 G. S. Kumar, R. G. Shrestha, Q. Ji, J. P. Hill, K. Ariga, S. 44458.

244 | Bull. Chem. Soc. Jpn. 2019, 92, 216–244 | doi:10.1246/bcsj.20180280 © 2019 The Chemical Society of Japan

Вам также может понравиться