You are on page 1of 37

1

1 Type of the Paper (Review)

2 Emerging Applications of Nanobiosensors in Pathogen


3 Detection in Water and Food
4 Hiram Martin Valenzuela-Amaro† 1,2, Alberto Aguayo-Acosta † 1,2, Edgar Ricardo Meléndez-Sánchez 1,2, Orlando de
5 la Rosa 1,2, Perla Guadalupe Vázquez-Ortega3, Mariel Araceli Oyervides-Muñoz1,2, Juan Eduardo Sosa-Hernández1, 2
6 * Roberto Parra-Saldivar1,2*

7 1
Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey, 64849,
8 Mexico
9 2
Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, 64849, Mexico
10 3
Tecnologico Nacional de México/Instituto Tecnológico de Durango. Blvd. Felipe Pescador 1830-Ote. Col.
11 Nueva Vizcaya. C.P. 34080, Durango, Dgo., México.
12 * Correspondence: Juan Eduardo Sosa-Hernández (eduardo.sosa@tec.mx); Roberto Parra-Saldivar
13 (r.parra@tec.mx)
14

15 Abstract: Food and waterborne illness are still a major concern in the health and food safety areas. every year there are almost
16 420,000 deaths related to foodborne illness and 2.2 million of deaths related to waterborne illness worldwide, the major concern
17 pathogenic agents for these diseases are bacteria such as: Salmonella, Shiga-toxin producer E. coli, Campylobacter, Listeria
18 monocytogenes among other, in the other side major concern waterborne pathogens are Vibrio cholerae, leptospirosis, Schistosoma
19 mansoni, S. japonicum. Despite the major efforts for the control of food and water quality, safety and prevent the presence of
20 pathogens on this kind of sources, many foodborne and waterborne illness cases still occur globally. Due to the mentioned it is
21 required the development of novel, faster and more functional real-time surveillance methods for the detection of pathogenic
22 microorganisms in these sources. Methods based in biosensor devices emerged has a novel tool for faster detection of pathogens in
23 food and water, to displace the time-consuming pathogen detection methods, that usually takes a few days to obtain a precise
24 result and in many cases these methods do not generate considerable data. The biosensor devices can be summarized as a device
25 that uses biochemical reactions mediated by isolated enzymes, antibodies, tissues, DNA, or aptamers to detect the presence of
26 pathogens usually using electrical, thermal, or optical signals in response of the presence particular pathogen biomarkers, such as
27 specific genetic material sequence. The application of Nano and Molecular technologies permits
28 Citation: To be added by editorial the identification of pathogens in a faster and high sensibility process, available to discriminate at
29 staff during production. very low concentration of the pathogen for this objective the application of nanoparticles of gold,
30 Received: date silver, iron, and magnetics nanoparticles has been used to improve the function of the biosensor
31 Revised: date devices. In this review are summarized a principal application of nanomaterials and biosensor-
32 Accepted: date based devices for the detection of pathogens in food and water samples. It has been observed that
33 Published: date the application of nanomaterials has been improving the biosensor devices performance, also in
34 combination the nanomaterials used in biosensors offer unique advantages for pathogen detection.
35 Their small size and high specific surface area allow for more effective interaction with pathogenic
Copyright: © 2023 by the authors.
Submitted for possible open access
publication under the terms and
conditions of the Creative Commons
Attribution (CC BY) license
(https://creativecommons.org/license
s/by/4.0/).

3 Biosensors 2023, 13, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/biosensors


4 Biosensors 2023, 13, x FOR PEER REVIEW 2 of 37
5

36 agents, enhancing the sensitivity and selectivity of the biosensors. Additionally, their ability to be functionalized with specific
37 molecules such as antibodies or nucleic acids facilitates the specific detection of the target pathogens.

38 Keywords: Nanobiosensors, Nanomaterials, Foodborne diseases, Waterborne diseases, Food


39 safety.
40
41
42
43

44 Graphical abstract

45

46 1. Introduction
47 Every year contaminated food is responsible for 420,000 deaths and 600 million
48 cases of foodborne illnesses caused by spoiled food [1]. This is not just a problem in
49 underdeveloped countries, also developed countries has several troubles related to
50 foodborne pathogens, Only in US there are more than 9.4 million deaths per year due to
6 Biosensors 2023, 13, x FOR PEER REVIEW 3 of 37
7

51 the ingestion of pathogenic bacteria in food (Riu & Giussani, 2020), only in 2010, 420,000
52 people (one-third of them being children under the age of five) died from illnesses
53 related to salmonellosis and Escherichia coli infection [2]. Foodborne illnesses arise from
54 the presence of pathogens, toxins, and contaminants in food products. These illnesses
55 are typically associated with gastrointestinal symptoms such as diarrhea, vomiting,
56 abdominal pain, and fever. However, certain foodborne illnesses can have a significant
57 impact on human health, leading to neurological, hepatic, and renal complications, and
58 in some cases, they can be life-threatening if not appropriately addressed [3,4]. In recent
59 years the majority of reported foodborne illness outbreaks are caused by pathogens such
60 as: Norovirus [4], Campylobacter [5], Salmonella [4,5], Listeria monocytogenes [6], and
61 Shiga toxin-producing Escherichia coli [7]. Also, other pathogens that are occasionally
62 reported to cause illness include Staphylococcus aureus [8], species of Clostridium [9],
63 Bacillus cereus [10], Yersinia enterocolitica [11].
64 Similarly, to food pathogen, the presence of pathogens in water is a major concern
65 problem for health safety and biosafety [12], in fact it is estimated that 663 million
66 people consume untreated water obtained from different sources (surface and
67 groundwater) [13]. Diseases caused by the ingestion of contaminated water cause more
68 than 2.2 million deaths per year and more cases of diseases such as diarrhea,
69 gastrointestinal and systematic diseases are related to this cause [14], the major
70 pathogens related are: Salmonella, Shigella, Campylobacter, Staphylococcus aureus, and E.
71 coli [15,16], however the virus and parasite related diseases has become an incoming
72 problem [17], some parasite and pathogens linked to waterborne outbreaks include
73 Vibrio cholerae, leptospirosis, Schistosoma mansoni, S. japonicum, [15,18,19].
74 Monitoring the presence of pathogens such as viruses, bacteria and parasites in
75 water is very important as a measure to prevent diseases, derived from in drinking
76 contaminated water, also to surveillance the quality of water, these can be achieved
77 using protocol such as a wastewater-based epidemiology protocols, which allow the
78 detection of these pathogens [20,21], and can be applied to verify the quality of the
79 water that is discarded and how it has to be treated so as not to cause adverse effects on
80 the environment and to ensure water sustainability for future generations.
81 Pathogen detection methods has a crucial role in this quality preservation activity
82 and ensure food and water safety, however actual monitoring methods are time-
83 consuming process that usually takes a few days to obtain a precise result, and in some
84 cases they do not generate considerable data, limiting a real time pathogen monitoring
85 [22]. In fact, for the identification of pathogenic agents such as bacteria and viruses,
86 however, gold standards methodologies are traditional techniques: viable plate counts,
87 flow cytometry and staining methods, among others [23] [24,25]. Nevertheless, the
88 detection time is one of the major limitations of this technique, in fact, the necessary to
89 growth the pathogen in laboratory conditions is a limitans of this protocols, that can take
8 Biosensors 2023, 13, x FOR PEER REVIEW 4 of 37
9

90 several days to get a result, hindering the response time for the control of pathogens
91 [24]. Among other fast techniques used for pathogen detection involve, polymerase
92 chain reaction techniques (PCR) [20,26,27], immunological methods [28] next-
93 generation sequencing [29], whole-genome sequencing [30], flow cytometry [31], and
94 the inclusions of the use of biosensors and nanoparticles (NPs ) in recent years [32–34].
95 The last-mentioned methods help to perform faster monitoring (real-time
96 surveillance systems), reducing response times to the presence of pathogens in water
97 bodies [35], the use of biosensors improved with NPs, allowing a better performance of
98 the device by making it faster, more specific and portable, among other improvements in
99 its characteristics [36]. In fact, the nature of the NPs is a subject of study as they show
100 diverse detection capabilities, when applied under the assumption of being part of a
101 pathogen detection system [37].
102 Due the above mentioned and the necessity of increasing of concern of other kind of
103 pathogens, it is becoming necessary the development of rapid methods for the detection
104 of pathogenic bacteria, virus and parasite [38]. Unfortunately, and despite recent
105 advances in new pathogen detection approaches, their applicability is still limited, this is
106 why technologies capable of obtaining better results in a fast and affordable way have
107 been studied and developed new technologies that are "rapid, sensitive and specific" for
108 the detection of pathogens is a priority assignment to ensure health security and prevent
109 food and water borne outbreaks, for this purpose the development of biosensor-devices
110 emerge as a novel solution [39].
111 In order to improve the pathogen detection method, this is why multiple
112 methodologies and technologies have been developed to allow for rapid and accurate
113 pathogen detection [40]. The tools offered by molecular biology have emerged as an
114 alternative for faster analysis [41], e.g., quantitative polymerase chain reaction (qPCR)
115 [42], conventional polymerase chain reaction (PCR) [43] digital droplet PCR (ddPCR)
116 (Chen et al., 2021), fluorescence in situ hybridization (FISH) [43], surface plasmon
117 resonance imaging (SPR) [45], enzyme-linked immunosorbent assay (ELISA) [46],
118 surface-enhanced Raman spectroscopy (SERS) [47], these technique has been applied
119 has a pathogen detection methodologies in different matrices such as food and water
120 [4,24].
121 The Biosensors devices are composed as follows; biorecognition element, a
122 transducer, amplifier, and processor. The biorecognition element recognizes the analyte
123 of interest, the transducer generates a signal from the recognition of the analyte into a
124 measurable signal which is then processed by a processor and amplifier to obtain a
125 signal output [48,49]. In summary is a bioanalytical device that detect specific
126 biomarkers using biochemical reactions [50] mediated by isolated enzymes, antibodies,
127 tissues, organelles, or whole cells to detect biomarkers related to the presence of
10 Biosensors 2023, 13, x FOR PEER REVIEW 5 of 37
11

128 pathogen, using electrical, thermal, or optical signals [51], correlating the presence of
129 specific pathogen in a mentioned matrixes and signal emission [32].
130 The application of biosensor have garnered attention in the field of pathogen
131 detection due to their precision, decent selectivity, and short analysis times [25].
132 However, these methodologies also have certain disadvantages (use of expensive
133 enzymes and equipment), not to mention the extensive laboratory work involved.
134 Therefore, there is a need to develop fast, highly specific, and sensitive techniques for
135 pathogen detection [35]. Currently, biosensor technology has greater possibilities for
136 application due to its unique sensitivity, low detection limit, and simple operation
137 compared to traditional techniques [52].
138 In recent years, the development of biosensors has been focused on the
139 miniaturization of the devices without affecting the detection efficacy. To achieve this
140 priority NPs are employed for the miniaturization of the biosensor through the
141 development of the nano-scale platform, indeed, in the different sections of the
142 biosensor, NPs are used as signal transducers to convert a biomolecular interaction into
143 an electrical, optical, or magnetic signal [53]. This functionality inside of biosensor is
144 because of unique properties at nanometric scales (surface area, small size, affinity for
145 some biomolecules, catalytic activity, and autofluorescence) [54,55].
146 The nanobiosensors are composed of three parts: biorecognition probe, transducer,
147 and amplifier (fig. 1) [56]. NPs are often used as transducers which combine a
148 biochemical, electrical, magnetic or optical signal [53] these signals can be read simply
149 and effectively (fig.1) shows the structure of a biosensor using NPs functionalized with
150 biorecognition and as transducer. [57]
151

Figure 1. General structure of nanobiosensor with differents agents of biorecognition


12 Biosensors 2023, 13, x FOR PEER REVIEW 6 of 37
13

152 Nanomaterials have been identified as ideal candidates to enhance biosensors in


153 terms of sensitivity and the reduction of detection limits, enhances detection capabilities,
154 being highly specific and making it one of the best alternatives for early diagnosis [37];
155 [36]. Through the detection of electrochemical signals, the specificity of signal
156 recognition is results of the adequate selection of ligands, and functionality, directly
157 conjugated with the NPs that help to attracts biomarkers of interest, also NPs convert
158 signals from one form to another or act as detectors of the generated signals [58],[59].
159 The classification based on the type of bio-recognition immobilized on the
160 nanomaterial [60] is divided into: enzymes [61], antibodies [62], antigens [63], DNA-
161 RNA [64], organelle [65], cell membrane [66], and phage particle [67]. The conversion of
162 this signal can be achieved in different ways, and this can be classified according to the
163 type of conversion used [68]. The classification of conversion systems can include optical
164 systems. [69] electrochemical nanobiosensors [70] thermoelectric [71] and piezoelectric
165 [72]. The classification based on the type of bio-recognition immobilized on the
166 nanomaterial [60] is divided into: enzymes [61], antibodies [62], antigens [63], DNA-
167 RNA [64], organelle [65], cell membrane [66], and phage particle [67]. The conversion of
168 this signal can be achieved in different ways, and this can be classified according to the
169 type of conversion used [68]. The classification of conversion systems can include optical
170 systems. [69] electrochemical nanobiosensors [70] thermoelectric [71] and piezoelectric
171 [72].

172 2. Biofunctionalization of Nanostructured Surfaces for Interaction with


173 Biorecognition Agents
174 It is essential for the biomaterial surface to be biocompatible in order to allow
175 protein adsorption without altering the natural structure of the bioactive molecule [73].
176 Bioconjugation involves the interaction of chemical or functional groups between NPs
177 and biomolecules [74,75]. Also, it is important to mention the properties that can affect
178 the efficiency of the connection with the biomarkers are: the optimal distance between
179 biorecognition molecules and the nanostructure, the pH of the storage buffer used, as
180 well as potential modifications to the biological and antigenic properties of biomolecules
181 after conjugation, developing different approaches for nanostructures conjugation
182 [76,77], by custom functional groups (such as primary amines, carboxylates, cis-diols,
183 and sulfhydryls) on nanosurfaces.[54,78].
184 Therefore, the adsorption of bioactive molecules into biosensor and nanobiosensor
185 devices can be classified into the following categories: (i) non-covalent immobilization
186 strategies, those techniques are based in electrostatic interaction, hydrophobic
187 adsorption, and coordination bond formation between biorecognition molecules and the
188 surface of nanomaterials (fig. 2) [75,79], (ii) covalent immobilization strategies,
189 involving chemical bonds, chemically activate and modify biorecognition molecules,
14 Biosensors 2023, 13, x FOR PEER REVIEW 7 of 37
15

190 achieving a stable binding. They enhance sensitivity and selectivity in pathogen
191 detection, ensuring a robust interaction [80], and finally (iii) combined techniques
192 [76,81].
193

194

195 Figure 2. Different techniques of bioconjugation in nanomaterials

196 2.1 Biorecognition section of bionsensor device: Enzymes applications


197 Enzymatic reactions in biodetection processes involve the following steps: Firstly,
198 enzymes recognize and bind to the target molecule in the environment / solution, often
199 through specific binding sites or active sites on the enzyme, enzymes are typically
200 immobilized on the surface, electrode or substrate, of the sensor in order to provide the
201 best conditions to react with a specific target molecule and produce a detectable signal
202 [82]. Subsequently, enzymes catalyze chemical reactions and produce detectable signals.
203 Enzymatic activity can be used as a signal through variations in the concentration of
204 protons, entrance or exit of gases, heat emission (Dziąbowska et al., 2018). Finally, the
205 signal generated is detected and quantified using detection techniques (electrochemical
206 and fluorescence techniques) [82,83]. In fact, a biosensor-based in enzymes immobilized
207 on Au-NP was used to detect the microbial pathogen , Campylobacter jejuni in chicken
208 breast samples, in this biosensor the nuclease enzymes and deoxyribozymes was
209 immobilized to detect the presence of the pathogen for the reaction of the scission
210 enzymatic generating a heteroduplex of DNA-RNA, which finally induce a detectable
211 signal based in a fluorescence detection model with a LOD of 10 pM [84]
16 Biosensors 2023, 13, x FOR PEER REVIEW 8 of 37
17

212 To enhance the biosensor recognition capacities, the immobilization and


213 stabilization of the enzyme are a normal process, in fact, for the immobilization of
214 enzymes on nanostructures, different techniques are commonly applied, such as
215 covalent binding, crosslinking, and self-assembled monolayers [85]. However, using
216 enzymes as biorecognition agents has certain disadvantages in in situ applications.
217 Enzymatic activity can be influenced by environmental conditions such as temperature
218 and pH, which affect the stability of the biosensor [86].
219
220 2.2 Antibody applications
221 Antibodies (immunoglobulins) are proteins produced by cells of the immune
222 system called B lymphocytes [87]. They consist of a basic structure composed of four
223 polypeptide chains: two identical heavy chains and two identical light chains with their
224 typical "Y" shape [87,88]. Antibodies are among the many biological molecules that
225 have gained importance due to their high specificity and in vivo uniqueness, for the
226 detection of pathogen biomarker [89]. The monoclonal antibodies, have been designed
227 to precisely target antigens or receptors [90]. The antibodies used as a part of biosensor
228 device are derived from animals and commonly are immobilized . This is the case with
229 Majdi et al., (2019) for the immovilized an antibody with NP or nanomaterial includes
230 adsorption by weak interaction such as electrostatic, hydrophobic or van de Waals forces
231 [92], in the other hand the preferred method for immobilizing antibodies onto NPs and
232 other surfaces is through covalent bonding, specifically using carbodiimide chemistry
233 and maleimide conjugation. This approach allows for longer lasting and reusable
234 devices, as well as better control over antibody orientation, resulting in enhanced
235 detection capabilities [92,93].
236 These type of antibody-functionalized biosensor has been used for the biodetection
237 of different pathogens, in the case of Guo et al., (2020) developed a method that utilizes
238 NPs etching. These techniques allow for specific detection of Salmonella Typhimurium.
239 Using a catalase-modified antibodies bind to the bacteria and catalyze the conversion of
240 H2O2 to H2O. In the absence of Salmonella Typhimurium, the catalase-modified
241 antibodies do not bind to the bacteria, resulting in a significant accumulation of residual
242 H2O2. HRP triggers the production of •OH, causing a color change in the Au nanowires
243 from dark blue to pink. The linear detection range is between 18 UFC mL−1 to 1.8 × 10^5
244 CFU mL−1, with a detection limit of 35 CFU mL−1 [94]. However, antibody-
245 functionalized biosensors have limitations including lack of specificity, long-term
246 stability, high production cost, challenges in antibody immobilization, potential cross-
247 reactivity, limited antibody availability, and batch-to-batch variability [32].
248
249 2.3 DNA applications
18 Biosensors 2023, 13, x FOR PEER REVIEW 9 of 37
19

250 Nucleic acid-based biosensors, such as DNA, stand out as biorecognition elements
251 due to their simplicity, speed, and high specificity. For this reason, they are widely used
252 for the detection of pathogens and other substances of interest in various biodetection
253 applications [77]. These characteristics make DNA a powerful and versatile tool in the
254 field of biodetection [95,96]. These molecular probes can be used in different ways and
255 methods, such as DNAzyme [97], DNA hairpin [98], DNA hybridization [99], and DNA
256 origami [100]. It is widely recognized that DNA and its assembly structure can be
257 applied to detect specific targets, including nucleic acids, proteins, metal ions, and small
258 biological molecules. Common bioreceptors in this category include deoxyribonucleic
259 acid (DNA), ribonucleic acid (RNA), and peptide nucleic acids (PNA) [101].
260 These biomarkers have been functionalized with nanomaterials to enhance their
261 selectivity and durability in pathogen biodetection. An application of pathogenic
262 bacteria detection in milk, was development using a combination of photo-induced
263 electron transfer (PET) between a G-quadruplex DNAzyme and silver nanocluster-
264 labeled DNA, along with exponential circular amplification based on hairpin probe,
265 achieving an ultra-low detection limit of 8 cfu·mL−1 for S. Typhimurium. This strategy
266 represents a promising platform for highly sensitive and specific detection of pathogenic
267 bacteria in food analysis [102]. In another study a fluorescent DNA hairpin template
268 was develop by designing two hairpin probes with Au-NPs for the detection of
269 Staphylococcus aureus 16S rRNA. HP1 was biofunctionalized with thiol groups and a
270 fluorescent chromophore, and a thiol group was attached to the NP surface. The
271 addition of HP2 causes the target sequence to walk along the surface of the Au-NPs,
272 thus opening the hairpin structure of HP2 and enabling the recycling of the target
273 sequence. They achieved a LOD of 7.73 CFU mL-1 with an FM of 4.36x10-5,
274 demonstrating a novel and efficient method for the detection of S. aureus [103].
275
276 2.4 Aptamer applications
277 Aptamers are short sequences of RNA or DNA (oligonucleotides) capable of folding
278 into unique three-dimensional structures and binding to targets such as proteins, lipids,
279 ions, small-molecular-weight metabolites, and even whole cells with high specificity and
280 affinity [104]. To produce aptamers, the SELEX (Systematic Evolution of Ligands by
281 Exponential Enrichment) process is utilized. In this process, aptamers are generated
282 through in vitro synthesis of combinatorial libraries with diverse sequences [35].
283 Through an iterative selection process, aptamers with higher affinity for the desired
284 target are enriched and amplified, while those with lower affinity are discarded. This
285 enables the generation of highly specific and high-affinity aptamers for various
286 biomolecular targets [105]. The analytes of aptamer-based biosensors can vary between
287 size, and complexity as it can detect specific molecules such as proteins or more complex
288 analytes such as whole cells. Aptamers modify its structure once reacting with a specific
289 analyte and the conformational change can be transduced by different types of signals
20 Biosensors 2023, 13, x FOR PEER REVIEW 10 of 37
21

290 such as optical or electrochemical (Sharifi et al. 2020; Bakhshandeh et al. 2022;
291 Sadanandan et al. 2023).
292 Some of the applications of aptamer-based sensor were developed for the detection
293 of S. aureus, E. coli and Campylobacter jejuni pathogen biomarkers [35] another example
294 was the results for S. aures detection protocolos where they designed an ultra-sensitive
295 magnetic fluorescence aptasensor based on fluorescence resonance energy transfer, the
296 aptamers on the surface of Fe3O4 and modified carbon dots (CDs). CDs were used as the
297 fluorescence donor and Fe3O4 as the "off-on" sensor receptor. Due to the strong affinity of
298 the aptamers for bacteria, the presence of target bacteria led to the disassembly of the
299 Fe3O4/CDs aptasensor, resulting in the recovery of CD fluorescence whit a range of
300 detection exhibited between 50x10^7 CFU mL-1 and 8 CFU mL-1 [106].
301 Other example is the application for Escherichia coli detection using graphene oxide
302 (GO)-modified Au-NPs, enhanced with aptamers E8 aptamer for E. coli detection. The
303 detection limit was found to be 10 cells mL-1 in water and coconut water-enriched
304 samples. Furthermore, the aptamer-based nanosensor exhibited selectivity towards its
305 target without any cross-reactivity with other bacteria. The color change from red to
306 blue, based on aggregation, can be easily visualized by naked eye [107], other protocol
307 for E. coli detection in water, was the nanobiosensor using QDs functionalized with
308 aptamer II and coated with magnetic NPs. Fluorescence values were recorded for 100,
309 200, 300, 400, and 500 CFU, each with CdTe-MPA QDs at 100 μg/mL, resulting in digital
310 signals of 29.3 mV, 34.18 mV, 39.06 mV, 43.94 mV, and 48.82 mV respectively,
311 demonstrate that CdTe-MPA QDs conjugated with aptamer II were capable of
312 selectively capture and detect E. coli [108].
313 Aptamers exhibit significant advantages for the application in pathogen detection,
314 including lower molecular weight, easier and more cost-effective production methods,
315 and good chemical stability [35], also their ability to be generated against a wide range
316 of targets, ranging from small molecules to large proteins, even whole live cells such as
317 bacteria [109], leading to their utilization in various nanobiosensor technologies for
318 pathogen detection. Different technologies are employed in these sensors, including
319 surface plasmon resonance (SPR), electrochemistry, piezoelectric effect, and
320 chemiluminescence [68].
321 For example, SPR sensors utilize the reflection of light on a modified metal surface
322 to detect changes in refractive index due to analyte binding. This enables precise and
323 sensitive detection of the target analytes [68]. In the case of electrochemical sensors, the
324 analyte interaction is translated into an electrical signal, providing a quantitative means
325 of detection and enabling real-time measurements [110]. Piezoelectric sensors, on the
326 other hand, leverage the piezoelectric effect to convert the mechanical energy generated
327 by analyte interaction into electrical energy, allowing highly sensitive and accurate
328 detection [111]. Furthermore, chemiluminescence is another technology used in
329 biosensors, where the analyte interaction triggers a chemical reaction that generates
22 Biosensors 2023, 13, x FOR PEER REVIEW 11 of 37
23

330 light. This emitted chemiluminescent light can be measured to detect and quantify the
331 analyte, providing highly sensitive and specific detection [112].
332
333 Functionalizing nanomaterials with aptamers have allowed combining various
334 signal transduction strategies (including colorimetric, electrochemical, and fluorescence
335 methods) for detecting foodborne pathogens. This versatility in detection provides the
336 ability to utilize different approaches according to the specific needs of each application,
337 enhancing the sensitivity and selectivity of detection systems. Thus, aptamers become a
338 powerful and promising tool in the fight against food contamination and the protection
339 of public health [113]. These technologies enable the detection and quantification of
340 substances in biological samples, providing versatile and efficient options for
341 applications in fields such as medical diagnosis, food safety, and environmental

Figure 3. Combination of different nanomaterials and sensors for the detection of pathogens
342 monitoring (fig 3).

343 3. Nanomaterials for the detection of pathogens in water and food


344
345 The precise detection of pathogens in water and food is of vital importance to
346 ensure sanitary safety. In this context, nanomaterials play a crucial role by offering
347 innovative and sensitive solutions. The exploration of nanomaterials in combination
348 with highly efficient aptamers is revolutionizing the detection of pathogens in water and
24 Biosensors 2023, 13, x FOR PEER REVIEW 12 of 37
25

349 food. This fusion of nanotechnology and aptamers opens new possibilities for more
350 effective control and quicker responses to potential public health risks. The following
351 table summarizes the last five years of nanobiosensor production for the detection of
352 viruses, bacteria, and parasites using aptamers in complex matrices.
353

354 Table 1 NPs application for detection of pathogenic bacteria in food and water matrices

Nanomateria Pathogen Matrix LOD Signal Bioconjugate Reference


l d material
Au-NPs S. aureus Tap- 101 to 104 Fluorescence Aptamer [114]
water CFU
mL-1.
Au-NPs E. coli Tap 10 cell Fluorescence - [115]
S. water mL-1 in
Typhimurium and lake tap
water water
and 100
in lake
Au- P. aeruginosa Water 1 cell LSPR Aptamer [116]
N.triangles
AuNPs Staphylococcus Luria 1.5x107 Colorimetric Aptamer [117]
aureus bertani cells/mL
media
AuNPs Ochratoxin A Peanut, 28.18 Colorimetric Aptamer [118]
soybean, pg/mL
and corn
AuNPs E. coli Flour 2.5 ng/µ Colorimetric Probe [119]
L
Graphene E. coli Bacterial 1x103 Colorimetric Antibody [120]
oxide coated S. suspensio CFU
AuNPs Typhimurium n

Ag-NPs S. aureus Water 1,0 Electrochemical Aptamer [121]


ufc/mL
Ag-NPs E. coli Water 150 Electrochemical Aptamer [122]
CFU/ml
Chitosan- Glipopolysacc Bacterial 248 Electrochemical - [123]
AgNPs haride suspensi CFU/mL
26 Biosensors 2023, 13, x FOR PEER REVIEW 13 of 37
27

on
AgNPs S. aureus Bacterial 1 Electrochemical Aptamer [124]
suspensi CFU/mL
on and
human
serum
AgNPs E. coli Pork, 2 Photoelectroch Peptide [125]
cabbage CFU/mL emical Magainin
and milk
Au-NPs and E. coli Water 9,34 Electrochemical Aptamer [126]
oxide of CFU mL
graphene -1
NPs
Multiwalled E. coli Water 0,8 Electrochemical Antibody [127]
carbon CFU/mL
nanotubes
Graphene Salmonella Water 102–108 Colorimetric Antibody [128]
and carbon enteritidis cfu mL-
nanotubes 1
FeO-NPS and E. coli Water 1 × 102 Fluorescence Aptamer [108]
Quantum dots CFU
Quantum S. aureus, S. Water 16-28 Colorimetric Aptamers [129]
dots typhimurium CFU/mL
SiNPs E. coli Bacterial 103 Electrochemical Polyclonalanti [130].
suspensio CFu/mL bodies
n
SiNPs E. coli Bacterial 8 Fluorescence Rhodamine B [131]
suspensio CFU/mL
n

SiNPs AFB1 from Peanut, 0.214 Fluorescence Aptamer [132]


filamentous maize pg/mL
fungi and
badam
MNPs S. aureus Milk, 2.5 ng/µ Colorimetric Probes [133]
Romaine L
lettuce,
ham,
28 Biosensors 2023, 13, x FOR PEER REVIEW 14 of 37
29

sausage
Iron oxide Bacillus cereus Inoculate 12 Electrochemical Vancomycin [134]
MNPs assisted and Shigella d media cells/mL
AuNPs flexneri and
3
cells/mL
Iron core gold Salmonella Beverage 32 Fluorescence Antibody [55]
NPs enteritidis samples Salmonell
a/mL
MNPs Salmonella Abrir en 53 Abrir en tec Abrir en tec [135]
Typhimurium tec CFU/nL
Iron oxide Hepatitis E Clinical 56 RNA Fluorescence Antinody [136]
encapsulated virus samples copies/m Electrochemical
quantum dots Norovirus L
69 RNA
copies/m
L
QDs Salmonella Chicken 43 Fluorescence Antibody [137]
Typhimurium meats CFU/mL
QDs Salmonella Aquatic 10 Fluorescence Aptamer [138]
Typhimurium samples CFU/mL
and Vibrio
parahaemolyti
cus
102
CFU/mL
QDs nanobeds Salmonella Potable 10−1 Fluorescence Antibody [139]
Typhimurium water, CFU/mL
orange
juice,
lettuce,
and
chicken
355
356 3.1 Gold nanopartícles (Au-NPs)
357 Among the different types of NPs, metallic nanoparticles (MNPs) exhibit many
358 useful characteristics for their application in various fields of biotechnology, such as
359 high surface-to-volume ratio, conductivity, selectivity, and excellent optical and
360 chemical properties [140,141]. The application can vary depending on the metal used,
30 Biosensors 2023, 13, x FOR PEER REVIEW 15 of 37
31

361 size, shape, surface properties, and functionalization of the MNPs (Patel et al., 2022). On
362 one hand, Au-NPs have been successfully used in pathogen detection because they can
363 easily be conjugated with recognition and biorecognition elements such as aptamers,
364 DNA, antibodies, carbohydrates, and proteins, which can enhance the reactivity and
365 selectivity of the NPs towards specific pathogens [33,143].
366 Au-NPs are one of the most stable MNPs, not to mention their unique
367 characteristics such as good chemical reactivity, conductivity, and high resistance, which
368 have attracted attention for their use in biosensor development [144]. The surface of Au-
369 NPs has been functionalized with various biocomponents [145]. These nanobiosensors
370 have a very low limit of detection (LOD) for different chemical and biological analytes,
371 not to mention their high stability against oxidation [144].
372 Their characteristics, such as stability, conjugation, amplification properties, and
373 their ability to serve as colorimetric biosensors [146–148], as are especially relevant in the
374 case of Au-NPs due to their localized surface plasmon resonance, which is a
375 phenomenon that gives unique optical properties to MNPs, particularly Au-NPs. This is
376 due to the interaction of electromagnetic waves with NPs of specific sizes and shapes,
377 resulting in differential absorption of the light spectrum and different colors exhibited
378 by the NPs [32,33]. These properties can be altered in the presence of different analytes,
379 making Au-NPs highly suitable for biosensor development.
380 3.2 Silver nanoparticles (Ag-NPs)
381 Ag-NPs stand out for their wide range of applications. These nanomaterials have
382 been incorporated into textiles, healthcare products, consumer goods, medical devices,
383 biodetection, among others [149]. These materials are highly attractive in the field of
384 diagnostics due to their high conductivity, catalytic activity, and plasmonic properties
385 that can be leveraged to enhance the performance of biosensors [150]. Sensitivity is a
386 crucial factor in biosensors for detecting low concentrations of an analyte. Ag-NPs have
387 been used to increase the electroactive surface area of electrodes, thereby enhancing the
388 electron transfer rate and improving biosensor sensitivity biosensor [149,150]. In a
389 biosensor, Ag-NPs have the characteristic of amplifying signals or improving the
390 detection of nucleic acids. Their plasmonic resonance absorption band below 500 nm
391 confers selective absorption in the visible and near-infrared spectrum [144]. In the
392 phenomenon of surface plasmon resonance (SPR), electrons on the surface of a metal are
393 excited by photons of specific wavelengths and incidence angles (Li, 2018; Varghese
394 Alex et al., 2020), This phenomenon is utilized to detect a target based on the refractive
395 index [151]. When bound to a biosensor, the biological recognition event between the
396 analyte and the biological recognition element results in a change in the SPR resonance
397 angle [153].
398 Conjugated polymers such as those that include silver nano particles are promising
399 materials for the addressing of current and emerging issues such as pandemic
400 monitoring [154], and pathogen detection both in food [125] and in water [121]
32 Biosensors 2023, 13, x FOR PEER REVIEW 16 of 37
33

401
402 3.3 Carbon-based nanoparticles
403 Similar to Au-NPs, carbon-based NPs are useful for the implementation of
404 detection techniques for pathogen monitoring in water [155,156]. Carbon-based NPs
405 such as carbon nanotubes, graphene and carbon nanodots have great potential in
406 biosensing of pathogens because of their ability to be coated with different biomolecules
407 for association of molecular patterns from pathogens and generate a signal for specific
408 pathogens as functionalized NPs can mimic the specific surface structure of pathogens
409 [157]. Carbon NPs have been used in the fluorescence resonance energy transfer (FRET)
410 technique with quantum dots as donors modified with aptamers for the detection of
411 Vibrio parahaemolyticus and Salmonella typhimurium in the range of from 25 and 35 cfu/mL
412 and up to 50 to106 cfu/mL respectively [158]. Also, these NPs can be used in combination
413 with aptamers to amplify the sensitivity and specificity of the device.
414
415 3.4 Magnetic nanomaterials (MNPs)
416 Magnetic NPs possess their own versatility when used for biosensing pathogens,
417 because of their specific attributes particularly fast separation and concentration that
418 makes them an easy tool for pathogen detection [159]. MNPs have been used for
419 detecting pathogen by nucleic acid detection and quantification in devices for point of
420 care testing in the detection of hepatitis B virus (limit of detection of 50 IU/mL) and
421 SARS-CoV-2 (500 copies/mL) [160].
422 The magnetic NPs (MNPs) can conform a section of the transducer part of the
423 biosensor, or to be suspended in solution in direct contact with the analyte of interest
424 [161], when the MNPs are in contact with the sample, bind to the target molecule by
425 interaction of the label in the NPs (a functional group) and a protein, once the complex
426 of MNP and target is formed an external magnetic field attract it to the active detection
427 surface and after a wash of the not binding molecules targets are detected [162]
428 When talking about magnetic NPs s in biosensing, it is important to mention the
429 magnetic relaxation switching mechanism (MRS). This phenomenon describes the time
430 when cross-linking occurs between the MNPs in the binding and recognition of targets.
431 When these MNPs clusters are formed, a change in the transverse relaxation of the
432 sample is reflected as motional averaging or static dephasing according to the MNPs
433 cluster size and this change can be monitored by nuclear magnetic resonance (Min et al.,
434 2012).
435
436 3.5 Silica nanoparticles (Si-NPs)
437 Si-NPs have applications in biomedical field [164], they present good optical
438 properties and also have a good biocompatibility [165] NPs possesses the quality of
439 being mesoporous witch in combination with other metals result in attractive and
34 Biosensors 2023, 13, x FOR PEER REVIEW 17 of 37
35

440 profitable characteristics for biosensing purposes [165] Its uniformity and easily pore
441 size change among the gating mechanism makes the very useful in biosensing for drugs
442 delivery, for example [166] another important characteristic of SiNPs is that there are
443 considered as a GRAS (generally recognized as safe) material by the FDA. [167,168].
444 The mesoporous nature of the silica NPs is a characteristic of big interest, this
445 feature can be employed to separate bacteria from complex samples even preserving its
446 viability, colloidal stabilization of magnetic NPs for the same purpose. Also, the silanol
447 functional groups from SiNPs make possible the use and design of various bio-
448 recognition systems that help to increase the sensibility and selectivity while reducing
449 detection time of different pathogens [168].
450
451 3.6 Quantum Dots (QD)
452 Quantum dots (QD) are colloidal nanocrystalline semiconductors that possess
453 properties such as quantum confinement effect, allowing them to emit and absorb light
454 at specific wavelengths [169]. Because of this, QDs exhibit excellent optical properties,
455 including a broad absorption spectrum, a narrow emission spectrum, and tunable
456 luminescence, which show great prospects in biodetection [170]
457 QD-based biosensors include and may not be limited to fluorescence,
458 bioluminescent, chemiluminescent and photoelectrochemical approaches [171]. Some of
459 the characteristics that make attractive the use of quantum dots for biosensing
460 applications are that the possess high quantum yield, better photobleaching resistance,
461 wide absorption spectrum, a narrow emission spectrum and their specificity with
462 biologic targets in comparison with common fluorophores and dyes (Pourmadadi et al.,
463 2023). Also, it is very remarkable that its surface is easily functionalized with biologic
464 components in order to integrate QD probes [171]. In the field of nanomaterials, also the
465 use of combinations of magnetic compounds displays attractive characteristics for
466 current applications, these nanocomposites besides maintain magnetic behavior
467 complementary add functional proprieties to the final product, [136]
468 As presented above numerous studies focus its determinations on Salmonella
469 Typhimurium mainly because it is the most common pathogen related to food poisoning
470 in western countries causing gastro ententeritis [173]. If well it is the model or the target
471 of the experimentations, the modifications in for example primers design or binding
472 proteins may allow to replicate the studies carried with this strain to any other food
473 pathogen [138,139,174].

474 4. Challenges and limitations to detect pathogens by DNA using nanobiosensors


475 The importance of exceptionally responsive devices is essential in advancing
476 biosensors. Insufficient sensitivity and affinity towards biomarkers can significantly
477 impact the performance of the device, in fact some of the biomarkers are at ultra-low
36 Biosensors 2023, 13, x FOR PEER REVIEW 18 of 37
37

478 concentration in the samples (pM) and in consequence it is necessary that the device
479 could detect these concentrations. [175]. For the application of biosensor-base detection
480 protocols a range of detectable concentration are dependent of biomarker selected, also
481 of the analyte source, in fact some limit of detection has a range of pM to mM of
482 concentrations, for pathogen detection [176] However, using a genetic and whole cell-
483 based biomarker targets, some of the bionsensor has limit of detection of 3 genetic copies
484 per sample [177], 1CFUmL-1 [178] or even reaching a detection limit of 3x10 6 gene copies
485 per sample [8] or 5x104 CFUmL-1 biomarker concentration in the sample [179]
486
487 As is mentioned above, the choice of signal recognition technology can determine
488 the sensitivity level required to identify biochemical, genetic or whole cell biomarker
489 concentration in the sample [180,181] affect the performance of the device, in fact the
490 detection ultra-low concentration of pathogen-diseases biomarkers concentration in
491 samples is a mandatory request to early detection in clinical diagnoses [182]
492
493 Some of the disadvantages of biosensor-based pathogens detection are firstly: the
494 factors to be determined is the target molecule to be sensed, in fact the sensitivity and
495 specificity are compromised by the biomarker choice [96]. The use of genetic markers
496 makes the device more sensitive, however it necessitates intricate systems and laborious
497 sample/re-agent handling procedures [183].
498
499 On the other hand, signal emission technology is another important factor in
500 pathogen detection, colorimetric-based biosensor has several factors that may alter its
501 detection capability, such has colorimetric substrate, incubation time and even the
502 temperature in the signal is measured [184], particularly in DNA-based biosensor other
503 factors can be, Lack of the ability to form complex, complication in large-scale patterns,
504 reaction induction by mistake and are sensitive to enzymatic degradation and oxidation
505 [96].
506
507 However, there are several applied technologies in the biorecognition section of the
508 biosensor device, for microbial pathogens detection, are the use of gene sequence
509 biomarkers such as, CRISPR/Cas9-based technology, where the low limit of detection
510 was 3x100 genetic copies per sample, reaching to 3x10 6 gene copies per sample [8] in this
511 study the detection signal was dose-response intensity. CRISPR/Cas12 based lateral
512 flow, where the low limit of detection was 4x10 0 gene copies in the sample [177]. Other
513 of the limitations to consider in the application of these device is the ability of the
514 biosensor to discriminate between Live/death cells (LOD 1 CFUmL -1), the use
515 functionalized NPs with bacteriophages as a biorecognition agent is a solution applied
516 for successfully discrimination between live/death [178].
517
38 Biosensors 2023, 13, x FOR PEER REVIEW 19 of 37
39

518 Conclusions
519 Over the years, significant advancements have been made in the field of biosensors,
520 particularly in areas related to food safety and the monitoring of pathogenic
521 microorganisms associated with food and waterborne illnesses. Despite these
522 achievements, progress in technologies for the development of biosensors applied to
523 pathogen detection remains a highly promising area of study. This is due to the presence
524 of various nanomaterials (MNPs, QD, carbon nanotubes, among others) with specific
525 properties that enable the identification of specific pathogens.
526 The nanomaterials used in biosensors offer unique advantages for pathogen detection.
527 Thanks to their small size and large specific surface area, they facilitate more effective
528 interaction with pathogenic agents, enhancing the sensitivity and selectivity of
529 biosensors. Furthermore, their capacity to be functionalized with specific molecules,
530 such as antibodies, nucleic acids, or aptamers, provides intrinsic advantages. In
531 particular, aptamers, due to their chain-like structure, offer greater flexibility and ease of
532 design, making them highly selective and sensitive agents for the precise detection of
533 pathogens.
534 In addition, pathogen detection through biosensors has a substantial impact on public
535 health. By enabling the precise and rapid identification of pathogenic agents in food and
536 water, this technology contributes to preventing disease outbreaks and taking
537 appropriate measures to ensure consumer safety.
538 In summary, the nanomaterials employed in biosensors present diverse advantages for
539 pathogen detection. Their reduced size, high specific surface area, functionalization
540 capacity, and signal amplification properties contribute to the sensitivity, selectivity, and
541 precision of biosensors. These qualities, combined with the potential to use techniques
542 such as fluorescence and others, make nanomaterial-based biosensors promising tools
543 for pathogen monitoring and detection, enhancing safety in the food industry and public
544 health overall.
545 Author Contributions:
546 Conceptualization. H.M.V.A., A.A.A. E.R.M.S; Writing— original draft preparation: H.M.V.A.,
547 A.A.A, E.R.M.S;; Data curation: H.M.V.A, A.A:A, E.R.M.S;; Writing—review and editing:
548 H.M.V.A., E.R.M-S; A.A.A, xxxxxxxxxx Funding acquisition: xxxxxxx; Supervision. J.E.S.H., R.P.S.

549 Supplementary Materials: The following supporting information can be downloaded at:
550 www.mdpi.com/xxx/s1, Figure S1: title; Table S1: title; Video S1: title.
551 Author Contributions: For research articles with several authors, a short paragraph specifying
552 their individual contributions must be provided. The following statements should be used
553 “Conceptualization, X.X. and Y.Y.; methodology, X.X.; software, X.X.; validation, X.X., Y.Y. and
554 Z.Z.; formal analysis, X.X.; investigation, X.X.; resources, X.X.; data curation, X.X.; writing—
40 Biosensors 2023, 13, x FOR PEER REVIEW 20 of 37
41

555 original draft preparation, X.X.; writing—review and editing, X.X.; visualization, X.X.; supervision,
556 X.X.; project administration, X.X.; funding acquisition, Y.Y. All authors have read and agreed to
557 the published version of the manuscript.” Please turn to the CRediT taxonomy for the term
558 explanation. Authorship must be limited to those who have contributed substantially to the work
559 reported.
560 Funding: Please add: “This research received no external funding” or “This research was funded
561 by NAME OF FUNDER, grant number XXX” and “The APC was funded by XXX”. Check carefully
562 that the details given are accurate and use the standard spelling of funding agency names at
563 https://search.crossref.org/funding. Any errors may affect your future funding.
564 Institutional Review Board Statement: In this section, you should add the Institutional Review
565 Board Statement and approval number, if relevant to your study. You might choose to exclude this
566 statement if the study did not require ethical approval. Please note that the Editorial Office might
567 ask you for further information. Please add “The study was conducted in accordance with the
568 Declaration of Helsinki, and approved by the Institutional Review Board (or Ethics Committee) of
569 NAME OF INSTITUTE (protocol code XXX and date of approval).” for studies involving humans.
570 OR “The animal study protocol was approved by the Institutional Review Board (or Ethics
571 Committee) of NAME OF INSTITUTE (protocol code XXX and date of approval).” for studies
572 involving animals. OR “Ethical review and approval were waived for this study due to REASON
573 (please provide a detailed justification).” OR “Not applicable” for studies not involving humans or
574 animals.
575 Informed Consent Statement: Any research article describing a study involving humans should
576 contain this statement. Please add “Informed consent was obtained from all subjects involved in
577 the study.” OR “Patient consent was waived due to REASON (please provide a detailed
578 justification).” OR “Not applicable.” for studies not involving humans. You might also choose to
579 exclude this statement if the study did not involve humans.
580 Written informed consent for publication must be obtained from participating patients who
581 can be identified (including by the patients themselves). Please state “Written informed consent
582 has been obtained from the patient(s) to publish this paper” if applicable.
583 Data Availability Statement: We encourage all authors of articles published in MDPI journals to
584 share their research data. In this section, please provide details regarding where data supporting
585 reported results can be found, including links to publicly archived datasets analyzed or generated
586 during the study. Where no new data were created, or where data is unavailable due to privacy or
587 ethical restrictions, a statement is still required. Suggested Data Availability Statements are
588 available in section “MDPI Research Data Policies” at https://www.mdpi.com/ethics.
589 Acknowledgments: In this section, you can acknowledge any support given which is not covered
590 by the author contribution or funding sections. This may include administrative and technical
591 support, or donations in kind (e.g., materials used for experiments).
592 Conflicts of Interest: Declare conflicts of interest or state “The authors declare no conflict of
593 interest.” Authors must identify and declare any personal circumstances or interest that may be
594 perceived as inappropriately influencing the representation or interpretation of reported research
595 results. Any role of the funders in the design of the study; in the collection, analyses or
596 interpretation of data; in the writing of the manuscript; or in the decision to publish the results
42 Biosensors 2023, 13, x FOR PEER REVIEW 21 of 37
43

597 must be declared in this section. If there is no role, please state “The funders had no role in the
598 design of the study; in the collection, analyses, or interpretation of data; in the writing of the
599 manuscript; or in the decision to publish the results”.

600 Appendix A
601 The appendix is an optional section that can contain details and data supplemental
602 to the main text—for example, explanations of experimental details that would disrupt
603 the flow of the main text but nonetheless remain crucial to understanding and
604 reproducing the research shown; figures of replicates for experiments of which
605 representative data is shown in the main text can be added here if brief, or as
606 Supplementary data. Mathematical proofs of results not central to the paper can be
607 added as an appendix.

608 Appendix B
609 All appendix sections must be cited in the main text. In the appendices, Figures,
610 Tables, etc. should be labeled starting with “A”—e.g., Figure A1, Figure A2, etc.

611 References
612 1. Estimating the burden of foodborne diseases Available online: https://www.who.int/activities/estimating-the-
613 burden-of-foodborne-diseases (accessed on 14 March 2023).
614 2. Haughton, P. La OMS intensifica sus esfuerzos para mejorar la salubridad de los alimentos y proteger a la pobla-
615 ción de las enfermedades Available online: https://www.who.int/es/news/item/07-06-2021-who-steps-up-action-
616 to-improve-food-safety-and-protect-people-from-disease (accessed on 10 May 2023).
617 3. AL-Mamun, M.; Chowdhury, T.; Biswas, B.; Absar, N. Food Poisoning and Intoxication: A Global Leading Con-
618 cern for Human Health. In Food Safety and Preservation; Elsevier, 2018; pp. 307–352 ISBN 978-0-12-814956-0.
619 4. Chin, N.A.; Salihah, N.T.; Shivanand, P.; Ahmed, M.U. Recent Trends and Developments of PCR-Based Meth-
620 ods for the Detection of Food-Borne Salmonella Bacteria and Norovirus. J Food Sci Technol 2022, 59, 4570–4582,
621 doi:10.1007/s13197-021-05280-5.
622 5. Thomas, K.M.; De Glanville, W.A.; Barker, G.C.; Benschop, J.; Buza, J.J.; Cleaveland, S.; Davis, M.A.; French,
623 N.P.; Mmbaga, B.T.; Prinsen, G.; et al. Prevalence of Campylobacter and Salmonella in African Food Animals
624 and Meat: A Systematic Review and Meta-Analysis. International Journal of Food Microbiology 2020, 315, 108382,
625 doi:10.1016/j.ijfoodmicro.2019.108382.
626 6. Dos Santos, J.S.; Biduski, B.; Dos Santos, L.R. Listeria Monocytogenes: Health Risk and a Challenge for Food
627 Processing Establishments. Arch Microbiol 2021, 203, 5907–5919, doi:10.1007/s00203-021-02590-2.
44 Biosensors 2023, 13, x FOR PEER REVIEW 22 of 37
45

628 7. EFSA BIOHAZ Panel; Koutsoumanis, K.; Allende, A.; Alvarez‐Ordóñez, A.; Bover‐Cid, S.; Chemaly, M.; Davies,
629 R.; De Cesare, A.; Herman, L.; Hilbert, F.; et al. Pathogenicity Assessment of Shiga Toxin‐producing Escherichia
630 Coli (STEC) and the Public Health Risk Posed by Contamination of Food with STEC. EFS2 2020, 18, doi:10.2903/
631 j.efsa.2020.5967.
632 8. Zhou, J.; Yin, L.; Dong, Y.; Peng, L.; Liu, G.; Man, S.; Ma, L. CRISPR-Cas13a Based Bacterial Detection Platform:
633 Sensing Pathogen Staphylococcus Aureus in Food Samples. Analytica Chimica Acta 2020, 1127, 225–233,
634 doi:10.1016/j.aca.2020.06.041.
635 9. Mora, Z.V.L.; Macías-Rodríguez, M.E.; Arratia-Quijada, J.; Gonzalez-Torres, Y.S.; Nuño, K.; Villarruel-López, A.
636 Clostridium Perfringens as Foodborne Pathogen in Broiler Production: Pathophysiology and Potential Strategies
637 for Controlling Necrotic Enteritis. Animals 2020, 10, 1718, doi:10.3390/ani10091718.
638 10. Enosi Tuipulotu, D.; Mathur, A.; Ngo, C.; Man, S.M. Bacillus Cereus: Epidemiology, Virulence Factors, and
639 Host–Pathogen Interactions. Trends in Microbiology 2021, 29, 458–471, doi:10.1016/j.tim.2020.09.003.
640 11. Gupta, V.; Gulati, P.; Bhagat, N.; Dhar, M.S.; Virdi, J.S. Detection of Yersinia Enterocolitica in Food: An Over-
641 view. Eur J Clin Microbiol Infect Dis 2015, 34, 641–650, doi:10.1007/s10096-014-2276-7.
642 12. Pichel, N.; Vivar, M.; Fuentes, M. The Problem of Drinking Water Access: A Review of Disinfection Technologies
643 with an Emphasis on Solar Treatment Methods. Chemosphere 2019, 218, 1014–1030, doi:10.1016/
644 j.chemosphere.2018.11.205.
645 13. Adelodun, B.; Ajibade, F.O.; Ighalo, J.O.; Odey, G.; Ibrahim, R.G.; Kareem, K.Y.; Bakare, H.O.; Tiamiyu, A.O.;
646 Ajibade, T.F.; Abdulkadir, T.S.; et al. Assessment of Socioeconomic Inequality Based on Virus-Contaminated
647 Water Usage in Developing Countries: A Review. Environmental Research 2021, 192, 110309, doi:10.1016/
648 j.envres.2020.110309.
649 14. Ramírez-Castillo, F.; Loera-Muro, A.; Jacques, M.; Garneau, P.; Avelar-González, F.; Harel, J.; Guerrero-Barrera,
650 A. Waterborne Pathogens: Detection Methods and Challenges. Pathogens 2015, 4, 307–334, doi:10.3390/
651 pathogens4020307.
652 15. Cissé, G. Food-Borne and Water-Borne Diseases under Climate Change in Low- and Middle-Income Countries:
653 Further Efforts Needed for Reducing Environmental Health Exposure Risks. Acta Tropica 2019, 194, 181–188,
654 doi:https://doi.org/10.1016/j.actatropica.2019.03.012.
655 16. Mahagamage, M.G.Y.L.; Pathirage, M.V.S.C.; Manage, P.M. Contamination Status of Salmonella Spp., Shigella
656 Spp. and Campylobacter Spp. in Surface and Groundwater of the Kelani River Basin, Sri Lanka. Water 2020, 12,
657 2187, doi:10.3390/w12082187.
658 17. Schoenen, D. Role of Disinfection in Suppressing the Spread of Pathogens with Drinking Water: Possibilities and
659 Limitations. Water Research 2002, 36, 3874–3888, doi:10.1016/S0043-1354(02)00076-3.
46 Biosensors 2023, 13, x FOR PEER REVIEW 23 of 37
47

660 18. Wen, X.; Chen, F.; Lin, Y.; Zhu, H.; Yuan, F.; Kuang, D.; Jia, Z.; Yuan, Z. Microbial Indicators and Their Use for
661 Monitoring Drinking Water Quality—A Review. Sustainability 2020, 12, 2249, doi:10.3390/su12062249.
662 19. Semenza, J.C.; Rocklöv, J.; Ebi, K.L. Climate Change and Cascading Risks from Infectious Disease. Infect Dis Ther
663 2022, 11, 1371–1390, doi:10.1007/s40121-022-00647-3.
664 20. Jahne, M.A.; Schoen, M.E.; Kaufmann, A.; Pecson, B.M.; Olivieri, A.; Sharvelle, S.; Anderson, A.; Ashbolt, N.J.;
665 Garland, J.L. Enteric Pathogen Reduction Targets for Onsite Non-Potable Water Systems: A Critical Evaluation.
666 Water Research 2023, 233, 119742, doi:https://doi.org/10.1016/j.watres.2023.119742.
667 21. Parra-Arroyo, L.; Martínez-Ruiz, M.; Lucero, S.; Oyervides-Muñoz, M.A.; Wilkinson, M.; Melchor-Martínez,
668 E.M.; Araújo, R.G.; Coronado-Apodaca, K.G.; Bedran, H.V.; Buitrón, G.; et al. Degradation of Viral RNA in
669 Wastewater Complex Matrix Models and Other Standards for Wastewater-Based Epidemiology: A Review.
670 TrAC Trends in Analytical Chemistry 2023, 158, 116890, doi:https://doi.org/10.1016/j.trac.2022.116890.
671 22. Kumar, H.; Kuča, K.; Bhatia, S.K.; Saini, K.; Kaushal, A.; Verma, R.; Bhalla, T.C.; Kumar, D. Applications of Nan-
672 otechnology in Sensor-Based Detection of Foodborne Pathogens. Sensors 2020, 20, 1966, doi:10.3390/s20071966.
673 23. Kabiraz, M.P.; Majumdar, P.R.; Mahmud, M.M.C.; Bhowmik, S.; Ali, A. Conventional and Advanced Detection
674 Techniques of Foodborne Pathogens: A Comprehensive Review. Heliyon 2023, 9, e15482, doi:10.1016/
675 j.heliyon.2023.e15482.
676 24. Clais, S.; Boulet, G.; Van kerckhoven, M.; Lanckacker, E.; Delputte, P.; Maes, L.; Cos, P. Comparison of Viable
677 Plate Count, Turbidity Measurement and Real‐time PCR for Quantification of Porphyromonas Gingivalis. Letters
678 in Applied Microbiology 2015, 60, 79–84, doi:10.1111/lam.12341.
679 25. Rajapaksha, P.; Elbourne, A.; Gangadoo, S.; Brown, R.; Cozzolino, D.; Chapman, J. A Review of Methods for the
680 Detection of Pathogenic Microorganisms. Analyst 2019, 144, 396–411, doi:10.1039/C8AN01488D.
681 26. Aw, T.G.; Rose, J.B. Detection of Pathogens in Water: From Phylochips to QPCR to Pyrosequencing. Current
682 Opinion in Biotechnology 2012, 23, 422–430, doi:https://doi.org/10.1016/j.copbio.2011.11.016.
683 27. Fu, Y.; Peng, H.; Liu, J.; Nguyen, T.H.; Hashmi, M.Z.; Shen, C. Occurrence and Quantification of Culturable and
684 Viable but Non-Culturable (VBNC) Pathogens in Biofilm on Different Pipes from a Metropolitan Drinking Wa-
685 ter Distribution System. Science of The Total Environment 2021, 764, 142851,
686 doi:https://doi.org/10.1016/j.scitotenv.2020.142851.
687 28. Bu, T.; Jia, P.; Liu, J.; Liu, Y.; Sun, X.; Zhang, M.; Tian, Y.; Zhang, D.; Wang, J.; Wang, L. Diversely Positive-
688 Charged Gold Nanoparticles Based Biosensor: A Label-Free and Sensitive Tool for Foodborne Pathogen Detec-
689 tion. Food Chemistry: X 2019, 3, 100052, doi:https://doi.org/10.1016/j.fochx.2019.100052.
48 Biosensors 2023, 13, x FOR PEER REVIEW 24 of 37
49

690 29. DeMone, C.; Hwang, M.-H.; Feng, Z.; McClure, J.T.; Greenwood, S.J.; Fung, R.; Kim, M.; Weese, J.S.; Shapiro, K.
691 Application of next Generation Sequencing for Detection of Protozoan Pathogens in Shellfish. Food and Water-
692 borne Parasitology 2020, 21, e00096, doi:https://doi.org/10.1016/j.fawpar.2020.e00096.
693 30. Eyre, D.W. Infection Prevention and Control Insights from a Decade of Pathogen Whole-Genome Sequencing.
694 Journal of Hospital Infection 2022, 122, 180–186, doi:https://doi.org/10.1016/j.jhin.2022.01.024.
695 31. Dąbrowiecki, Z.; Dąbrowiecka, M.; Olszański, R.; Siermontowski, P. Developing a Methodology for Testing and
696 Preliminary Determination of the Presence of and in Environmental Water Samples by Immunomagnetic Sepa-
697 ration Combined with Flow Cytometry. Polish Hyperbaric Research 2019, 68, 71–92, doi:10.2478/phr-2019-0013.
698 32. Hegde, M.; Pai, P.; Shetty, M.G.; Babitha, K.S. Gold Nanoparticle Based Biosensors for Rapid Pathogen Detec-
699 tion: A Review. Environmental Nanotechnology, Monitoring & Management 2022, 18, 100756,
700 doi:https://doi.org/10.1016/j.enmm.2022.100756.
701 33. Sadanandan, S.; S, M.V.; Ramkumar, K.; Pillai, N.P.; P, A.; J, S.P.; V, D.; K, R.; R, J.S.; Sreejaya, M.M. Biorecogni -
702 tion Elements Appended Gold Nanoparticle Biosensors for the Detection of Food-Borne Pathogens - A Review.
703 Food Control 2023, 148, 109510, doi:https://doi.org/10.1016/j.foodcont.2022.109510.
704 34. Wen, C.-Y.; Liang, X.; Liu, J.; Zhao, T.-Y.; Li, X.; Zhang, Y.; Guo, G.; Zhang, Z.; Zeng, J. An Achromatic Colori -
705 metric Nanosensor for Sensitive Multiple Pathogen Detection by Coupling Plasmonic Nanoparticles with Mag-
706 netic Separation. Talanta 2023, 256, 124271, doi:https://doi.org/10.1016/j.talanta.2023.124271.
707 35. Li, D.; Liu, L.; Huang, Q.; Tong, T.; Zhou, Y.; Li, Z.; Bai, Q.; Liang, H.; Chen, L. Recent Advances on Aptamer-
708 Based Biosensors for Detection of Pathogenic Bacteria. World J Microbiol Biotechnol 2021, 37, 45, doi:10.1007/
709 s11274-021-03002-9.
710 36. Chamundeeswari, M.; Jeslin, J.; Verma, M.L. Nanocarriers for Drug Delivery Applications. Environ Chem Lett
711 2019, 17, 849–865, doi:10.1007/s10311-018-00841-1.
712 37. Ghorbani, F.; Abbaszadeh, H.; Mehdizadeh, A.; Ebrahimi-Warkiani, M.; Rashidi, M.-R.; Yousefi, M. Biosensors
713 and Nanobiosensors for Rapid Detection of Autoimmune Diseases: A Review. Microchim Acta 2019, 186, 838,
714 doi:10.1007/s00604-019-3844-4.
715 38. Nnachi, R.C.; Sui, N.; Ke, B.; Luo, Z.; Bhalla, N.; He, D.; Yang, Z. Biosensors for Rapid Detection of Bacterial
716 Pathogens in Water, Food and Environment. Environment International 2022, 166, 107357, doi:10.1016/
717 j.envint.2022.107357.
718 39. Sharifi, S.; Vahed, S.Z.; Ahmadian, E.; Dizaj, S.M.; Eftekhari, A.; Khalilov, R.; Ahmadi, M.; Hamidi-Asl, E.; Labib,
719 M. Detection of Pathogenic Bacteria via Nanomaterials-Modified Aptasensors. Biosensors and Bioelectronics 2020,
720 150, 111933, doi:10.1016/j.bios.2019.111933.
50 Biosensors 2023, 13, x FOR PEER REVIEW 25 of 37
51

721 40. Cho, I.-H.; Ku, S. Current Technical Approaches for the Early Detection of Foodborne Pathogens: Challenges
722 and Opportunities. IJMS 2017, 18, 2078, doi:10.3390/ijms18102078.
723 41. López, M.M.; Ilop, P.; Noales Are Molecular Tools Solving the Challenges Posed by Detection of Plant Patho-
724 genic Bacteria and Viruses? Current Issues in Molecular Biology 2009, doi:10.21775/cimb.011.013.
725 42. Kim, J.-H.; Oh, S.-W. Rapid and Sensitive Detection of E. Coli O157:H7 and S. Typhimurium in Iceberg Lettuce
726 and Cabbage Using Filtration, DNA Concentration, and QPCR without Enrichment. Food Chemistry 2020, 327,
727 127036, doi:10.1016/j.foodchem.2020.127036.
728 43. Srivastava, K.R.; Awasthi, S.; Mishra, P.K.; Srivastava, P.K. Biosensors/Molecular Tools for Detection of Water-
729 borne Pathogens. In Waterborne Pathogens; Elsevier, 2020; pp. 237–277 ISBN 978-0-12-818783-8.
730 44. Chen, B.; Jiang, Y.; Cao, X.; Liu, C.; Zhang, N.; Shi, D. Droplet Digital PCR as an Emerging Tool in Detecting
731 Pathogens Nucleic Acids in Infectious Diseases. Clinica Chimica Acta 2021, 517, 156–161, doi:10.1016/
732 j.cca.2021.02.008.
733 45. Bulard, E.; Bouchet-Spinelli, A.; Chaud, P.; Roget, A.; Calemczuk, R.; Fort, S.; Livache, T. Carbohydrates as New
734 Probes for the Identification of Closely Related Escherichia Coli Strains Using Surface Plasmon Resonance Imag-
735 ing Available online: https://pubs.acs.org/doi/pdf/10.1021/ac5037704 (accessed on 26 April 2023).
736 46. Zhao, Y.; Zeng, D.; Yan, C.; Chen, W.; Ren, J.; Jiang, Y.; Jiang, L.; Xue, F.; Ji, D.; Tang, F.; et al. Rapid and Accu -
737 rate Detection of Escherichia Coli O157:H7 in Beef Using Microfluidic Wax-Printed Paper-Based ELISA. Analyst
738 2020, 145, 3106–3115, doi:10.1039/D0AN00224K.
739 47. Liu, S.; Hu, Q.; Li, C.; Zhang, F.; Gu, H.; Wang, X.; Li, S.; Xue, L.; Madl, T.; Zhang, Y.; et al. Wide-Range, Rapid,
740 and Specific Identification of Pathogenic Bacteria by Surface-Enhanced Raman Spectroscopy. ACS Sens. 2021, 6,
741 2911–2919, doi:10.1021/acssensors.1c00641.
742 48. Ali, A.A.; Altemimi, A.B.; Alhelfi, N.; Ibrahim, S.A. Application of Biosensors for Detection of Pathogenic Food
743 Bacteria: A Review. Biosensors 2020, 10, 58, doi:10.3390/bios10060058.
744 49. Kuswandi, B. Nanobiosensor Approaches for Pollutant Monitoring. Environ Chem Lett 2019, 17, 975–990,
745 doi:10.1007/s10311-018-00853-x.
746 50. Nanobiomaterial Engineering: Concepts and Their Applications in Biomedicine and Diagnostics; Chandra, P., Prakash,
747 R., Eds.; Springer Singapore: Singapore, 2020; ISBN 978-981-329-839-2.
748 51. Fracchiolla, N.; Artuso, S.; Cortelezzi, A. Biosensors in Clinical Practice: Focus on Oncohematology. Sensors 2013,
749 13, 6423–6447, doi:10.3390/s130506423.
750 52. Zhang, Z.; Zhou, J.; Du, X. Electrochemical Biosensors for Detection of Foodborne Pathogens. Micromachines
751 2019, 10, 222, doi:10.3390/mi10040222.
52 Biosensors 2023, 13, x FOR PEER REVIEW 26 of 37
53

752 53. Saha, K.; Agasti, S.S.; Kim, C.; Li, X.; Rotello, V.M. Gold Nanoparticles in Chemical and Biological Sensing.
753 Chem. Rev. 2012, 112, 2739–2779, doi:10.1021/cr2001178.
754 54. Tuteja, S.K.; Mutreja, R.; Neethirajan, S.; Ingebrandt, S. Bioconjugation of Different Nanosurfaces With Biorecog-
755 nition Molecules for the Development of Selective Nanosensor Platforms. In Advances in Nanosensors for Biologi-
756 cal and Environmental Analysis; Elsevier, 2019; pp. 79–94 ISBN 978-0-12-817456-2.
757 55. Zhao, X.; Smith, G.; Javed, B.; Dee, G.; Gun’ko, Y.K.; Curtin, J.; Byrne, H.J.; O’Connor, C.; Tian, F. Design and
758 Development of Magnetic Iron Core Gold Nanoparticle-Based Fluorescent Multiplex Assay to Detect Salmo-
759 nella. Nanomaterials 2022, 12, 3917, doi:10.3390/nano12213917.
760 56. Verma, M.L.; Rani, V. Biosensors for Toxic Metals, Polychlorinated Biphenyls, Biological Oxygen Demand, En-
761 docrine Disruptors, Hormones, Dioxin, Phenolic and Organophosphorus Compounds: A Review. Environ Chem
762 Lett 2021, 19, 1657–1666, doi:10.1007/s10311-020-01116-4.
763 57. Mirres, A.C. de M.; Silva, B.E.P. de M. da; Tessaro, L.; Galvan, D.; Andrade, J.C. de; Aquino, A.; Joshi, N.; Conte-
764 Junior, C.A. Recent Advances in Nanomaterial-Based Biosensors for Pesticide Detection in Foods. Biosensors
765 2022, 12, 572, doi:10.3390/bios12080572.
766 58. Munawar, A.; Ong, Y.; Schirhagl, R.; Tahir, M.A.; Khan, W.S.; Bajwa, S.Z. Nanosensors for Diagnosis with Opti-
767 cal, Electric and Mechanical Transducers. RSC Adv. 2019, 9, 6793–6803, doi:10.1039/C8RA10144B.
768 59. Javaid, M.; Haleem, A.; Singh, R.P.; Rab, S.; Suman, R. Exploring the Potential of Nanosensors: A Brief Over-
769 view. Sensors International 2021, 2, 100130, doi:10.1016/j.sintl.2021.100130.
770 60. Selvolini, G.; Marrazza, G. MIP-Based Sensors: Promising New Tools for Cancer Biomarker Determination. Sen-
771 sors 2017, 17, 718, doi:10.3390/s17040718.
772 61. Hroncekova, S.; Lorencova, L.; Bertok, T.; Hires, M.; Jane, E.; Bučko, M.; Kasak, P.; Tkac, J. Amperometric Minia-
773 turised Portable Enzymatic Nanobiosensor for the Ultrasensitive Analysis of a Prostate Cancer Biomarker. JFB
774 2023, 14, 161, doi:10.3390/jfb14030161.
775 62. Farrokhnia, M.; Amoabediny, G.; Ebrahimi, M.; Ganjali, M.; Arjmand, M. Ultrasensitive Early Detection of In -
776 sulin Antibody Employing Novel Electrochemical Nano-Biosensor Based on Controllable Electro-Fabrication
777 Process. Talanta 2022, 238, 122947, doi:10.1016/j.talanta.2021.122947.
778 63. Zhang, L.; Mazouzi, Y.; Salmain, M.; Liedberg, B.; Boujday, S. Antibody-Gold Nanoparticle Bioconjugates for
779 Biosensors: Synthesis, Characterization and Selected Applications. Biosensors and Bioelectronics 2020, 165, 112370,
780 doi:10.1016/j.bios.2020.112370.
781 64. Park, D.H.; Choi, M.Y.; Choi, J.-H. Recent Development in Plasmonic Nanobiosensors for Viral DNA/RNA
782 Biomarkers. Biosensors 2022, 12, 1121, doi:10.3390/bios12121121.
54 Biosensors 2023, 13, x FOR PEER REVIEW 27 of 37
55

783 65. Rawat, N.K.; Ghosh, R. Chapter 8 - Conducting Polymer–Based Nanobiosensors. In Nanosensors for Smart Cities;
784 Han, B., Tomer, V.K., Nguyen, T.A., Farmani, A., Kumar Singh, P., Eds.; Micro and Nano Technologies; Elsevier,
785 2020; pp. 129–142 ISBN 978-0-12-819870-4.
786 66. Fang, R.H.; Jiang, Y.; Fang, J.C.; Zhang, L. Cell Membrane-Derived Nanomaterials for Biomedical Applications.
787 Biomaterials 2017, 128, 69–83, doi:10.1016/j.biomaterials.2017.02.041.
788 67. Aliakbar Ahovan, Z.; Hashemi, A.; De Plano, L.M.; Gholipourmalekabadi, M.; Seifalian, A. Bacteriophage Based
789 Biosensors: Trends, Outcomes and Challenges. Nanomaterials 2020, 10, 501, doi:10.3390/nano10030501.
790 68. Solaimuthu, A.; Vijayan, A.N.; Murali, P.; Korrapati, P.S. Nano-Biosensors and Their Relevance in Tissue Engi -
791 neering. Current Opinion in Biomedical Engineering 2020, 13, 84–93, doi:10.1016/j.cobme.2019.12.005.
792 69. Song, M.; Yang, M.; Hao, J. Pathogenic Virus Detection by Optical Nanobiosensors. Cell Reports Physical Science
793 2021, 2, 100288, doi:10.1016/j.xcrp.2020.100288.
794 70. Negahdary, M.; Angnes, L. Electrochemical Nanobiosensors Equipped with Peptides: A Review. Microchim Acta
795 2022, 189, 94, doi:10.1007/s00604-022-05184-x.
796 71. Tsao, Y.-H.; Husain, R.A.; Lin, Y.-J.; Khan, I.; Chen, S.-W.; Lin, Z.-H. A Self-Powered Mercury Ion Nanosensor
797 Based on the Thermoelectric Effect and Chemical Transformation Mechanism. Nano Energy 2019, 62, 268–274,
798 doi:10.1016/j.nanoen.2019.05.032.
799 72. Yu, R.; Niu, S.; Pan, C.; Wang, Z.L. Piezotronic Effect Enhanced Performance of Schottky-Contacted Optical, Gas,
800 Chemical and Biological Nanosensors. Nano Energy 2015, 14, 312–339, doi:10.1016/j.nanoen.2014.11.037.
801 73. Tavakolian, M.; Jafari, S.M.; Van De Ven, T.G.M. A Review on Surface-Functionalized Cellulosic Nanostructures
802 as Biocompatible Antibacterial Materials. Nano-Micro Lett. 2020, 12, 73, doi:10.1007/s40820-020-0408-4.
803 74. Borse, V.B.; Konwar, A.N.; Jayant, R.D.; Patil, P.O. Perspectives of Characterization and Bioconjugation of Gold
804 Nanoparticles and Their Application in Lateral Flow Immunosensing. Drug Deliv. and Transl. Res. 2020, 10, 878–
805 902, doi:10.1007/s13346-020-00771-y.
806 75. Zhou, Y.; Fang, Y.; Ramasamy, R. Non-Covalent Functionalization of Carbon Nanotubes for Electrochemical
807 Biosensor Development. Sensors 2019, 19, 392, doi:10.3390/s19020392.
808 76. Fratila, R.M.; Mitchell, S.G.; Del Pino, P.; Grazu, V.; De La Fuente, J.M. Strategies for the Biofunctionalization of
809 Gold and Iron Oxide Nanoparticles. Langmuir 2014, 30, 15057–15071, doi:10.1021/la5015658.
810 77. Wang, D.-X.; Wang, J.; Wang, Y.-X.; Du, Y.-C.; Huang, Y.; Tang, A.-N.; Cui, Y.-X.; Kong, D.-M. DNA Nanostruc-
811 ture-Based Nucleic Acid Probes: Construction and Biological Applications. Chem. Sci. 2021, 12, 7602–7622,
812 doi:10.1039/D1SC00587A.
813 78. Yaraki, M.T.; Tan, Y.N. Bioconjugation of Different Nanosurfaces With Biorecognition Molecules for the Devel-
814 opment of Selective Nanosensor Platforms. Chem. Asian J. 2020, 15, 3180–3208, doi:10.1002/asia.202000847.
56 Biosensors 2023, 13, x FOR PEER REVIEW 28 of 37
57

815 79. Valenzuela-Amaro, H.M.; Vázquez Ortega, P.G.; Zazueta-Alvarez, D.E.; López-Miranda, J.; Rojas-Contreras, J.A.
816 Síntesis verde de nanopartículas de magnetita (NPs-Fe3O4): factores y limitaciones. MN 2022, 16, 1e–18e,
817 doi:10.22201/ceiich.24485691e.2023.30.69744.
818 80. Carnerero, J.M.; Jimenez‐Ruiz, A.; Castillo, P.M.; Prado‐Gotor, R. Covalent and Non‐Covalent DNA–Gold‐
819 Nanoparticle Interactions: New Avenues of Research. ChemPhysChem 2017, 18, 17–33, doi:10.1002/
820 cphc.201601077.
821 81. Greca, L.G.; Lehtonen, J.; Tardy, B.L.; Guo, J.; Rojas, O.J. Biofabrication of Multifunctional Nanocellulosic 3D
822 Structures: A Facile and Customizable Route. Mater. Horiz. 2018, 5, 408–415, doi:10.1039/C7MH01139C.
823 82. Rocchitta, G.; Spanu, A.; Babudieri, S.; Latte, G.; Madeddu, G.; Galleri, G.; Nuvoli, S.; Bagella, P.; Demartis, M.;
824 Fiore, V.; et al. Enzyme Biosensors for Biomedical Applications: Strategies for Safeguarding Analytical Perfor-
825 mances in Biological Fluids. Sensors 2016, 16, 780, doi:10.3390/s16060780.
826 83. Rahmawati, I.; Einaga, Y.; Ivandini, T.A.; Fiorani, A. Enzymatic Biosensors with Electrochemiluminescence
827 Transduction. ChemElectroChem 2022, 9, e202200175, doi:10.1002/celc.202200175.
828 84. McVey, C.; Huang, F.; Elliott, C.; Cao, C. Endonuclease Controlled Aggregation of Gold Nanoparticles for the
829 Ultrasensitive Detection of Pathogenic Bacterial DNA. Biosensors and Bioelectronics 2017, 92, 502–508,
830 doi:https://doi.org/10.1016/j.bios.2016.10.072.
831 85. Karthik, V.; Senthil Kumar, P.; Vo, D.-V.N.; Selvakumar, P.; Gokulakrishnan, M.; Keerthana, P.; Audilakshmi, V.;
832 Jeyanthi, J. Enzyme-Loaded Nanoparticles for the Degradation of Wastewater Contaminants: A Review. Environ
833 Chem Lett 2021, 19, 2331–2350, doi:10.1007/s10311-020-01158-8.
834 86. Banakar, M.; Hamidi, M.; Khurshid, Z.; Zafar, M.S.; Sapkota, J.; Azizian, R.; Rokaya, D. Electrochemical Biosen-
835 sors for Pathogen Detection: An Updated Review. Biosensors 2022, 12, 927, doi:10.3390/bios12110927.
836 87. Chiu, M.L.; Goulet, D.R.; Teplyakov, A.; Gilliland, G.L. Antibody Structure and Function: The Basis for Engi-
837 neering Therapeutics. Antibodies 2019, 8, 55, doi:10.3390/antib8040055.
838 88. Zahavi, D.; Weiner, L. Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34, doi:10.3390/an-
839 tib9030034.
840 89. McKeague, M.; DeRosa, M.C. Challenges and Opportunities for Small Molecule Aptamer Development. Journal
841 of Nucleic Acids 2012, 2012, 1–20, doi:10.1155/2012/748913.
842 90. Chavda, V.P.; Balar, P.C.; Teli, D.; Davidson, M.; Bojarska, J.; Apostolopoulos, V. Antibody–Biopolymer Conju-
843 gates in Oncology: A Review. Molecules 2023, 28, 2605, doi:10.3390/molecules28062605.
844 91. Majdi, H.; Salehi, R.; Pourhassan-Moghaddam, M.; Mahmoodi, S.; Poursalehi, Z.; Vasilescu, S. Antibody Conju-
845 gated Green Synthesized Chitosan-Gold Nanoparticles for Optical Biosensing. Colloid and Interface Science Com-
846 munications 2019, 33, 100207, doi:10.1016/j.colcom.2019.100207.
58 Biosensors 2023, 13, x FOR PEER REVIEW 29 of 37
59

847 92. Marques, A.C.; Costa, P.J.; Velho, S.; Amaral, M.H. Functionalizing Nanoparticles with Cancer-Targeting Anti-
848 bodies: A Comparison of Strategies. Journal of Controlled Release 2020, 320, 180–200, doi:10.1016/
849 j.jconrel.2020.01.035.
850 93. Lara, S.; Perez-Potti, A. Applications of Nanomaterials for Immunosensing. Biosensors 2018, 8, 104, doi:10.3390/
851 bios8040104.
852 94. Guo, R.; Huang, F.; Cai, G.; Zheng, L.; Xue, L.; Li, Y.; Liao, M.; Wang, M.; Lin, J. A Colorimetric Immunosensor
853 for Determination of Foodborne Bacteria Using Rotating Immunomagnetic Separation, Gold Nanorod Indica-
854 tion, and Click Chemistry Amplification. Microchim Acta 2020, 187, 197, doi:10.1007/s00604-020-4169-z.
855 95. Dolatabadi, J.E.N.; Mashinchian, O.; Ayoubi, B.; Jamali, A.A.; Mobed, A.; Losic, D.; Omidi, Y.; De La Guardia, M.
856 Optical and Electrochemical DNA Nanobiosensors. TrAC Trends in Analytical Chemistry 2011, 30, 459–472,
857 doi:10.1016/j.trac.2010.11.010.
858 96. Hua, Y.; Ma, J.; Li, D.; Wang, R. DNA-Based Biosensors for the Biochemical Analysis: A Review. Biosensors 2022,
859 12, 183, doi:10.3390/bios12030183.
860 97. Ma, X.; Ding, W.; Wang, C.; Wu, H.; Tian, X.; Lyu, M.; Wang, S. DNAzyme Biosensors for the Detection of Path-
861 ogenic Bacteria. Sensors and Actuators B: Chemical 2021, 331, 129422, doi:10.1016/j.snb.2020.129422.
862 98. Ahmed, M.; Patel, R. Electrochemical/Voltammetric/Amperometric Nanosensors for the Detection of Pathogenic
863 Bacteria. In Nanosensors for Point-of-Care Diagnostics of Pathogenic Bacteria; Acharya, A., Singhal, N.K., Eds.;
864 Springer Nature: Singapore, 2023; pp. 113–141 ISBN 978-981-9912-18-6.
865 99. Wu, Q.; Zhang, Y.; Yang, Q.; Yuan, N.; Zhang, W. Review of Electrochemical DNA Biosensors for Detecting
866 Food Borne Pathogens. Sensors 2019, 19, 4916, doi:10.3390/s19224916.
867 100. D’Agata, R.; Bellassai, N.; Jungbluth, V.; Spoto, G. Recent Advances in Antifouling Materials for Surface Plas -
868 mon Resonance Biosensing in Clinical Diagnostics and Food Safety. Polymers 2021, 13, 1929, doi:10.3390/
869 polym13121929.
870 101. Bhardwaj, N.; Bhardwaj, S.K.; Nayak, M.K.; Mehta, J.; Kim, K.-H.; Deep, A. Fluorescent Nanobiosensors for the
871 Targeted Detection of Foodborne Bacteria. TrAC Trends in Analytical Chemistry 2017, 97, 120–135, doi:10.1016/j.-
872 trac.2017.09.010.
873 102. Leng, X.; Wang, Y.; Li, R.; Liu, S.; Yao, J.; Pei, Q.; Cui, X.; Tu, Y.; Tang, D.; Huang, J. Circular Exponential Ampli-
874 fication of Photoinduced Electron Transfer Using Hairpin Probes, G-Quadruplex DNAzyme and Silver Nan-
875 ocluster-Labeled DNA for Ultrasensitive Fluorometric Determination of Pathogenic Bacteria. Microchim Acta
876 2018, 185, 168, doi:10.1007/s00604-018-2698-5.
60 Biosensors 2023, 13, x FOR PEER REVIEW 30 of 37
61

877 103. Zhou, Y.; Wang, Z.; Zhang, S.; Deng, L. An Ultrasensitive Fluorescence Detection Template of Pathogenic Bacte-
878 ria Based on Dual Catalytic Hairpin DNA Walker@Gold Nanoparticles Enzyme-Free Amplification. Spectrochim-
879 ica Acta Part A: Molecular and Biomolecular Spectroscopy 2022, 277, 121259, doi:10.1016/j.saa.2022.121259.
880 104. Chinnappan, R.; AlZabn, R.; Abu-Salah, K.M.; Zourob, M. An Aptamer Based Fluorometric Microcystin-LR As -
881 say Using DNA Strand-Based Competitive Displacement. Microchim Acta 2019, 186, 435, doi:10.1007/s00604-019-
882 3504-8.
883 105. Tessaro, L.; Aquino, A.; De Almeida Rodrigues, P.; Joshi, N.; Ferrari, R.G.; Conte-Junior, C.A. Nucleic Acid-
884 Based Nanobiosensor (NAB) Used for Salmonella Detection in Foods: A Systematic Review. Nanomaterials 2022,
885 12, 821, doi:10.3390/nano12050821.
886 106. Cui, F.; Sun, J.; De Dieu Habimana, J.; Yang, X.; Ji, J.; Zhang, Y.; Lei, H.; Li, Z.; Zheng, J.; Fan, M.; et al. Ultrasensi-
887 tive Fluorometric Angling Determination of Staphylococcus Aureus in Vitro and Fluorescence Imaging in Vivo
888 Using Carbon Dots with Full-Color Emission. Anal. Chem. 2019, 91, 14681–14690, doi:10.1021/
889 acs.analchem.9b03916.
890 107. Gupta, R.; Kumar, A.; Kumar, S.; Pinnaka, A.K.; Singhal, N.K. Naked Eye Colorimetric Detection of Escherichia
891 Coli Using Aptamer Conjugated Graphene Oxide Enclosed Gold Nanoparticles. Sensors and Actuators B: Chemi-
892 cal 2021, 329, 129100, doi:10.1016/j.snb.2020.129100.
893 108. Pandit, C.; Alajangi, H.K.; Singh, J.; Khajuria, A.; Sharma, A.; Hassan, Md.S.; Parida, M.; Semwal, A.D.; Gopalan,
894 N.; Sharma, R.K.; et al. Development of Magnetic Nanoparticle Assisted Aptamer-Quantum Dot Based Biosen-
895 sor for the Detection of Escherichia Coli in Water Samples. Science of The Total Environment 2022, 831, 154857,
896 doi:10.1016/j.scitotenv.2022.154857.
897 109. Sande, M.G.; Rodrigues, J.L.; Ferreira, D.; Silva, C.J.; Rodrigues, L.R. Novel Biorecognition Elements against
898 Pathogens in the Design of State-of-the-Art Diagnostics. Biosensors 2021, 11, 418, doi:10.3390/bios11110418.
899 110. Singh, A.; Sharma, A.; Ahmed, A.; Sundramoorthy, A.K.; Furukawa, H.; Arya, S.; Khosla, A. Recent Advances in
900 Electrochemical Biosensors: Applications, Challenges, and Future Scope. Biosensors 2021, 11, 336, doi:10.3390/
901 bios11090336.
902 111. Feyziazar, M.; Amini, M.; Jahanban-Esfahlan, A.; Baradaran, B.; Oroojalian, F.; Kamrani, A.; Mokhtarzadeh, A.;
903 Soleymani, J.; De La Guardia, M. Recent Advances on the Piezoelectric, Electrochemical, and Optical Biosensors
904 for the Detection of Protozoan Pathogens. TrAC Trends in Analytical Chemistry 2022, 157, 116803, doi:10.1016/j.-
905 trac.2022.116803.
906 112. Saleh Ibrahim, Y.; Alexis Ramírez-Coronel, A.; Kumar Sain, D.; Haleem Al-qaim, Z.; Hassan Jawhar, Z.; Yaseen
907 Mahmood Alabdali, A.; Hayif Jasim Ali, S.; Althomali, R.H.; Fakri Mustafa, Y.; Mireya Romero-Parra, R. Ad-
62 Biosensors 2023, 13, x FOR PEER REVIEW 31 of 37
63

908 vances in Nanomaterials-Based Chemiluminescence (Bio)Sensor for Specific and Sensitive Determination of
909 Pathogenic Bacteria. Microchemical Journal 2023, 191, 108860, doi:10.1016/j.microc.2023.108860.
910 113. Sun, F.; Zhang, J.; Yang, Q.; Wu, W. Quantum Dot Biosensor Combined with Antibody and Aptamer for Tracing
911 Food-Borne Pathogens. Food Quality and Safety 2021, 5, fyab019, doi:10.1093/fqsafe/fyab019.
912 114. Sun, R.; Zou, H.; Zhang, Y.; Zhang, X.; Chen, L.; Lv, R.; Sheng, R.; Du, T.; Li, Y.; Wang, H.; et al. Vancomycin
913 Recognition and Induced-Aggregation of the Au Nanoparticles through Freeze-Thaw for Foodborne Pathogen
914 Staphylococcus Aureus Detection. Analytica Chimica Acta 2022, 1190, 339253,
915 doi:https://doi.org/10.1016/j.aca.2021.339253.
916 115. Singh, P.; Gupta, R.; Choudhary, M.; Pinnaka, A.K.; Kumar, R.; Bhalla, V. Drug and Nanoparticle Mediated
917 Rapid Naked Eye Water Test for Pathogens Detection. Sensors and Actuators B: Chemical 2018, 262, 603–610,
918 doi:https://doi.org/10.1016/j.snb.2018.02.033.
919 116. Hu, J.; Fu, K.; Bohn, P.W. Whole-Cell Pseudomonas Aeruginosa Localized Surface Plasmon Resonance Aptasen-
920 sor. Anal. Chem. 2018, 90, 2326–2332, doi:10.1021/acs.analchem.7b04800.
921 117. Lim, S.H.; Ryu, Y.C.; Hwang, B.H. Aptamer-Immobilized Gold Nanoparticles Enable Facile and On-Site Detec-
922 tion of Staphylococcus Aureus. Biotechnology and Bioprocess Engineering 2021, 26, 107–113, doi:10.1007/s12257-020-
923 0161-z.
924 118. Yang, X.; Huang, R.; Xiong, L.; Chen, F.; Sun, W.; Yu, L. A Colorimetric Aptasensor for Ochratoxin A Detection
925 Based on Tetramethylrhodamine Charge Effect-Assisted Silver Enhancement. Biosensors 2023, 13, 468,
926 doi:10.3390/bios13040468.
927 119. Dester, E.; Kao, K.; Alocilja, E.C. Detection of Unamplified E. Coli O157 DNA Extracted from Large Food Sam-
928 ples Using a Gold Nanoparticle Colorimetric Biosensor. Biosensors 2022, 12, 274, doi:10.3390/bios12050274.
929 120. Kaushal, S.; Pinnaka, A.K.; Soni, S.; Singhal, N.K. Antibody Assisted Graphene Oxide Coated Gold Nanoparti-
930 cles for Rapid Bacterial Detection and near Infrared Light Enhanced Antibacterial Activity. Sensors and Actuators
931 B: Chemical 2021, 329, 129141, doi:https://doi.org/10.1016/j.snb.2020.129141.
932 121. Abbaspour, A.; Norouz-Sarvestani, F.; Noori, A.; Soltani, N. Aptamer-Conjugated Silver Nanoparticles for Elec-
933 trochemical Dual-Aptamer-Based Sandwich Detection of Staphylococcus Aureus. Biosensors and Bioelectronics
934 2015, 68, 149–155, doi:https://doi.org/10.1016/j.bios.2014.12.040.
935 122. Dabhade, A.H.; Verma, R.P.; Paramasivan, B.; Kumawat, A.; Saha, B. Development of Silver Nanoparticles and
936 Aptamer Conjugated Biosensor for Rapid Detection of E. Coli in a Water Sample. 3 Biotech 2023, 13, 244,
937 doi:10.1007/s13205-023-03663-3.
64 Biosensors 2023, 13, x FOR PEER REVIEW 32 of 37
65

938 123. Imran, M.; Ehrhardt, C.J.; Bertino, M.F.; Shah, M.R.; Yadavalli, V.K. Chitosan Stabilized Silver Nanoparticles for
939 the Electrochemical Detection of Lipopolysaccharide: A Facile Biosensing Approach for Gram-Negative Bacteria.
940 Micromachines 2020, 11, 413, doi:10.3390/mi11040413.
941 124. Shahdost-Fard, F.; Faridfar, S.; Keihan, A.H.; Aghaei, M.; Petrenko, I.; Ahmadi, F.; Ehrlich, H.; Rahimi-
942 Nasrabadi, M. Applicability of a Green Nanocomposite Consisted of Spongin Decorated Cu2WO4(OH)2 and
943 AgNPs as a High-Performance Aptasensing Platform in Staphylococcus Aureus Detection. Biosensors 2023, 13,
944 271, doi:10.3390/bios13020271.
945 125. Yin, M.; Liu, C.; Ge, R.; Fang, Y.; Wei, J.; Chen, X.; Chen, Q.; Chen, X. Paper-Supported near-Infrared-Light-Trig -
946 gered Photoelectrochemical Platform for Monitoring Escherichia Coli O157:H7 Based on Silver Nanoparticles-
947 Sensitized-Upconversion Nanophosphors. Biosensors and Bioelectronics 2022, 203, 114022,
948 doi:https://doi.org/10.1016/j.bios.2022.114022.
949 126. Qaanei, M.; Taheri, R.A.; Eskandari, K. Electrochemical Aptasensor for Escherichia Coli O157:H7 Bacteria Detec-
950 tion Using a Nanocomposite of Reduced Graphene Oxide, Gold Nanoparticles and Polyvinyl Alcohol. Anal.
951 Methods 2021, 13, 3101–3109, doi:10.1039/D1AY00563D.
952 127. Ertaş, T.; Dinç, B.; Üstünsoy, R.; Eraslan, H.; Ergenç, A.F.; Bektaş, M. Novel Electrochemical Biosensor for Es -
953 cherichia Coli Using Gold-Coated Tungsten Wires and Antibody Functionalized Short Multiwalled Carbon
954 Nanotubes. Instrumentation Science & Technology 2023, 0, 1–16, doi:10.1080/10739149.2023.2222801.
955 128. Wang, Z.; Yao, X.; Wang, R.; Ji, Y.; Yue, T.; Sun, J.; Li, T.; Wang, J.; Zhang, D. Label-Free Strip Sensor Based on
956 Surface Positively Charged Nitrogen-Rich Carbon Nanoparticles for Rapid Detection of Salmonella Enteritidis.
957 Biosensors and Bioelectronics 2019, 132, 360–367, doi:https://doi.org/10.1016/j.bios.2019.02.061.
958 129. Kurt, H.; Yüce, M.; Hussain, B.; Budak, H. Dual-Excitation Upconverting Nanoparticle and Quantum Dot Ap-
959 tasensor for Multiplexed Food Pathogen Detection. Biosensors and Bioelectronics 2016, 81, 280–286,
960 doi:https://doi.org/10.1016/j.bios.2016.03.005.
961 130. Mathelié-Guinlet, M.; Cohen-Bouhacina, T.; Gammoudi, I.; Martin, A.; Béven, L.; Delville, M.-H.; Grauby-Hey-
962 wang, C. Silica Nanoparticles-Assisted Electrochemical Biosensor for the Rapid, Sensitive and Specific Detection
963 of Escherichia Coli. Sensors and Actuators B: Chemical 2019, 292, 314–320,
964 doi:https://doi.org/10.1016/j.snb.2019.03.144.
965 131. Jenie, S.N.A.; Kusumastuti, Y.; Krismastuti, F.S.H.; Untoro, Y.M.; Dewi, R.T.; Udin, L.Z.; Artanti, N. Rapid Fluo-
966 rescence Quenching Detection of Escherichia Coli Using Natural Silica-Based Nanoparticles. Sensors 2021, 21,
967 881, doi:10.3390/s21030881.
66 Biosensors 2023, 13, x FOR PEER REVIEW 33 of 37
67

968 132. Wu, Z.; Sun, D.-W.; Pu, H.; Wei, Q. A Dual Signal-on Biosensor Based on Dual-Gated Locked Mesoporous Silica
969 Nanoparticles for the Detection of Aflatoxin B1. Talanta 2023, 253, 124027,
970 doi:https://doi.org/10.1016/j.talanta.2022.124027.
971 133. Boodoo, C.; Dester, E.; David, J.; Patel, V.; Kc, R.; Alocilja, E.C. Multi-Probe Nano-Genomic Biosensor to Detect S.
972 Aureus from Magnetically-Extracted Food Samples. Biosensors 2023, 13, 608, doi:10.3390/bios13060608.
973 134. Diouani, M.F.; Sayhi, M.; Djafar, Z.R.; Ben Jomaa, S.; Belgacem, K.; Gharbi, H.; Ghita, M.; Popescu, L.-M.;
974 Piticescu, R.; Laouini, D. Magnetic Separation and Centri-Chronoamperometric Detection of Foodborne Bacteria
975 Using Antibiotic-Coated Metallic Nanoparticles. Biosensors 2021, 11, 205, doi:10.3390/bios11070205.
976 135. Wen, J.; Ren, L.; He, Q.; Bao, J.; Zhang, X.; Pi, Z.; Chen, Y. Contamination-Free V-Shaped Ultrafast Reaction Cas -
977 cade Transferase Signal Amplification Driven CRISPR/Cas12a Magnetic Relaxation Switching Biosensor for Bac-
978 teria Detection. Biosensors and Bioelectronics 2023, 219, 114790, doi:https://doi.org/10.1016/j.bios.2022.114790.
979 136. Ganganboina, A.B.; Chowdhury, A.D.; Khoris, I.M.; Doong, R.; Li, T.-C.; Hara, T.; Abe, F.; Suzuki, T.; Park, E.Y.
980 Hollow Magnetic-Fluorescent Nanoparticles for Dual-Modality Virus Detection. Biosensors and Bioelectronics
981 2020, 170, 112680, doi:https://doi.org/10.1016/j.bios.2020.112680.
982 137. Kulkarni, M.B.; Ayachit, N.H.; Aminabhavi, T.M. Biosensors and Microfluidic Biosensors: From Fabrication to
983 Application. Biosensors 2022, 12, 543, doi:10.3390/bios12070543.
984 138. Liu, L.; Hong, J.; Wang, W.; Xiao, S.; Xie, H.; Wang, Q.; Gan, N. Fluorescent Aptasensor for Detection of Live
985 Foodborne Pathogens Based on Multicolor Perovskite-Quantum-Dot-Encoded DNA Probes and Dual-Stirring-
986 Bar-Assisted Signal Amplification. Journal of Pharmaceutical Analysis 2022, 12, 913–922,
987 doi:https://doi.org/10.1016/j.jpha.2022.07.001.
988 139. Shang, Y.; Cai, S.; Ye, Q.; Wu, Q.; Shao, Y.; Qu, X.; Xiang, X.; Zhou, B.; Ding, Y.; Chen, M.; et al. Quantum Dot
989 Nanobeads-Labelled Lateral Flow Immunoassay Strip for Rapid and Sensitive Detection of Salmonella Ty-
990 phimurium Based on Strand Displacement Loop-Mediated Isothermal Amplification. Engineering 2022, 19, 62–
991 70, doi:https://doi.org/10.1016/j.eng.2021.03.024.
992 140. Alafeef, M.; Moitra, P.; Pan, D. Nano-Enabled Sensing Approaches for Pathogenic Bacterial Detection. Biosensors
993 and Bioelectronics 2020, 165, 112276, doi:https://doi.org/10.1016/j.bios.2020.112276.
994 141. Ayodhya, D. Recent Progress on Detection of Bivalent, Trivalent, and Hexavalent Toxic Heavy Metal Ions in
995 Water Using Metallic Nanoparticles: A Review. Results in Chemistry 2023, 5, 100874,
996 doi:https://doi.org/10.1016/j.rechem.2023.100874.
997 142. Patel, R.; Mitra, B.; Vinchurkar, M.; Adami, A.; Patkar, R.; Giacomozzi, F.; Lorenzelli, L.; Baghini, M.S. A Review
998 of Recent Advances in Plant-Pathogen Detection Systems. Heliyon 2022, 8, e11855, doi:10.1016/
999 j.heliyon.2022.e11855.
68 Biosensors 2023, 13, x FOR PEER REVIEW 34 of 37
69

1000 143. Loiseau, A.; Asila, V.; Boitel-Aullen, G.; Lam, M.; Salmain, M.; Boujday, S. Silver-Based Plasmonic Nanoparticles
1001 for and Their Use in Biosensing. Biosensors 2019, 9, 78, doi:10.3390/bios9020078.
1002 144. Ibrahim, N.; Jamaluddin, N.D.; Tan, L.L.; Mohd Yusof, N.Y. A Review on the Development of Gold and Silver
1003 Nanoparticles-Based Biosensor as a Detection Strategy of Emerging and Pathogenic RNA Virus. Sensors 2021, 21,
1004 5114, doi:10.3390/s21155114.
1005 145. Yu, X.; Jiao, Y.; Chai, Q. Applications of Gold Nanoparticles in Biosensors. Nano LIFE 2016, doi:10.1142/
1006 S1793984416420010.
1007 146. Baetsen-Young, A.M.; Vasher, M.; Matta, L.L.; Colgan, P.; Alocilja, E.C.; Day, B. Direct Colorimetric Detection of
1008 Unamplified Pathogen DNA by Dextrin-Capped Gold Nanoparticles. Biosensors and Bioelectronics 2018, 101, 29–
1009 36, doi:https://doi.org/10.1016/j.bios.2017.10.011.
1010 147. Hui, C.; Hu, S.; Yang, X.; Guo, Y. A Panel of Visual Bacterial Biosensors for the Rapid Detection of Genotoxic
1011 and Oxidative Damage: A Proof of Concept Study. Mutation Research/Genetic Toxicology and Environmental Muta-
1012 genesis 2023, 888, 503639, doi:https://doi.org/10.1016/j.mrgentox.2023.503639.
1013 148. Liu, J.; Xu, J.-Z.; Rao, Z.-M.; Zhang, W.-G. An Enzymatic Colorimetric Whole-Cell Biosensor for High-Through-
1014 put Identification of Lysine Overproducers. Biosensors and Bioelectronics 2022, 216, 114681,
1015 doi:https://doi.org/10.1016/j.bios.2022.114681.
1016 149. Miranda, R.R.; Sampaio, I.; Zucolotto, V. Exploring Silver Nanoparticles for Cancer Therapy and Diagnosis. Col-
1017 loids and Surfaces B: Biointerfaces 2022, 210, 112254, doi:https://doi.org/10.1016/j.colsurfb.2021.112254.
1018 150. Douaki, A.; Demelash Abera, B.; Cantarella, G.; Shkodra, B.; Mushtaq, A.; Ibba, P.; Inam, A.S.; Petti, L.; Lugli, P.
1019 Flexible Screen Printed Aptasensor for Rapid Detection of Furaneol: A Comparison of CNTs and AgNPs Effect
1020 on Aptasensor Performance. Nanomaterials 2020, 10, 1167, doi:10.3390/nano10061167.
1021 151. Li, G. Nano-Inspired Biosensors for Protein Assay with Clinical Applications; Elsevier, 2018; ISBN 978-0-12-815054-2.
1022 152. Varghese Alex, K.; Tamil Pavai, P.; Rugmini, R.; Shiva Prasad, M.; Kamakshi, K.; Sekhar, K.C. Green Synthesized
1023 Ag Nanoparticles for Bio-Sensing and Photocatalytic Applications. ACS Omega 2020, 5, 13123–13129,
1024 doi:10.1021/acsomega.0c01136.
1025 153. Srivastava, K.R.; Awasthi, S.; Mishra, P.K.; Srivastava, P.K. Chapter 13 - Biosensors/Molecular Tools for Detec-
1026 tion of Waterborne Pathogens. In Waterborne Pathogens; Prasad, M.N.V., Grobelak, A., Eds.; Butterworth-Heine-
1027 mann, 2020; pp. 237–277 ISBN 978-0-12-818783-8.
1028 154. Nguyen, T.N.; Phung, V.-D.; Tran, V.V. Recent Advances in Conjugated Polymer-Based Biosensors for Virus
1029 Detection. Biosensors 2023, 13, 586, doi:10.3390/bios13060586.
70 Biosensors 2023, 13, x FOR PEER REVIEW 35 of 37
71

1030 155. Bakhshandeh, B.; Sorboni, S.G.; Haghighi, D.M.; Ahmadi, F.; Dehghani, Z.; Badiei, A. New Analytical Methods
1031 Using Carbon-Based Nanomaterials for Detection of Salmonella Species as a Major Food Poisoning Organism in
1032 Water and Soil Resources. Chemosphere 2022, 287, 132243, doi:https://doi.org/10.1016/j.chemosphere.2021.132243.
1033 156. Sharma, A.; Sharma, N.; Kumari, A.; Lee, H.-J.; Kim, T.; Tripathi, K.M. Nano-Carbon Based Sensors for Bacterial
1034 Detection and Discrimination in Clinical Diagnosis: A Junction between Material Science and Biology. Applied
1035 Materials Today 2020, 18, 100467, doi:https://doi.org/10.1016/j.apmt.2019.100467.
1036 157. Bhattacharya, K.; Mukherjee, S.P.; Gallud, A.; Burkert, S.C.; Bistarelli, S.; Bellucci, S.; Bottini, M.; Star, A.; Fadeel,
1037 B. Biological Interactions of Carbon-Based Nanomaterials: From Coronation to Degradation. Nanomedicine: Nan-
1038 otechnology, Biology and Medicine 2016, 12, 333–351, doi:https://doi.org/10.1016/j.nano.2015.11.011.
1039 158. Duan, N.; Wu, S.; Dai, S.; Miao, T.; Chen, J.; Wang, Z. Simultaneous Detection of Pathogenic Bacteria Using an
1040 Aptamer Based Biosensor and Dual Fluorescence Resonance Energy Transfer from Quantum Dots to Carbon
1041 Nanoparticles. Microchim Acta 2015, 182, 917–923, doi:10.1007/s00604-014-1406-3.
1042 159. Shen, Y.; Zhang, Y.; Gao, Z.F.; Ye, Y.; Wu, Q.; Chen, H.-Y.; Xu, J.-J. Recent Advances in Nanotechnology for Si -
1043 multaneous Detection of Multiple Pathogenic Bacteria. Nano Today 2021, 38, 101121,
1044 doi:https://doi.org/10.1016/j.nantod.2021.101121.
1045 160. Fang, Y.; Wang, Y.; Zhu, L.; Liu, H.; Su, X.; Liu, Y.; Chen, Z.; Chen, H.; He, N. A Novel Cartridge for Nucleic
1046 Acid Extraction, Amplification and Detection of Infectious Disease Pathogens with the Help of Magnetic
1047 Nanoparticles. Chinese Chemical Letters 2023, 34, 108092, doi:https://doi.org/10.1016/j.cclet.2022.108092.
1048 161. Hussain, C.M. Analytical Applications of Functionalized Magnetic Nanoparticles; Royal Society of Chemistry, 2021;
1049 ISBN 978-1-83916-276-3.
1050 162. Huang, H.T.; Garu, P.; Li, C.H.; Chang, W.C.; Chen, B.W.; Sung, S.Y.; Lee, C.M.; Chen, J.Y.; Hsieh, T.F.; Sheu,
1051 W.J.; et al. Magnetoresistive Biosensors for Direct Detection of Magnetic Nanoparticle Conjugated Biomarkers
1052 on a Chip. SPIN 2019, 09, 1940002, doi:10.1142/S2010324719400022.
1053 163. Min, C.; Shao, H.; Liong, M.; Yoon, T.-J.; Weissleder, R.; Lee, H. Mechanism of Magnetic Relaxation Switching
1054 Sensing. ACS Nano 2012, 6, 6821–6828, doi:10.1021/nn301615b.
1055 164. Arya, S.; Singh, A.; Sharma, A.; Gupta, V. 11 - Silicon-Based Biosensor. In Silicon-Based Hybrid Nanoparticles;
1056 Thomas, S., Nguyen, T.A., Ahmadi, M., Yasin, G., Joshi, N., Eds.; Micro and Nano Technologies; Elsevier, 2022;
1057 pp. 247–267 ISBN 978-0-12-824007-6.
1058 165. Bagheri, E.; Ansari, L.; Sameiyan, E.; Abnous, K.; Taghdisi, S.M.; Ramezani, M.; Alibolandi, M. Sensors Design
1059 Based on Hybrid Gold-Silica Nanostructures. Biosensors and Bioelectronics 2020, 153, 112054,
1060 doi:https://doi.org/10.1016/j.bios.2020.112054.
72 Biosensors 2023, 13, x FOR PEER REVIEW 36 of 37
73

1061 166. Costanzo, H.; Gooch, J.; Frascione, N. Nanomaterials for Optical Biosensors in Forensic Analysis. Talanta 2023,
1062 253, 123945, doi:https://doi.org/10.1016/j.talanta.2022.123945.
1063 167. Aggett, P.; Aguilar, F.; Crebelli, R.; Dusemund, B.; Filipič, M.; Frutos, M.J.; Galtier, P.; Gott, D.; Gundert‐Remy,
1064 U.; Kuhnle, G.G.; et al. Re‐evaluation of Silicon Dioxide (E 551) as a Food Additive. EFS2 2018, 16, doi:10.2903/
1065 j.efsa.2018.5088.
1066 168. Şen Karaman, D.; Pamukçu, A.; Karakaplan, M.B.; Kocaoglu, O.; Rosenholm, J.M. Recent Advances in the Use of
1067 Mesoporous Silica Nanoparticles for the Diagnosis of Bacterial Infections. IJN 2021, Volume 16, 6575–6591,
1068 doi:10.2147/IJN.S273062.
1069 169. Wen, L.; Qiu, L.; Wu, Y.; Hu, X.; Zhang, X. Aptamer-Modified Semiconductor Quantum Dots for Biosensing
1070 Applications. Sensors 2017, 17, 1736, doi:10.3390/s17081736.
1071 170. Sun, H.; Zhou, P.; Su, B. Electrochemiluminescence of Semiconductor Quantum Dots and Its Biosensing Appli-
1072 cations: A Comprehensive Review. Biosensors 2023, 13, 708, doi:10.3390/bios13070708.
1073 171. Ma, F.; Li, C.; Zhang, C. Development of Quantum Dot-Based Biosensors: Principles and Applications. J. Mater.
1074 Chem. B 2018, 6, 6173–6190, doi:10.1039/C8TB01869C.
1075 172. Pourmadadi, M.; Rahmani, E.; Rajabzadeh-Khosroshahi, M.; Samadi, A.; Behzadmehr, R.; Rahdar, A.; Ferreira,
1076 L.F.R. Properties and Application of Carbon Quantum Dots (CQDs) in Biosensors for Disease Detection: A Com -
1077 prehensive Review. Journal of Drug Delivery Science and Technology 2023, 80, 104156,
1078 doi:https://doi.org/10.1016/j.jddst.2023.104156.
1079 173. Smith, R.P.; Barraza, I.; Quinn, R.J.; Fortoul, M.C. Chapter One - The Mechanisms and Cell Signaling Pathways
1080 of Programmed Cell Death in the Bacterial World. In Cell Death Regulation in Health and Disease - Part B; Spetz,
1081 J.K.E., Galluzzi, L., Eds.; International Review of Cell and Molecular Biology; Academic Press, 2020; Vol. 352, pp.
1082 1–53.
1083 174. Hao, L.; Xue, L.; Huang, F.; Cai, G.; Qi, W.; Zhang, M.; Han, Q.; Wang, Z.; Lin, J. A Microfluidic Biosensor Based
1084 on Magnetic Nanoparticle Separation, Quantum Dots Labeling and MnO2 Nanoflower Amplification for Rapid
1085 and Sensitive Detection of Salmonella Typhimurium. Micromachines 2020, 11, 281, doi:10.3390/mi11030281.
1086 175. Schmitz, F.R.W.; Valério, A.; De Oliveira, D.; Hotza, D. An Overview and Future Prospects on Aptamers for
1087 Food Safety. Appl Microbiol Biotechnol 2020, 104, 6929–6939, doi:10.1007/s00253-020-10747-0.
1088 176. Pirzada, M.; Altintas, Z. Nanomaterials for Healthcare Biosensing Applications. Sensors 2019, 19, 5311,
1089 doi:10.3390/s19235311.
1090 177. Mukama, O.; Wu, J.; Li, Z.; Liang, Q.; Yi, Z.; Lu, X.; Liu, Y.; Liu, Y.; Hussain, M.; Makafe, G.G.; et al. An Ultrasen -
1091 sitive and Specific Point-of-Care CRISPR/Cas12 Based Lateral Flow Biosensor for the Rapid Detection of Nucleic
1092 Acids. Biosensors and Bioelectronics 2020, 159, 112143, doi:https://doi.org/10.1016/j.bios.2020.112143.
74 Biosensors 2023, 13, x FOR PEER REVIEW 37 of 37
75

1093 178. Li, Y.; Xie, G.; Qiu, J.; Zhou, D.; Gou, D.; Tao, Y.; Li, Y.; Chen, H. A New Biosensor Based on the Recognition of
1094 Phages and the Signal Amplification of Organic-Inorganic Hybrid Nanoflowers for Discriminating and Quanti-
1095 tating Live Pathogenic Bacteria in Urine. Sensors and Actuators B: Chemical 2018, 258, 803–812,
1096 doi:https://doi.org/10.1016/j.snb.2017.11.155.
1097 179. Zhang, X.; Li, G.; Wu, D.; Li, X.; Hu, N.; Chen, J.; Chen, G.; Wu, Y. Recent Progress in the Design Fabrication of
1098 Metal-Organic Frameworks-Based Nanozymes and Their Applications to Sensing and Cancer Therapy. Biosen-
1099 sors and Bioelectronics 2019, 137, 178–198, doi:https://doi.org/10.1016/j.bios.2019.04.061.
1100 180. Ahmed, A.; Rushworth, J.V.; Hirst, N.A.; Millner, P.A. Biosensors for Whole-Cell Bacterial Detection. Clin Micro-
1101 biol Rev 2014, 27, 631–646, doi:10.1128/CMR.00120-13.
1102 181. Sivakumar, R.; Lee, N.Y. Recent Advances in Airborne Pathogen Detection Using Optical and Electrochemical
1103 Biosensors. Analytica Chimica Acta 2022, 1234, 340297, doi:https://doi.org/10.1016/j.aca.2022.340297.
1104 182. Echeverri, D.; Orozco, J. Glycan-Based Electrochemical Biosensors: Promising Tools for the Detection of Infec-
1105 tious Diseases and Cancer Biomarkers. Molecules 2022, 27, 8533, doi:10.3390/molecules27238533.
1106 183. Puchkova, A.; Vietz, C.; Pibiri, E.; Wünsch, B.; Sanz Paz, M.; Acuna, G.P.; Tinnefeld, P. DNA Origami Nanoan-
1107 tennas with over 5000-Fold Fluorescence Enhancement and Single-Molecule Detection at 25 ΜM. Nano Lett.
1108 2015, 15, 8354–8359, doi:10.1021/acs.nanolett.5b04045.
1109 184. Chen, J.; Alcaine, S.D.; Jiang, Z.; Rotello, V.M.; Nugen, S.R. Detection of Escherichia Coli in Drinking Water Using
1110 T7 Bacteriophage-Conjugated Magnetic Probe. Anal. Chem. 2015, 87, 8977–8984, doi:10.1021/
1111 acs.analchem.5b02175.
1112

1113 Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
1114 author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury
1115 to people or property resulting from any ideas, methods, instructions or products referred to in the content.

You might also like