Open access peer-reviewed chapter

The Novel Nanomaterials Based Biosensors and Their Applications

Written By

Kübra Gençdağ Şensoy and Mihrican Muti

Submitted: 07 June 2020 Reviewed: 06 November 2020 Published: 07 December 2020

DOI: 10.5772/intechopen.94930

From the Edited Volume

Novel Nanomaterials

Edited by Karthikeyan Krishnamoorthy

Chapter metrics overview

716 Chapter Downloads

View Full Metrics

Abstract

Since the development of the first biosensor reported, biosensor has received considerable attention due to its high selectivity and sensitivity. Biosensors are highly pursued in order to meet the growing demands and challenges in a large number of analytic applications such as medical diagnosis, food safety control, environmental monitoring, or even military defense. Due to the unique physical, chemical, mechanical and electrical properties, nanomaterials have been widely investigated for their ability and used to fabricate sensors. High surface to volume ratio, good stability, excellent electrocatalytic properties of the nanomaterials plays an important role in the sensitive and selective detection of biomolecules. The synthesis of new nanomaterials with different properties is increasingly common in order to improve these counted properties of nanomaterials. This chapter gives an overview of the importance of the development of novel nanomaterials based biosensors technologies. The use of different funtionalized carbon nanomaterilas, metal oxide nanoparticles, metal nanoparticles, polymeric nanoparticles, quantum dots, graphene sheets and other novel nanomaterials in biosensor technology, and their innovations and advantages are discussed.

Keywords

  • novel nanomaterials
  • biosensor
  • biorecognition
  • nanosensors

1. Introduction

A biosensor device is defined as a biological or bio-inspired receptor unit with unique specificities for analytes. These analytes are generally of biological origin. One of the challenges in biosensor development is that efficient signal capture can be achieved with biological recognition. Novel nanomaterials represent a rapidly developing field in bioanalysis applications. The sensitivity and performance of biosensors can be improved by using nanomaterials. Typical schemeatic presentation of a biosensor is illuatrated in Figure 1.

Figure 1.

Schematic presentation of a biosensor.

With the development of nanotechnology, many new nanomaterials such as gold nanostructure, magnetic nanoparticles, nanozymes, and carbon-based nanomaterials have been synthesized [1]. Nanomaterials have been widely applied in the areas of invivo imaging [2], cancer treatment [3], drug delivery [4], catalysis [5], bacteriostasis [6], and so on. Due to the outstanding physical and chemical properties of nanomaterials, nanomaterial-based biosensors have been developed [7].

In this chapter, synthesis, properties and possible applications of these materials in biosensors were examined. The high sensitivity and selectivity of nanomaterial-based biosensors have led to major advances in the development of new methodologies for early detection. Due to its submicron dimensions, it allows simple and fast analysis in vivo. Their reactivity, toughness and other properties are also dependent on their unique shape, size and structure. In addition, the application of nanomaterials to biosensors provides different detection limits depending on the samples to be analyzed and facilitates the adjustment of the sensitivity level according to the needs.

Advertisement

2. Experimental

2.1 Synthesize of the novel nanomaterials

2.1.1 Graphdiyne

The synthesis of the GDY was reported in the literature as follows:

Graphdiyne (GDY) was prepared on the copper surface by a cross-linking reaction using hexaetynylbenzene (HEB) as a monomer. Firstly, hexakis [(trimethylsilyl) ethynyl] benzene (HEB-TMS) was prepared using Negishi cross-linking reaction. Then HEB monomer was obtained by the addition of tetrabutylammonium fluoride into tetrahydrofuran solution of HEB-TMS with stirring at 0°C for 10 minutes. Finally, GDY in the presence of pyridine was successfully grown on the surface of copper foils by a cross-coupling reaction of the HEB monomer for 72 hours at 60°C under a nitrogen atmosphere. After the reaction was completed, GDY grown on copper foils was removed by ultrasonic treatment and concentrated by rotary evaporator, and then washed with heated acetone and N,N-dimethylformamide. The GDY powder was refluxed with dilute hydrochloric acid and sodium hydroxide, respectively at 80°C for 3 hours. It was then washed repeatedly and centrifuged. Finally, black GDY powder was obtained by centrifugation and drying vacuum [8].

2.1.2 Gold nanostructures

According to the modified El-Sayed method the synthesis of the gold nanorods (GNRs) was performed as follows:

Ttwo solutions were prepared as seed solution and growth solution. For the seed solution, ice-cold sodium borohydride (NaBH4) (0.3 mL, 0.01 M) was added to the solution mixture containing hydrogen tetrachloroaurate (HAuCl4) (0.5 mM) and cetyltrimethylammonium bromide (CTAB) (0.2 M) in a volume ratio of 1:1, and the entire reaction mixture was incubated at room temperature for 3 hours. For the growth solution, a 200 mL solution containing HAuCl4 (0.5 mM) and CTAB (0.1 M) was made and 6 mL of silver nitrate (AgNO3) (4 mM) was added to it. Following this, 0.5 M sulfuric acid (H2SO4) (1 mL) and 0.0788 M ascorbic acid (1.4 mL) were added and mixed gently. In the last step, the seed solution (0.24 mL) was added to the growth solution mixture and left at room temperature for a period of 12 hours. The brownish colored solution was centrifuged at 9000 rpm (2 times) for 30 minutes to remove unbound CTAB and stored at room temperature (28°C) [9].

2.1.3 Inorganic nanomaterials

The synthesis of Zirconium Phosphate Nanoparticle (ZrP-NP) is described in this section as one of the inorganic nanomaterials.

Zirconium Phosphate (ZrP), one of the inorganic nanomaterials, has been synthesized by hydrothermal process. First, 1.6 g of ZrOCl2.8H2O was added to 30 mL of DI water and stirred continuously. Then 15 M H3PO4 (10 mL) was added to this prepared solution and stirred continuously for 30 minutes. The solution was transferred to a hydrothermal autoclave (50 mL) and heated in an air oven at 200°C for 24 hours. The products obtained were collected by centrifugation and washed several times with ethanol and deionized water. In the last step, the purified ZrP powder was dried in an air oven at 50°C [10].

2.1.4 Nanozymes

The synthesis of the core-shell Au@Co-Fe hybrid nanoparticles is described as peroxidase mimetic nanozyme.

In the synthesis of the core-shell Au@Co-Fe hybrid nanoparticles as the peroxidase mimetic nanozyme, gold nanoparticles (AuNPs) with the average diameter of 22 nm were synthesized by citrate reduction of HAuCl4. Briefly, 1.5 mL of 1% (w/v) sodium citrate solution was added to 21 mL of 0.8 mM HAuCl4.3H2O solution at boiling point while the solution was stirred vigorously. After hanging its color from pale yellow to deep red, the mixture was stirred for 15 min and let to cool to room temperature and, then stored at 4°C until use. In the second step, 1 μL tween 20 was added to 1.5 mL of the synthesized AuNPs. Then, 100 μL of FeSO4 0.18 M and 180 μL of CoCl2 0.1 M were added to the mixture, and incubated at the room temperature for 24 h. After that, the mixture was centrifuged and washed with deionized water [11].

2.1.5 Hybrid nanocomposites

One of the hybrid nanocomposites is reduced graphene oxide-magnetite nanoparticle (RGO-Fe3O4 NP) and its synthesis is described below according to the literature [12].

Reduce graphene oxide magnetite nanoparticle (RGO-Fe3O4 NP) hybrid was synthesized by alkaline reduction. For this purpose, the powder was redispersed in the 0.5 mg mL−1 graphene oxide (GO) suspension. Citric and ascorbic acids were added and the mixture was stirred at 55°C (12 hours). 1 M NaOH was added and the mixture It was stirred again at 95°C (6 hours). After centrifugation at 10000 rpm (RCF = 1118 x g), the solid is filtered, washed, and dried in vacuum during 24 hours [12].

2.1.6 DNA nanomaterials

Y-DNA was prepared by mixing equimolar amounts of three single stranded DNA (ssDNA), two long and one short. The two long sequences have regions that hybridize to the shorter one. One of the fields is not completely linked to the corresponding fragment. Thus, the target miRNA became able to replace this fragment and remove the Y-DNA nanostructure. ssDNAs were dissolved in hybridization buffer at 10 μM final concentration per sequence and annealed to form the desired Y-shaped DNA: annealed at 95°C for 2 minutes, cooled to 65°C and incubated for 5 minutes, followed by 2 minutes while its temperature dropped to 60°C and cooled to 20°C at a rate of 1° per minute. The final products were stored at 4°C. Double stranded substrates were formed by mixing in the hybridization buffer. The mixture was heated to 95°C for 5 minutes and slowly cooled to 4°C, then allowed to stand at room temperature for 20 minutes to form a specific double stranded substrate [13].

2.1.7 DNAzyme

DNA phosphorylation was made by incubating 200 pmol of FS1 with 20 units of T4 polynucleotide kinase (PNK) at 37°C for 30 min in a 100 μL reaction mixture containing 50 mM Tris–HCl (pH 7.6 at 25°C), 10 mM MgCl2, 5 mM 1,4-Dithiothreitol (DTT), 0.1 mM spermidine and 1 mM adenosine 5′-triphosphate (ATP). The reaction was stopped by heating the mixture at 90° C for 5 minutes. RFT1 (100 μM) and 2 μL RFS1 (100 μM) were then added to the solution, and the mixture was heated to 90°C for 40 seconds and cooled to room temperature for 10 minutes. In the last step, 10 units of T4 DNA ligase were added for DNA ligation at 25°C for 2 hours. The ligation mix contains 10 mM MgCl2, (150 μL) 40 mM Tris–HCl (pH 7.6 at 25°C), 10 mM DTT and 0.5 mM ATP. The products were concentrated by standard ethanol precipitation and further purified by polyacrylamide gel electrophoresis [14].

2.1.8 Carbon Nanodots

The syntheses of carbon nanodots (CDs) will describe according to the literature [15].

CDs were synthesized hydrothermally with citric acid and ethylenediamine (EDA). Initially citric acid (3.0 g) and ethylenediamine (1875 μL) were dissolved in 30 mL of distilled water. The solution was then transferred to a 500 mL round bottom flask and heated at 150°C for 5 hours. The product was dialyzed against ddH2O to obtain CDs. CDs powder was obtained by evaporating, redispersed in deionized water, and stored at 4°C for later use [15].

2.1.9 Carbon black nanomaterials

Carbon black (CB) is produced by the reaction of a hydrocarbon fuel such as gas or oil with a limited supply of combustion air at temperatures of 1320 to 1540°C. The hydrocarbons which were degraded from polyethylene (PE) or high density polyethylene (HDPE) at the pyrolysis step were injected into decomposing chamber. They were introduced to pass through dc-plasma jet, and were decomposed into the carbon particles. The carbon particles were cooled down in the stream of nitrogen and they were deposited on the surface of outer graphite chamber after decomposition by the plasma jet. As-synthesized carbon black samples were characterized by the analytical instrument without further purification in the case of carbon black synthesis. Two major processes are the oil furnace process and the thermal process. The oil furnace process accounts for about 90 percent of production, and the thermal, about 10 percent. Two other processes are, the lamp for production of lamp black and the cracking of acetylene to produce acetylene black. However, these are small-volume specialty black operations that constitute less than 1 percent of total production in this country [16].

2.1.10 Nanodiamonds

For the nanodiamond synthesis the graphitic C3N4 (g-C3N4) used for the starting material which prepared by a benzenethermal reaction between C3N3Cl3 and NaNH2 at 220°C for 12 hours. For the synthesis of the C3N4, 1.10 g (6.0 mmol) C3N3Cl3(1,3,5-trichlorotriazine) and 0.70 g (18.0 mmol) NaNH2 (sodium amide) powders were put into a 50 mL teflon-lined autoclave, which was then filled with benzene up to 90% of the total volume. The autoclave was sealed and maintained at 180–220°C for 8–12 h, then allowed to cool to room temperature naturally. The mixed product was washed three times with distilled water, acetone and again distilled water to remove NaCl impurities, some organic-like impurities. The g-C3N4 obtained in such a way is a light yellowish brown powder of amorphous-like, poorly crystalline particles [8]. The resulting yellow powders was dried in vacuum at 50°C for several hours. The sample was compressed to a desired pressure at room temperature, heated to 800–2000°C for 5–30 min, and then quenched and decompressed to ambient condition [17].

2.1.11 Magnetic nanoparticles

Magnetic nanoparticles (MNP) were prepared by chemical co-precipitation and then processed under hydrothermal conditions. Briefly, iron (II) chloride and iron (III) chloride (1:2) were chemically precipitated at room temperature (25°C) by adding 30% ammonium hydroxide at pH=10.0–10.4. The precipitates were heated at 80°C for 35 minutes with continuous stirring and washed in deionized water and ethanol [18].

Advertisement

3. Result and discussion

3.1 Graphdiyne

Graphdiyne (GDY) is a new two-dimensional all-carbon allotrope composed of benzene rings and alkyne unites.

The carbon based nanomaterials are usually used to build electrochemical biosensors because of their physical and chemical properties. According to conventional carbon nanomaterials, GDY possesses richer carbon chemical bonds, which are of great importance for their practical applications. More importantly, GDY has a typical 2D structure similar to graphene, but also has the properties of three-dimensional materials such as a hard carbon network and uniformly distributed pores that can greatly increase active bonding areas [19, 20].

Figure 2 illustrates surface characterization of GDY [21].

Figure 2.

A) SEM, B) TEM and C) HRTEM of GDY.

As can be seen from this figure it is clear that GDY has a porous structure which is very important in sensor design to the effective diffusion of the analyte to the sensor surface.

There are studies in which GDY has been used in the preparation of electrochemical enzyme biosensors [21], for microRNA testing [22] and in the determination of bacterium [23]. GDY was investigated as matrix for tyrosinase (a model enzyme) immobilization to create a mediator-free GDY based biosensor for rapid detection of bisphenol A (BPA). In this study between different carbon nanomaterial based biosensors including carbon nanotube and graphene was compared and it was reported, GDY-based tyrosinase biosensor performed better analytical for BPA detection than CNTs and graphene-based biosensors [21]. A new photoactive material has been synthesized that integrates the properties of MoS2 and GDY to implement ultra-sensitive detection of microRNA [22]. Controllable synthesis of two-dimensional graphite nanosheet (GDY NS) is of great importance for the clinical diagnosis and treatment of tuberculosis [23].

It is though that as a new promising 2D all-carbon nanomaterial after graphene, graphdiyne with intriguing properties would inevitably attract the general interest of scientists.

3.2 Gold nanostructures

Metal nanoparticles (NPs) such as gold and silver NPs have gained immense recognition in nanosensing and diagnostic applications [24, 25]. Therefore, ease of synthesis, versatile surface functionalization and long term stability of gold nanomaterials increases their potential as efficient detection probes [26].

Gold nanostars modified with biotin were used for streptavidin determination [27]. Sensing applications using other shapes of gold nanomaterials include the use of gold nanowires and nanocubes for detection of bacteria in human kidney infection and catechol, respectively [28, 29].

Gold nanorods have also employed as a SERS substrate where in they have achieved highly sensitive and selective detection of DNA [30].

It has been reported that the nanosensor based on gold nanorodes is highly reproducible and has excellent selectivity. It was also reported the nanosensing platform is reliable, facile, cost-effective and less labor intensive. The nanomaterial with aspect ratio tunable property can be possibly used for several biomedical applications.

Figure 3 illusrates TEM and SEM images of some kind of gold nanosructures [27, 28, 29, 30].

Figure 3.

TEM (A,B) and SEM (C,D) images of gold nanostar (A) [27], gold nanorods (B) [28], gold nanoparticle (C) [29] and gold nanowire (D) [30].

3.3 Inorganic novel nanomaterials

Recently, inorganic nanostructured materials have gained widespread attention as potential electrode materials of electrochemical sensors with excellent structural adjustability and other properties [31, 32].

In the past few years, binary metal oxides (denoted BMOs) are considered as one of the state-of-the-art electrocatalyst materials for various electrochemical applications [33, 34]. Among the different categories of BMOs, transition-metal phosphates/phosphides (denoted TMPs) have attracted increasing attention as a promising electrocatalyst [35, 36, 37]. Ultrathin cobalt phosphate-based modified electrode was used for the non enzymatic electrochemical determination of glucose [38]. α-zirconium phosphate (α-ZrP) based electrocatalysts have been recognized as crucial for numerous electrochemical applications [39]. The sensitive electrochemical sensing probe using the ZrP nanoplates was successfully applied for Furazolidone detection [10].

Figure 4 illustrates surface characterization of ZrP [10].

Figure 4.

(A−C) FEG-SEM image, (D−F) TEM images, (G) EDX spectrum, and (H−J) elemental mapping of ZrP.

Figure 5.

Comparison of (A) natural enzyme-based immunoassays and (B) nanozyme-based immunoassays [46].

3.4 Nanozymes

In the last decade, artificial nanomaterials, which exhibit properties similar to enzymes, have been shown as highly stable and low-cost alternatives to enzymes in electrochemical biosensing.

Nanozymes, combining the advantages of chemical catalysts and enzymes [40, 41], outperform natural enzymes because they are usually synthetized using low-cost, simple, and mass-production methods and offer high operational stability and self-life, robust catalytic performance [42, 43, 44, 45]. Moreover, the smooth surface modification of nanomaterials provides more room for modifications than the natural enzymes. In addition, their inherent nanomaterial properties impart them both tunable and tenable catalytic activity [46, 47].

Figure 5 illustrates the schematic presentation of the enzyme-based and nanozyme-based immunoassay.

The lack of selectivity of nanozymes is compensated for by using specific bioreceptors. However, it is important to be aware of the current lack of bio-ligands for emerging analytes and that their use compromises both stability and the low cost of nanozymes [48].

Affinity ligand-based electrochemical biosensors using nanozymes have been successfully developed and exhibit some excellent merits such as higher selectivity and sensitivity, lower cost, shorter detection time, and better signal readout [49].

Nanozymes, being a special type of nanomaterial, can be exploited in electrochemical affinity biosensing as electrode modifiers, nanocarriers, and/or catalytic labels. These multi functional nanozymes, which include PtNPs/CoTPP/rGO [49], Pd/APTES-MCeO2-GS [50], rGO-NR-Au@Pt [51], Mn3O4 and Pd@Pt nanoflowers [52], Fe3O4/PDDA/Au@Pt [53], MWCNTs/ GQDs [54, 55], and FeS2-AuNPs [56], have been decorated with detector antibody (Ab2) [49, 50], detector antibody (Ab2) + HRP [5154, 55], AuNPs + Ab2 [56], detector aptamer (Apt2) + HRP [47], or (Apt2) + HRP + G-quadruplex/hemin DNAzyme [46]. It is important to note that these nanozymes are often dressed with the natural enzyme to further enhance the sensitivity [51, 54, 55].

The combination of nanozyme-based electrochemical affinity biosensors with personalized equipment such as smartphones and/or portable low-cost devices will also be exciting to move forward in point-of-care testing. This nanozymes development to achieve catalytic activity and efficiency comparable or even better than natural enzymes will bring a revolution to conventional electrochemical biosensing and more practical applications in other expectation fields.

3.5 Hybrid nanocomposites

Hybrid sensing materials, which are organized by interaction of organic molecules onto inorganic supports, have been developed as a novel and hopeful class of hybrid sensing probes. Magnetic silica hybrid rather than other hybrid materials such as polymer, titania, and selfassembled monolayers [57, 58, 59, 60] provides low toxicity, simple separation via external magnetic field, stability, biocompatibility and thermally stable advantages [61, 62, 63].

Biosensors prepared using hybrid materials were used to detect biological materials by thermal, electrical or optical signals. Examples of various applications of biosensors can be mentioned as environmental monitoring [64], forensic science [65, 66, 67], water characteristic testing [68], defense and the military [69], biomedicine, food industry and medical diagnosis [70].

Magnetic silica hybrids were reported as fluorescent, colorimetric, electrochemical and Surface-enhanced raman spectroscopy (SERS) sensing probes [71]. Inorganic mesoporous material is one of the best materials as molecular catalysts due to its thermal stability, easy production and modification. It used in the fields of biomedicine, electronics, and physicochemistry. Silica-coated Fe3O4 nanoparticle (Fe3O4@SiO2 NPs), have good excellent conductivity, electrochemical transducers, biocompatibility, catalytic activity, separation ability and low toxicity properties to produce “electronic wires” to increase the electron transfer between redox centers and electrode surfaces in proteins [72].

Figure 6 illustrates the surface characterization of Fe3O4@ SiO2 nanoparticles performed by transmission electron microscopy (TEM) [57].

Figure 6.

TEM images of Fe3O4@ SiO2 NPs with Fe3O4 sizes of (a) 8.8 nm and (b−d) 12.2 nm.

3.6 DNA nanomaterials

DNA nanomaterials have been widely used in bioassays due to their promising properties for sensitive and specific detection of biomolecules. The electrochemical biosensor has received greater attention in clinical diagnosis due to its high sensitivity, easy controllability and low cost [72]. For this reason, the biomolecular recognition and signal amplification based on electrochemical platform to achieve miRNAs detection still need to be considered.

In recent years, legion nucleic acid nanostructures have been applied to biological detection, including DNA tetrahedron, DNA gels, DNA dendrimers, and so on [73, 74, 75]. Y-shaped DNA (Y-DNA), as a constant nanostructure with high selectivity, provides an effective method for completely measuring target molecules [76]. Y-DNA consists of three oligonucleotides that are partially hybridized to each other. Some older biosensors used this feature to perform DNA detections where one DNA stand is fixed to the surface, another DNA stand and target DNA are added to form a specific structure [77].

Numbers of signal amplification strategies have been developed, including hybridization chain reaction (HCR), strand displacement amplification (SDA), catalytic hairpin assembly (CHA) and rolling circle amplification (RCA) [78, 79, 80, 81]. The HCR consists of a trigger sequence and two partially complementary hairpin probes. Once triggered, the two hairpin probes can autonomously hybridize continuously [82].

Compared to HCR, this reaction consists of more complex components, including a trigger sequence, two double stranded substrates with bridging loops in the middle, and two helper sequences [83]. Thus, non-linear HCR can achieve higher rates of amplification and molecular weights [84].

To join non-linear HCR and Y-DNA nanostructures, the Y-DNA’s terminals were designed as triggers that could initiate the amplification reaction. As a result, the new biosensing method can provide high-precision and selective detection of biological molecules. An unlabeled DNA nanostructured electrochemical biosensor was designed to detect miRNA-25, which is reported to be a potential molecular biomarker for non-small cell lung cancer and heart failure [85, 86].

Expanding the application of DNA nanomaterials to bioassays in the future may enable early and effective detection of various diseases.

3.7 DNAzyme

DNAzymes are single-stranded (ss) DNA sequences are able to catalyze a number of reactions, including cleavage of the phosphodiester backbone at a ribonucleotide or deoxyribonucleotide site [87]. It has been shown that metal ions play an important role in the catalytic process and are essential for the catalytic activity of most known DNAzymes [88].

The ability to select a DNAzyme with metal ion specific activity without previous chemical knowledge of the DNAzyme structure, and then to subsequently modify DNAzyme binding arms and other insignificant nucleotides with minimal to no effect on sensitivity and selectivity has made DNAzymes ideal metal-selective components for new metal ion sensing technologies. RNA-cleaving DNAzyme is a very useful biomaterial for the determination of metal ions, but some parts of DNAzymes can be cleaved by several metal ions, which makes different concentrations of metal ions difficult to distinguish [89].

In the last two decades, the rapid development of nanomaterials and biomaterials [90] offers more opportunities to improve electrochemical sensor performance. For the determination of Cu (II) and Hg (II), many highly sensitive sensors are manufactured using small molecules, peptides, proteins and antibodies at low cost.

The ligand sites of proteases composed of nitrogen, oxygen or sulfur can combine with heavy metal ions to form a stable complex [91]. Cu(II) is a small ion that has to be chelated first and then bind to the antibody recognition [92]. Both antibody and enzyme work best under physiological conditions that limit application in real environment. DNA is not only the genetic material of most living organisms, but also an excellent biological functional material [93].

Metal ions can be specifically bound with a single-stranded DNA to form a stable metal-mediated DNA, and this mechanism is applied to detect metal ions [9495]. Therefore, numerous studies have focused on the newly discovered biosensor using different DNA-based aptamers functionalized with nanomaterials to increase sensitivity. DNAzymes that break down RNA as DNA-based catalysts are obtained through in vitro selection, which turned out to be a very useful platform for the identification of metal ions. After binding with heavy metal ions, many biochemical and biophysical studies have been conducted on DNAzymes due to their high metal ion selectivity and high catalytic efficiency [96]. Therefore, DNAzymes have been applied in various biosensors (colorimetric, electrochemical and fluorescent) that realize the detection of various metal ions such as Mg(II) [97], Ag(I) [98], Pb(II) [99], Zn(II) [100], Hg(II) [101], UO2(II) [102].

The field of DNAzyme-based metal ion sensing is continuing to develop for future cellular and portable detection technologies.

3.8 Carbon nanodots

Carbon dots (CDs) are nanomaterials less than 10 nm in size and became the new potential material for the electrode modifier [103]. Formerly, CDs have been applied in electrochemical sensing platforms, mainly focusing on their electrocatalytic properties toward analytes of interest [104, 105] rather than electrode modifiers. Thus, the studies on carbon dots owing a noticeable potential to be used as electrode modifiers in electrochemical techniques to increase the sensitivity of the electrochemical sensor has been exploited.

Recently, a new member of CDs, have gained attention because of their water solubility, fine properties, high luminescence, low cytotoxicity and good conductivity [106]. Depending on the precursors employed in their synthesis, CNDs are surrounded by different functional groups including, among others, hydroxyl, amide groups and carboxyl which facilitate the immobilization of biomolecules. Hence, due to their ability to be modified with a wide variety of biomolecules, and in conjunction with the excellent properties mentioned above, CNDs have been employed in many biological applications such as solar cell development and photocatalysis [107, 108]. Concerning the employment of CNDs for electrochemical biosensors, it should be highlighted that despite the previously mentioned advantages, very few attempts to incorporate CNDs into electrodes are reported. Reporting the application of CNDs in electrochemical sensors are focused on the electrocatalytic properties of this nanomaterial toward oxygen reduction [109], biomedical application [110], exploited for glucose biosensing [111] and DNA sensing [112].

Transmission electron microscopy (TEM) of carbon nanodots in different scale from 20 nm to 2 nm are illustrate in Figure 7 [110].

Figure 7.

High resolution transmission electron microscopic images of fish scale derived carbon nanodots (a-c).

3.9 Carbon black nanomaterials

Since the discovery of carbon nanotubes, carbon-based nanomaterials being researched in various disciplines including electrochemistry. An old and cost-effective material recently called carbon black (CB) reinvented. CB has good electrical conductivity, dispersible in solvents, possibility of easy functionalization and has a large number of defect areas and fast electron transfer kinetics [113, 114, 115, 116].

Previously, CB’s main application in the electrochemical field was based on the design of sensors for analyte detection in fuel cell and gas phases for lithium and sodium batteries [117, 118]. However, until 2009, only a few CB-based electrochemical sensors were reported for analyte detection.

Among nanomaterials, CB demonstrated high potential in customizing all from the oldest carbon paste to glassy carbon and printed electrodes thanks to their fascinating electrochemical properties combined with cost effectiveness.

One of the main properties of CB is its ability to produce easily stable dispersions in a variety of solvents such as ethanol, acetonitrile, a mixture of dimethylformamide water [119], chitosan [120], or di hexadecylphosphate water solution [121], usually at a concentration of 1 mg/mL.

CB is widely used in the design of biosensors with a variety of biological recognition elements including enzymes, DNA and antibodies. The main potential of the enzyme combination with CB is based on the outstanding advantages this nanomaterial has in enhancing the biosensor sensitivity. CB can increase both conductivity and enzyme loading areas, thus causing increased signals and hence higher sensitivity. Some examples have shown that CB is a compatible substrate for the immobilization of enzymes in the design of amperometric biosensors [122].

Immunosensors have attracted great attention for specific, sensitive, cost-effective and in-field analysis. Examples of CB-based immunosensors in unlabeled configuration have been reported in the literature [123, 124].

Alongside traditional bioreceptors such as enzymes, antibodies, and nucleic acids, CB also demonstrated the ability to improve their analytical performance by combining with alternative biological recognition elements or molecularly imprinted polymers [125].

Besides the biosensor application, CB was used in sensor design for both single analyte detection and multiple analysis, showing increased sensitivity thanks to its high conductivity, number of defective areas and surface area. Nowadays, most CB-based detection systems are mainly sensors, but in recent years there has been a sharp increase in publications in the development of enzymatic, immuno, and DNA biosensors [126].

CB is a new generation material due to its environmentally friendly properties in terms of costs and environmental impact.

The morphological properties of the synthesized carbon black by using commercial and waste polystyrene (PS) and high density polyethylene in different pyrolysis conditions were illustrated in Figure 8 [16].

Figure 8.

FE-SEM images of the carbon black obtained from: A commercial PS pyrolyzed at 500°C; b commercial PS pyrolyzed at 900°C; c waste PS pyrolyzed at 500°C; d waste PS pyrolyzed at 900°C; e high density polyethylene (HDPE) pyrolyzed at 500°C; f HDPE pyrolyzed at 900°C.

3.10 Nanodiamonds

Nanodiamonds (ND), a new member of the carbon nanoparticle class, has recently received much attention in drug delivery, bio-imaging, and biosensor applications due to its physical and chemical properties [127].

Nanodiamond (ND) is of great interest in various fields of material science due to its various functional groups. An electrochemical biosensor containing copper, nano-diamond (ND) and carbon nanotube (CNT) was built to detect the amino acids of Parkia Seeds (PS). Electrochemical reaction of PS was carried out with composite electrodes prepared using nanodiamond [128].

The AFM and SEM characterization of nanocrystalline diamond (NCD) and boron doped nanocrystalline diamond (BDND) were illustrated in Figure 9 respectively [129, 130].

Figure 9.

(a) AFM topographic images of NCD films and (b) SEM image of BDND film grown on a Si substrate.

3.11 Magnetic nanoparticles

Nanomaterials provide high surface areas and a biocompatible environment for enzyme loading. In the last decade, research of magnetic particles has resulted in their use in a large number of nano-sensing devices, providing ease of separation in solution.

Various iron magnetic nanoparticles (MNPs) have proven to be an excellent nanomaterial for electrochemical biosensing applications due to their electroconductivity, biocompatibility and ease of synthesis properties. They make important contributions to the development of electrochemical nanobiosensors. Functionalized magnetic nanoparticles can be directed by the external magnetic field to site-specific drug delivery targets. Iron and iron oxide nanoparticles have been studied as signal amplification elements in biosensing [131]. Among these materials, magnetite (Fe3O4), a Fe2+ and Fe3+ complex oxide, is one of the most studied super paramagnetic nanoparticles. It has unique mesoscopic mechanical and physical properties and has many potential applications in various fields such as cell separation [132] and microwave absorption [133]. Fe3O4 nanoparticles have been widely used for in vivo examination [134]. The direct binding of cholesterol oxidase to Fe3O4 magnetic nanoparticles was investigated and the kinetic behavior, stability and activity of bound cholesterol were investigated [135]. Due to its easy preparation process, low toxicity, strong superparamagnetism and good biocompatibility, Fe3O4 has recently been used in biosensors for glucose, ethanol and acetaminophen. Prepared biosensors showed fast response and high sensitivity with a wide linear range [136, 137]. Fe3O4 - Au nanoparticles, have been successfully used for the first time in the dual-mode detection of carcinoembryonics antigens (CEA) and have correctly confirmed the presence of antigens [138].

Figure 10 illustrates TEM images of Fe3O4, Au and Fe3O4-Au nanoparticles [138].

Figure 10.

TEM images of (A) Fe3O4, (B) Au and (C) Fe3O4–Au nanoparticles; the corresponding HRTEM images are inserted.

Table 1 illustrates the studies based novel nanomaterials.

NanomaterialAnalyzedDetection limitLinear rangeMethodRef.
GraphdiyneBisphenol A1,0 × 10−7-3,5 × 10−6 mol/L24 nmol/LCV1[21]
Hybrid NanocompositeMetronidazole0,001–2444 μM0,8 nMCV and EIS2[34]
Inorganic nanomaterialFurazolidone (FZD)0,009–339 μM1,2 nMCV, EIS and Amperometry[10]
Noble metal nanoparticlesAlpha fetoprotein (AFP0,1 pg./mL to 50 ng/mL0,033 pg./mLCV[50]
Bimetallic Pt-Au/multi-walled carbon nanotubesOrganophosphorous pesticides50 to 500 nmol/L29,7 nmol/LCV, Amperometric i-t curve and EIS[64]
Quantum dotsDopamine0,375–450 μM100 nMElectro-
chemiluminescence
[66]
DNAzyme-functionalized single-walled carbon nanotubesCu(II) and Hg(II)Cu(II) 0,01–10,000 nM Hg(II) 5–10,000 nMCu(II) 6,7 pM Hg(II) 3,43 nMEIS[89]
DNAzyme Functionalized Single-Walled Carbon NanotubeSilver Ion10 pM to 106 pM5 pMUV–Vis Spectrometry[98]
Carbon nanodotsGene mutation0,001–20 μM.0,16 nMCV and DPV3[112]
Carbon-coated nickel magnetic nanoparticlesAcetaminophen2,0 × 10−6 to 2,3 × 10−4 mol/L.6,0 × 10−7 mol/LDPV[137]
Carbon blackBisphenol A0,03 μ M0,1–0,9 μM
1–50 μM
SVW4[139]
3D DNA nanonet structureMicroRNA36,083 fM10 fM-1 nMCV, DPV and EIS[140]
Carbon nanodot17ß-Estradiol0,5 × 10−12 M1,0 × 10−7 - 1,0 × 10−12 MCV and EIS[141]
Carbon blackPhotosynthetic herbicide0,1–5 mu M1 nMAmperometric measurement[142]
Metal-polymer hybrid nanomaterialHuman papillomavirus1–100 pg. mu/L2,74 pg. mu/LCV and EIS[143]
Nanozymes (magnetic metal organik framework)Hydrogen peroxide (H2O2)5 mu M-120 mM0,9 mu MCV, EIS and Amperometry[144]
Gold nanorodAflatoxin0,25–10 ng/mL0,11 ng/mLSPR5[145]
NanodiamondUrea0,1–0,9 mg/mL0,005 mg/mLDirect current voltage[146]
Carbon dots, chitosan, gold nanoparticlesPatulin1 × 10−12 - 1 × 10−9 mol/L7,57 × 10−13 mol/LCV and DPV[147]

Table 1.

Biosensor applications based navel nanomaterials.

1: Cyclic voltammetry, 2: Electrochemical impedance spectrometry, 3: Differential pulse voltammetry 4: Square wave voltammetry, 5: Surface plasmon resonance.

Advertisement

4. Conclusion

Nanomaterials offer significant advantages, especially in sensor technology, due to their large surface area. When biocompatible nanomaterials are used as biorecognition layers, it enables the design of highly sensitive biosensors. Many nanomaterials, which are widely used today, are now being replaced by novel nanomaterials due to their physical stability, easy synthesis, easy fabrication, and cheapness.

Nanomaterials became important components in bioanalytical devices since they clearly increase the performances in the sense of detection limits and sensitivity down to single molecules detection.

Over content of this chapter aims to evaluate developments in the fields of new nanomaterial-based biosensors. Their production and potential applications for the direct and reliable detection of bioanalytes are described. In addition, research interests for the production of nanomaterial-based biosensors were encouraged with examples.

References

  1. 1. Wu, L., Xiong, E. H., Zhang, X., Zhang, X. H., & Chen, J. H. (2014). Nanomaterials as signal amplification elements in DNA-based electrochemical sensing. Nano Today, 9(2), 197-211
  2. 2. Michalet, X., Pinaud, F. F., Bentolila, L. A., Tsay, J. M., Doose, S., Li, J. J., Weiss, S. (2005). Quantum dots for live cells, in vivo imaging, and diagnostics. Science, 307(5709), 538-544
  3. 3. .Cheng, L., Wang, X. W., Gong, F., Liu, T., & Liu, Z. (2020). 2D nanomaterials for cancer theranostic applications. Advanced Materials, 32(13), 1902333
  4. 4. Tsou, Y. H., Zhang, X. Q ., Zhu, H., Syed, S., & Xu, X. Y. (2018). Drug delivery to the brain across the blood–brain barrier using nanomaterials. Small, 14(25)
  5. 5. Zhu, W., Chen, Z., Pan, Y., Dai, R. Y., Wu, Y., Zhuang, Z. B., Li, Y. D. (2019). Functionalization of hollow nanomaterials for catalytic applications: Nanoreactor construction. Advanced Materials, 31(38)
  6. 6. Xin, Q ., Shah, H., Nawaz, A., Xie, W. J., Akram, M. Z., Batool, A., Gong, J. R. (2019). Antibacterial carbon-based nanomaterials. Advanced Materials, 31(45), 1804838
  7. 7. Chen, Y., Zhou, S. W., Li, L. L., & Zhu, J. J. (2017). Nanomaterialsbased sensitive electrochemiluminescence biosensing. Nano Today, 12, 98-115
  8. 8. Arduini, F., Cinti, S., Mazzaracchio, V., Scognamiglio, V., Amine, A., Moscone, D. (2020). Carbon black as an outstanding and affordable nanomaterial for electrochemical (bio)sensor design. Biosensors and Bioelectronics, 156, 112033
  9. 9. Nikoobakht, B., El-Sayed, M. A. (2003). Preparation and growth mechanism of gold nanorods (NRs) using seed-mediated growth method. Chem. Mater., 15, 1957-1962
  10. 10. Kokulnathan, T., Wang, T. J., Kumar, E. A., Suvina, V., Balakrishna, R. G. (2020). Development of an Electrochemical Platform Based on Nanoplate like Zirconium Phosphate for the Detection of Furazolidone. ACS Appl. Nano Mater., 3, 4522-4529
  11. 11. Mirhosseini, M., Shekari-Far, A., Hakimian, F., Haghiralsadat, B. F., Fatemi, S. K., Dashtestani, F. (2020) Core-shell Au@Co-Fe hybrid nanoparticles as peroxidase mimetic nanozyme for antibacterial application, Process Biochemistry 95, 131-138
  12. 12. Fernandes, P. M. V., Campiña, J. M., Silva, A. F. (2020). A layered nanocomposite of laccase, chitosan, and Fe3O4 nanoparticles-reduced graphene oxide for the nanomolar electrochemical detection of bisphenol A. Microchimica Acta, 187, 262
  13. 13. Zhoua, L., et al. (2019). A label-free electrochemical biosensor for microRNAs detection based on DNA nanomaterial by coupling with Y-shaped DNA structure and non-linear hybridization chain reaction. Biosensors and Bioelectronics, 126, 657-663
  14. 14. Liu, M., Zhang, Q ., Brennan, J. D., Li, Y. (2018). Graphene-DNAzyme-based fluorescent biosensor for Escherichia coli Detection. MRS Communications, 8, 687-694
  15. 15. Zhu, S., Meng, Q ., Wang, L., Zhang, J., Song, Y., Jin, H., Zhang, K., Sun, H., Wang, H., Yang, B. (2013). Highly photoluminescent carbon dots for multicolor patterning, sensors, and bioimaging. Angew. Chem. Int. Ed. Engl., 52, 3953-3957
  16. 16. Guo, X. F., Kim, G. J. (2009). Synthesis of Ultrafine Carbon Black by Pyrolysis of Polymers Using a Direct Current Thermal Plasma Process. Plasma Chemistry and Plasma Processing, 30, 75-90
  17. 17. Fang, L., Ohfuji, H., Irifune, T. (2013). A Novel Technique for the Synthesis of Nanodiamond Powder, Journal of Nanomaterials, 2013, 201845
  18. 18. Cornell, R. M., Schwertmann, U. (2006). The iron oxides structures, properties, reactions, occurrences and uses. 2nd, Completely Revised and Extended Edition, p 139-146
  19. 19. Li, Y., Guo, C., Li, J., Liao, W., Li, Z., Zhang, J., et al. (2017). Pyrolysis-induced synthesis of iron and nitrogen-containing carbon nanolayers modified graphdiyne nanostructure as a promising core-shell electrocatalyst for oxygen reduction reaction. Carbon, 119, 201-210
  20. 20. Wang, K., Wang, N., He, J., Yang, Z., Shen, X., Huang, C. (2017). Preparation of 3D architecture graphdiyne nanosheets for high-performance sodium-ion batteries and capacitors. ACS Appl. Mater, Interfaces 9 (46), 40604-40613
  21. 21. Wu, L., Gao, J., Lu, X., et al. (2020). Graphdiyne: A new promising member of 2D all-carbon nanomaterial as robust electrochemical enzyme biosensor platform, Carbon, 156, 568-575
  22. 22. Li, X., Li, Y., Zhang, J., et al. (2019). Molybdenum disulfide/graphdiyne-based photoactive material derived photoelectrochemical strategy for highly sensitive MicroRNA assay. Sensors and Actuators B-Chemical, 297, 126808
  23. 23. Chang, F., Huang, L., Guo, C., et al. (2019). Graphdiyne-Based One-Step DNA Fluorescent Sensing Platform for the Detection of Mycobacterium tuberculosis and Its Drug-Resistant Genes. ACS Applied Materials & Interfaces, 11(39), 35622-35629
  24. 24. Sau, T.K., Rogach, A.L., J€ackel, F., Klar, T.A., Feldmann, J. (2010). Properties and applications of colloidal nonspherical noble metal nanoparticles. Adv. Mater, 22(16), 1805-1825
  25. 25. Doria, G., Conde, J., Veigas, B., Giestas, L., Almeida, C., Assunç~ao, M., Rosa, J., Baptista, P. V. (2012). Noble metal nanoparticles for biosensing applications. Sensors, 12(2),1657-1687
  26. 26. Emrani, A.S., Danesh, N.M., Lavaee, P., Ramezani, M., Abnous, K., Taghdisi, S.M. (2016). Colorimetric and fluorescence quenching aptasensors for detection of streptomycinin blood serum and milk based on double-stranded DNA and gold nanoparticles. Food Chem., 190, 115-121
  27. 27. Dondapati, S.K., Sau, T.K., Hrelescu, C., Klar, T.A., Stefani, F.D., Feldmann, J. (2010) Label free biosensing based on single gold nanostars as plasmonic transducers. ACS Nano, 4 (11), 6318-6322
  28. 28. Basu, M., Seggerson, S., Henshaw, J., Jiang, J., del A. Cordona, R., Lefave, C., Boyle, P.J., Miller, A., Pugia, M., Basu, S. (2004). Nano-biosensor development for bacterial detection during human kidney infection: use of glycoconjugate-specific antibody bound gold nano wire arrays (GNWA). Glycoconj. J. 21(8-9), 487-496
  29. 29. Karim, M.N., Lee, J.E., Lee, H.J. (2014). Amperometric detection of catechol using tyrosinase modified electrodes enhanced by the layer-by-layer assembly of gold nanocubes and polyelectrolytes. Biosens. Bioelectron, 61, 147-151
  30. 30. Parab, H.J., Jung, C., Lee, J.-H., Park, H.G. (2010). A gold nanorod-based optical DNA biosensor for the diagnosis of pathogens, Biosens. Bioelectron. 26 (2), 667-673
  31. 31. Kokulnathan, T., Chen, S. M. (2019). Rational Design for the Synthesis of Europium Vanadate-Encapsulated Graphene Oxide Nanocomposite: An Excellent and Efficient Catalyst for the Electrochemical Detection of Clioquinol. ACS Sustainable Chem. Eng., 7, 4136-4146
  32. 32. Li, X., Zhu, J., Wei, B. (2016). Hybrid nanostructures of metal/two dimensional nanomaterials for plasmon-enhanced applications. Chem. Soc. Rev., 45, 3145-318
  33. 33. He, P., Huang, Q ., Huang, B., Chen, T. (2017). Controllable synthesis of Ni−Co−Mn multi-component metal oxides with various morphologies for high-performance flexible supercapacitors. RSC Adv., 7, 24353-24358
  34. 34. Kokulnathan, T.; Chen, S. M. Praseodymium Vanadate Decorated Sulfur-doped Carbon Nitride Hybrid Nanocomposite: The Role of Synergistic Electrocatalyst for the Detection of Metronidazole. ACS Appl. Mater. Interfaces 2019, 11, 7893-7905
  35. 35. Lin, R.; Ding, Y. A review on the synthesis and applications of mesostructured transition metal phosphates. Materials 2013, 6, 217-243
  36. 36. Liu, J., Meyns, M., Zhang, T., Arbiol, J., Cabot, A., Shavel, A. (2018). Triphenyl phosphite as the phosphorus source for the scalable and cost-effective production of transition metal phosphides. Chem. Mater., 30, 1799-1807
  37. 37. Zhang, Y., Xiao, J., Lv, Q ., Wang, S. (2018). Self-supported transition metal phosphide based electrodes as high-efficient water splitting cathodes. Front. Chem. Sci. Eng., 12, 494-508
  38. 38. Tomanin, P. P., Cherepanov, P. V., Besford, Q . A., Christofferson, A. J., Amodio, A., McConville, C. F., Yarovsky, I., Caruso, F., Cavalieri, F. (2018). Cobalt Phosphate Nanostructures for Non- Enzymatic Glucose Sensing at Physiological pH. ACS Appl. Mater. Interfaces, 10, 42786-42795
  39. 39. Pessoa, C. A., Gushikem, Y., Kubota, L. T., Gorton, L. (1997). Preliminary electrochemical study of phenothiazines and phenoxazines immobilized on zirconium phosphate. J. Electroanal. Chem., 431, 23-27
  40. 40. Singh, S. (2019). Nanomaterials exhibiting enzyme-like properties (nanozymes): current advances and future perspectives. Front Chem., 7, 46
  41. 41. Jiang, B., Fang, L., Wu, K., Yan, X., Fan, K. (2020). Ferritins as natural and artificial nanozymes for theranostics. Theranostics, 10, 687-706
  42. 42. Niu, X., Cheng, N., Ruan, X., Du, D., Lin, Y. (2020). Review— Nanozyme-based immunosensors and immunoassays: recent developments and future trends. J Electrochem Soc., 167, 037508
  43. 43. Liu, J. (2019). Special topic: nanozyme-based analysis and testing. J Anal Test, 3, 189-190
  44. 44. Liu, B., Liu, J. (2017). Surface modification of nanozymes. Nano Res., 10, 1125-1148
  45. 45. Chatterjee, B., Das, S. J., Anand, A., Sharma, T. K. (2020) Nanozymes and aptamer-based biosensing. Mater Sci Technol., 3, 127-135
  46. 46. Sun, D., Line, X., Lu, J., Wei, P., Luo, Z., Lu, X., et al. (2019). DNA nanotetrahedron-assisted electrochemical aptasensor for cardiac troponin I detection based on the co-catalysis of hybrid nanozyme, natural enzyme and artificial DNAzyme. Biosens Bioelectron, 142, 111578
  47. 47. Bazin, I., Tria, S. A., Hayat, A., Marty, J. L. (2017). New biorecognition molecules in biosensors for the detection of toxins. Biosens Bioelectron, 87, 285-298
  48. 48. Zhang, X., Wu, D., Zhou, X., Yu, Y., Liu, J., Hu, N., et al. (2019). Recent progress on the construction of nanozymes-based biosensors and their applications to food safety assay. TrAC Trends Anal Chem., 121, 115668
  49. 49. Shu, J., Qiu, Z., Wei, Q ., Zhuang, J., Tang, D. (2015). Cobalt-porphyrinplatinum- functionalized reduced graphene oxide hybrid nanostructures: a novel peroxidase mimetic system for improved electrochemical immunoassay. Sci Rep., 5, 15113
  50. 50. Wei, Y., Li, Y., Li, N., Zhang, Y., Yan, T., Man, H., Wei, Q . (2016). Sandwich-type electrochemical immunosensor for the detection of AFP based on Pd octahedral and APTES-M-CeO2-GS as signal labels. Biosens Bioelectron, 79, 482-487
  51. 51. Zhu, F., Zhao, G., Dou, W. (2018). Electrochemical sandwich immunoassay for Escherichia coli O157:H7 based on the use of magnetic nanoparticles and graphene functionalized with electrocatalytically active Au@Ptcore/shell nanoparticles. Microchim Acta, 185, 455
  52. 52. Golchin, J., Golchin, K., Alidadian, N., Ghaderi, S., Eslamkhah, S., Eslamkhah, M., Akbarzadeh, A. (2017). Nanozyme applications in biology and medicine: an overview. Artif Cells Nanomed Biotechnol., 45, 1-8
  53. 53. Serafín, V., Valverd,e A., Garranzo-Asensio, M., Barderas, R., Campuzano, S., Yáñez-Sedeño, P., Pingarrón, J. M. (2019). Simultaneous amperometric immunosensing of the metastasis related biomarkers IL-13Rα2 and CDH-17 by using grafted screen-printed electrodes and a composite prepared from quantum dots and carbon nanotubes for signal amplification. Microchim Acta, 186, 411
  54. 54. Serafín, V., Valverde, A., Martínez-García, G., Martínez-Periñán, E., Comba, F., Garranzo-Asensio, M., Barderas, R., Yáñez-Sedeño, P., Campuzano, S., Pingarrón, J. M. (2019). Graphene quantum dots functionalized multi-walled carbon nanotubes as nanocarriers in electrochemical immunosensing. Determination of IL-13 receptor α2 in colorectal cells and tumor tissues with different metastatic potential. Sensors Actuators B Chem., 284, 711-722
  55. 55. Zhang, L., Xie, X., Yuan, Y., Chai, Y., Yuan, R. (2019). FeS2-AuNPs nanocomposite as mimicking enzyme for constructing signal-off sandwich-type electrochemical immunosensor based on electroactive nickel hexacyanoferrate as matrix. Electroanalysis, 31, 1019-1025
  56. 56. Sun, D., Line, X., Lu, J., Wei, P., Luo, Z., Lu, X., et al. (2019). DNA nanotetrahedron-assisted electrochemical aptasensor for cardiac troponin I detection based on the co-catalysis of hybrid nanozyme, natural enzyme and artificial DNAzyme. Biosens Bioelectron, 142, 111578
  57. 57. Ding, H., Zhang, Y., Wang, S., Xu, J., Xu, S., Li, G. (2012). Fe3O4@ SiO2 core/shell nanoparticles: the silica coating regulations with a single core for different core sizes and shell thicknesses. Chem mater., 24: 4572-4580
  58. 58. Cheng, G., Zhang, J. L., Liu, Y. L., Sun, D. H., Ni, J. Z. (2011). Synthesis of novel Fe3O4@ SiO2@ CeO2 microspheres with mesoporous shell for phosphopeptide capturing and labeling. Chem commun., 47, 5732-5734
  59. 59. Shao, M., Ning, F., Zhao, J., Wei, M., Evans, D. G, Duan, X. (2012). Preparation of Fe3O4@ SiO2@ layered double hydroxide core–shell microspheres for magnetic separation of proteins. J am Chem Soc., 134, 1071-1077
  60. 60. Wang, L., Cole, M., Li, J., Zheng, Y., Chen, Y. P., Miller, K. P., Decho, A. W., Benicewicz, B. C. (2015). Polymer grafted recyclable magnetic nanoparticles, Polym Chem., 6, 248-255
  61. 61. Peng, X., Wang, Y., Tang, X., Liu, W. (2011). Functionalized magnetic core–shell Fe3O4@ SiO2 nanoparticles as selectivity enhanced chemosensor for Hg (II). Dyes pigments, 91, 26-32
  62. 62. Wang, Y., Peng, X., Sh,i J., Tang, X., Jiang, J., Liu, W. (2012). Highly selective fluorescent chemosensor for Zn2+ derived from inorganic organic hybrid magnetic core/shell Fe3O4@ SiO2 nanoparticles. Nanoscale Res. Lett., 7, 86
  63. 63. Zhao, Y., Li, J., Zhao, L., Zhang, S., Huang, Y., W,u X., Wang, X. (2014). Synthesis of amidoxime-functionalized Fe3O4@ SiO2 core–shell magnetic microspheres for highly efficient sorption of U (VI). Chem Eng., 235, 275-283
  64. 64. Miao, S. S., Wu, M. S., Ma, L. Y., He, H. J., Yang, H. (2016). Electrochemiluminescence biosensor for determination of organophosphorous pesticides based on bimetallic Pt- Au/multi-walled carbon nanotubes modified electrode. Talanta, 158, 142-151
  65. 65. Gao, W., Wang, C., Muzyka, Kitte, S. A., Li, J., Zhang, W., Xu, G. (2017). Artemisinin-luminol chemiluminescence for forensic bloodstain detection using a smart phone as a detector. Anal Chem., 89, 6160-6165
  66. 66. Stewart, A. J., Hendry, J., Dennany, L. (2015). Whole blood electrochemiluminescent detection of dopamine. Anal Chem, 87, 11847-11853
  67. 67. Takahashi, F., Nitta, S., Shimizu, R., Jin, J. (2018). Electrochemiluminescence and voltammetry of tris (2, 2-bipyridine) ruthenium (II) with amphetaminetype stimulants as coreactants: An application to the discrimination of methamphetamine. Forensic Toxicol., 36, 185-191
  68. 68. Zhang, J. J., Kang, T.F., Hao, Y. C., Lu, L. P., Cheng, S.Y. (2015). Electrochemiluminescent immunosensor based on CdS quantum dots for ultrasensitive detection of microcystin- LR, Sens. Actuators B Chem., 214, 117-123
  69. 69. Li, G., Yu, X., Liu, D., Liu, X., Li, F., Cui, H. (2015). Label-free electrochemiluminescence aptasensor for 2, 4, 6-trinitrotoluene based on bilayer structure of luminescence functionalized graphene hybrids. Anal Chem., 87, 10976-10981
  70. 70. Kitte, S. A., Gao, W., Zholudov, Y. T., Qi, L., Nsabimana, A., Liu, Z., Xu, G. (2017). Stainless steel electrode for sensitive luminol electrochemiluminescent detection of H2O2, glucose, and glucose oxidase activity. Anal Chem., 89, 9864-9869
  71. 71. Bagheri, E., Ansari, L., Abnous, K., Taghdisi, S. M., Naserifar, M., Ramezani, M., Alibolandi, M. (2020). Silica -magnetic inorganic hybrid nanomaterials as versatile sensing platform. Nanomed. J., 7(3), 183-193
  72. 72. Xu, T., Song, Y., Gao, W., Wu, T., Xu, L.P., Zhang, X., Wang, S. (2018). Superwettable Electrochemical Biosensor toward Detection of Cancer Biomarkers. ACS Sens., 3, 72-78
  73. 73. Brown 3rd, C. W., Buckhout-White, S., Díaz, S. A., Melinger, J. S., Ancona, M. G., Goldman, E. R., Medintz, I. L. (2017). Evaluating Dye-Labeled DNA Dendrimers for Potential Applications in Molecular Biosensing. ACS Sens., 2, 401-410
  74. 74. Zhou, L., Morel, M., Rudiuk, S., Baigl, D. (2017). Intramolecularly Protein-Crosslinked DNA Gels: new biohybrid nanomaterials with controllable size and catalytic activity. Small, 13
  75. 75. Zhou, X., Zhao, M., Duan, X., Guo, B., Cheng, W., Ding, S., Ju, H. (2017). Collapse of DNA tetrahedron nanostructure for “Off-On” fluorescence detection of DNA methyltransferase activity. ACS Appl. Mater. Interfaces, 9, 40087-40093
  76. 76. Li, Y., Chang, Y., Yuan, R., Chai, Y. (2018). Highly Efficient Target Recycling-Based Netlike Y-DNA for Regulation of Electrocatalysis toward Methylene Blue for Sensitive DNA Detection. ACS Appl. Mater. Interfaces, 10, 25213-25218
  77. 77. Wang, K., Lei, Y., Zhong, G.X., Zheng, Y.J., Sun, Z.L., Peng, H.P., Chen, W., Liu, A.L., Chen, Y.Z., Lin, X.H. (2015). Dual-probe electrochemical DNA biosensor based on the “Y” junction structure and restriction endonuclease assisted cyclic enzymatic amplification for detection of double-strand DNA of PML/RARα related fusion gene. Biosens. Bioelectron. 71, 463-469
  78. 78. Peng, X., Zhu, J., Wen, W., Bao, T., Zhang, X., He, H., Wang, S. (2018). Integrated amplified aptasensor with in-situ precise preparation of copper nanoclusters for ultrasensitive electrochemical detection of microRNA 21. Biosens. Bioelectron. 118, 174-180
  79. 79. Miao, P., Jiang, Y., Zhang, T., Huang, Y., Tang, Y. (2018). Electrochemical sensing of attomolar miRNA combining cascade strand displacement polymerization and reductant-mediated amplification. Chem. Commun., 54, 7366-7369
  80. 80. Liu, C., Chen, C., Li, S., Dong, H., Dai, W., Xu, T., Liu, Y., Yang, F., Zhang, X. (2018). Target-Triggered Catalytic Hairpin Assembly-Induced Core-Satellite Nanostructures for High-Sensitive "Off-to-On" SERS Detection of Intracellular MicroRNA. Anal. Chem., 90, 10591-10599
  81. 81. Peng, X., Liang, W.B., Wen, Z.B., Xiong, C.Y., Zheng, Y.N., Chai, Y.Q ., Yuan, R. (2018). Ultrasensitive Fluorescent Assay Based on a Rolling-Circle-Amplification-Assisted Multisite-Strand-Displacement-Reaction Signal-Amplification Strategy. Anal. Chem., 90, 7474-7479
  82. 82. Bi, S., Yue, S., Zhang, S. (2017). Hybridization chain reaction: a versatile molecular tool for biosensing, bioimaging, and biomedicine. Chem. Soc. Rev., 46, 4281-4298
  83. 83. Chang, C .C., Chen, C. Y., Chuang, T. L., Wu, T.H., Wei, S. C., Liao, H., Lin, C. W. (2016). Aptamer-based colorimetric detection of proteins using a branched DNA cascade amplification strategy and unmodified gold nanoparticles. Biosens. Bioelectron. 78, 200-205
  84. 84. Ding, X., Cheng, W., Li, Y., Wu, J., Li, X., Cheng, Q ., Ding, S. (2017). An enzyme-free surface plasmon resonance biosensing strategy for detection of DNA and small molecule based on nonlinear hybridization chain reaction. Biosens. Bioelectron. 87, 345-351
  85. 85. Xu, F.X., Su, Y.L., Zhang, H., Kong, J.Y., Yu, H., Qian, B.Y. (2014). Prognostic implications for high expression of MiR-25 in lung adenocarcinomas of female non-smokers. Asian Pac. J. Cancer Prev., 15, 1197-1203
  86. 86. Wahlquist, C., Jeong, D., Rojas-Muñoz, A., Kho, C., Lee, A., Mitsuyama, S., van Mil, A., Park, W.J., Sluijter, J.P., Doevendans, P.A., Hajjar, R.J., Mercola, M. (2014). Inhibition of miR-25 improves cardiac contractility in the failing heart. Nature, 508, 531-535
  87. 87. Breaker, R. R., Joyce, G. F. (1994). A DNA enzyme that cleaves RNA. Chem. Biol., 1, 223-229
  88. 88. Ihms, H. E., Lu, Y. (2012). In vitro selection of metal ion-selective DNAzymes. In Ribozymes: Methods and Protocols. Edited by Hartig JS. Totowa, NJ: Humana Press, 297-316
  89. 89. Wang, H., Liu, Y., Wang, J., Xiong, B., Hou, X. (2020). Electrochemical impedance biosensor array based on DNAzyme-functionalized single-walled carbon nanotubes using Gaussian process regression for Cu(II) and Hg(II) determination. Microchimica Acta, 187, 207
  90. 90. Oh, W. K., Kwon, O. S., Jang, J. (2013). Conducting polymer nanomaterials for biomedical applications: cellular interfacing and biosensing. Polym Rev., 53(3), 407-442
  91. 91. Syshchyk, O., Skryshevsky, V. A., Soldatkin, O. O., Soldatkin, A. P. (2015). Enzyme biosensor systems based on porous silicon photoluminescence for detection of glucose, urea and heavy metals. Biosens Bioelectron, 66, 89-94
  92. 92. Ouyang, H., Shu, Q ., Wang, W., Wang, Z., Yang, S., Wang, L., Fu, Z. (2016). An ultra-facile and label-free immunoassay strategy for detection of copper (II) utilizing chemiluminescence self-enhancement of Cu (II)-ethylenediaminetetraacetate chelate. Biosens Bioelectron, 85, 157-163
  93. 93. Seeman, N. C., Sleiman, H. F. (2017). DNA nanotechnology. Nat Rev Mater, 3(1), 1-23
  94. 94. Wang, H., Liu, Y., Liu, G. (2018). Reusable resistive aptasensor for Pb(II) based on the Pb(II)-induced despiralization of a dna duplex and formation of a G-quadruplex. Microchim Acta, 185(2), 142
  95. 95. Liu, L., Wu, H. C. (2016). Dna-based nanopore sensing. Angew Chem Int Ed., 55(49), 15216-15222
  96. 96. Liang, G., Man, Y., Li, A., Jin, X., Liu, X., Pan, L. (2017). Dnazyme-based biosensor for detection of lead ion: a review. MicrochemJ, 131, 145-153
  97. 97. Liu, S., Cheng, C., Gong, H., Wang, L. (2015). Programmable Mg2+ dependent DNAzyme switch by the catalytic hairpin DNA assembly for dual-signal amplification toward homogeneous analysis of protein and DNA. Chem Commun., 51(34), 7364-7367
  98. 98. Wang, H., Liu, Y., Liu, G. (2018). Label-free biosensor using a silver specific RNA-cleaving DNAzyme functionalized single-walled carbon nanotube for silver ion determination. Nanomaterials, 8, 258
  99. 99. Wang, H., Yin, Y., Gang, L. (2019). Single-gap microelectrode functionalized with single-walled carbon nanotubes and Pbzyme for the determination of Pb2+. Electroanal., 31(6), 1174-1181
  100. 100. Shen, W., Li, Y., Qi, T., Wang, S., Sun, J., Deng, H., Tang, S. (2018). Fluorometric determination of zinc (II) by using DNAzymemodified magnetic microbeads. Microchim Acta, 185(10), 447
  101. 101. Wang, H., Liu, Y., Liu, G. (2018). Electrochemical biosensor using DNA embedded phosphorothioate modified RNA for mercury ion determination. ACS Sens, 3(3), 624-631
  102. 102. Gupta, V. K., Singh, A. K., Kumawat, L. K., Mergu, N. (2016). An easily accessible switch-on optical chemosensor for the detection of noxious metal ions Ni (II), Zn (II), Fe (III) and UO2 (II). Sensor Actuat BChem., 222, 468-482
  103. 103. Wang, X., Feng, Y., Dong, P., Huang, J. (2019). A Mini Review on Carbon Quantum Dots: Preparation, Properties, and Electrocatalytic Application. Front. Chem., 7, 671
  104. 104. Peng, Z., Han, X., Li, S., Al-Youbic, A., O Bashammakh, A. S., SEl-Shahawi M. Leblan, R. M. (2017). Carbon dots: Biomacromolecule interaction, bioimaging and nanomedicine. Coord. Chem. Rev., 343, 256-277
  105. 105. Wang, T., Wang, A., Wang, R., Liu, Z., Sun, Y., Shan, G., Chen, Y., Liu, Y. (2019). Carbon dots with molecular fluorescence and their application as a “turn-o” fluorescent probe for ferricyanide detection. Sci. Rep., 9, 10723
  106. 106. Baker, S. N., Baker, G. A. (2010). Luminescent Carbon Nanodots: Emergent Nanolights. Angew. Chem. Int. Ed., 49, 6726-6744
  107. 107. Margraf, J. T., Lodermeyer, F., Strauss, V., Haines, P., Walter, J., Peukert, W., Costa, R. D., Clark, T., Guldi, D. M. (2016). Using Carbon Nanodots as Inexpensive and Environmentally Friendly Sensitizers in Mesoscopic Solar Cells. Nanoscale Horiz., 1, 220-226
  108. 108. Hutton, G. A. M., Martindale, B. C. M., Reisner, E. (2017). Carbon Dots as Photosensitisers for Solar-Driven Catalysis. Chem. Soc. Rev., 46, 6111-6123
  109. 109. Martinez-Perinan, E., Bravo, I., Rowley-Neale, S. J., Lorenzo, E., Banks, C. E. (2018). Carbon Nanodots as Electrocatalysts Towards the Oxygen Reduction Reaction. Electroanalysis, 30, 436-444
  110. 110. Athinarayanan, J., Periasamy, V. S., Alshatwi, A. A. (2020) Simultaneous fabrication of carbon nanodots and hydroxyapatite nanoparticles from fish scale for biomedical applications, Materials Science and Engineering C 117 111313
  111. 111. Li, H., Chen, L., Wu, H., He, H., Jin, Y. (2014). Ionic Liquid-Functionalized Fluorescent Carbon Nanodots and their Applications in Electrocatalysis, Biosensing, and Cell Imaging. Langmuir, 30, 15016-15021
  112. 112. Garcia-Mendiola, T., Bravo, I., Maria Lopez-Moreno, J., Pariente, F., Wannemacher, R., Weber, K., Popp, J., Lorenzo, E. (2018). Carbon Nanodots Based Biosensors for Gene Mutation Detection. Sens. Actuators B Chem., 256, 226-233
  113. 113. Arduini, F., Zanardi, C., Cinti, S., Terzi, F., Moscone, D., Palleschi, G., Seeber, R. (2015). Effective electrochemical sensor based on screen-printed electrodes modified with a carbon black-Au nanoparticles composite. Sens. Actuat. B, 212, 536-543
  114. 114. Vicentini, F.C., Ravanini, A.E., Figueiredo-Filho, L.C., Iniesta, J., Banks, C.E., Fatibello- Filho, O. (2015). Imparting improvements in electrochemical sensors: evaluation of different carbon blacks that give rise to significant improvement in the performance of electroanalytical sensing platforms. Electrochim. Acta, 157, 125-133
  115. 115. Cinti, S., Arduini, F., Carbone, M., Sansone, L., Cacciotti, I., Moscone, D., Palleschi, G. (2015). Screen-Printed Electrodes Modified with Carbon Nanomaterials: A Comparison among Carbon Black, Carbon Nanotubes and Graphene. Electroanal, 27, 2230-2238
  116. 116. Mazzaracchio, V., Tomei, M.R., Cacciotti, I., Chiodoni, A., Novara, C., Castellino, M.,Scordo, G., Amine, A., Moscone, D., Arduini, F. (2019). Inside the different types of carbon black as nanomodifiers for screen-printed electrodes. Electrochim. Acta, 317, 673-683
  117. 117. Alcantara, R., Jimenez-Mateos, L. M., Lavela, P., Tirado, J. L. (2001). Carbon black: a promising electrode material for sodium-ion batteries. Electrochem. Comm., 3, 639-642
  118. 118. Drofenik, J., Gaberscek, M., Dominko, R., Poulsen, F. W., Mogensen, M. B., Pejovnik, S., Jamnik, J. (2003). Cellulose as a binding material in graphitic anodes for Li ion batteries: A performance and degradation study. Electrochim. Acta, 48, 883-889
  119. 119. Mazzaracchio, V., Tomei, M. R., Cacciotti, I., Chiodoni, A., Novara, C., Castellino, M., Scordo, G., Amine, A., Moscone, D., Arduini, F. (2019). Inside the different types of carbon black as nanomodifiers for screen-printed electrodes. Electrochim. Acta, 317, 673-683
  120. 120. Talarico, D., Arduini, F., Amine, A., Cacciotti, I., Moscone, D., Palleschi, G. (2016). Screen-printed electrode modified with carbon black and chitosan: a novel platform for acetylcholinesterase biosensor development. Anal. Bioanal. Chem., 408, 7299-7309
  121. 121. Silva, T. A., Fatibello-Filho, O. (2017). Square-wave adsorptive anodic stripping voltammetric determination of ramipril using an electrochemical sensor based on nanostructured carbon black. Anal. Methods, 9, 4680-4687
  122. 122. Arduini, F., Cinti, S., Mazzaracchio, V., Scognamiglio, V., Amine, A., Moscone, D. (2020). Carbon black as an outstanding and affordable nanomaterial for electrochemical (bio)sensor design. Biosensors and Bioelectronics, 156, 112033
  123. 123. Aydın, E. B., Aydın, M., Sezgintürk, M. K. (2018). Electrochemical immunosensor based on chitosan/conductive carbon black composite modified disposable ITO electrode: An analytical platform for p53 detection. Biosensors and Bioelectronics,121, 80
  124. 124. Aydın, M,, Aydın, E. B., Sezgintürk, M. K. (2018). A highly selective electrochemical immunosensor based on conductive carbon black and star PGMA polymer composite material for IL-8 biomarker detection in human serum and saliva, Biosensors and Bioelectronics, 117, 720
  125. 125. Messaoud, N.B., Lahcen, A.A., Dridi, C., Amine, A. (2018). Ultrasound assisted magnetic imprinted polymer combined sensor based on carbon black and gold nanoparticles for selective and sensitive electrochemical detection of Bisphenol A. Sensors and Actuators B Chemical 276, 304
  126. 126. Arduini, F., Cinti, s., Mazzaracchio, V., Scognamiglio, V., Amine, A., Moscone, D. (2020). Carbon black as an outstanding and affordable nanomaterial for electrochemical (bio)sensor design. Biosensors and Bioelectronics, 156, 112033. Doi: 10.1016/j.bios.2020.112033
  127. 127. Zhang, X. Q ., Lam, R., Xu, X., Chow, E. K., Kim, H. J., Ho, D. (2011). Multimodal Nanodiamond Drug Delivery Carriers for Selective Targeting, Imaging, and Enhanced Chemotherapeutic Efficacy. Advanced Materials, 23, 4770-4775
  128. 128. Babadia, F. E., Hosseinia, S., Shavandib, A., Moghaddasc, H., Shotipruka, A., Kheawhoma, S. (2019). Electrochemical investigation of amino acids Parkia seeds using the composite electrode based on copper/carbon nanotube/nanodiamond. Journal of Environmental Chemical Engineering, 7, 102979
  129. 129. Azevedo, A. F., Matsushima, J. T., Vicentin, F. C., Baldan, M R., Ferreira, N. G. (2009). Surface characterization of NCD films as a function of sp2/sp3 carbon and oxygen content, Applied Surface Science 255, 6565-6570
  130. 130. Azevedo, A. F., Baldan, M. R., and Ferreira, N. G. (2012). Nanodiamond Films for Applications in Electrochemical Systems, International Journal of Electrochemistry, 508453, Doi: 10.1155/2012/508453
  131. 131. Hasanzadeh, M., Shadjou, N., And de la Guardia, M. (2015). Iron and iron-oxide magnetic nanoparticles as signal-amplification elements in electrochemical biosensing. Trends Anal Chem., 72, 1-9
  132. 132. Sieben, S., Bergemann, C., Lubbe, A., Brockmann, B., Rescheleit, D. (2001). Comparison of different particles and methods for magnetic isolation of circulating tumor cells. J Magn Mater., 225, 175
  133. 133. Pinho, M. S., Gregori, M. L., Nunes, R. C. R, Soares, B. G. (2001). Aging effect on the reflectivity measurements of polychloroprene matrices containing carbon black and carbonyl-iron powder. Polym Degrad Stab., 73,1-5
  134. 134. Abdelhamid, H. N. (2019). Nanoparticle based surface assisted laser desorption ionisation mass spectrometry: a review. Microchim Acta, 186, 682
  135. 135. Kouassi, G. K., Irudayaraj, J., McCarty, G. (2005). Examination of cholesterol oxidase attachment to magnetic nanoparticles. J Nanobiotechnol, 3, 1
  136. 136. Lu, B. W., Chen, W. C. (2006). A disposable glucose biosensor based on drop- coating of screen-printed carbon electrodes with magnetic nanoparticles. J Magn Magn Mater, 304, 400-402
  137. 137. Wang, S. F., Xie, F., Hu, R. F. (2007). Carbon-coated nickel magnetic nanoparticles modified electrodes as a sensor for determinationof acetaminophen. Sens Act B, 123, 495-500
  138. 138. Lou, L., Yu, K., Zhang, Z., Huang, R., Zhu, J., Wang, Y., Zhu, Z. (2012). Dual-mode protein detection based on Fe3O4– Au hybrid nanoparticles. Nano Res., 5, 272-282
  139. 139. Dhouha, J., Eleonora, M., Danila, M., et al. (2020). Highly sensitive paper-based electrochemical sensor for reagent free detection of bisphenol A. Talanta, 216, 120924
  140. 140. Wenqing, Z., Huan, X., Xianxian, Z. (2020). 3D DNA nanonet structure coupled with target -catalyzed hairpin assembly for dual -signal synergistically amplified electrochemical sensing of circulating microRNA, Analytica Chimica Acta, 1122, 39-47
  141. 141. Mat Zaid, M. H., Abdullah, J., Rozi, N., Rozlan, A. A. M., Hanifah, S. A. (2020). A Sensitive Impedimetric Aptasensor Based on Carbon Nanodots Modified Electrode for Detection of 17ß-Estradiol. Nanomaterials , 10, 1346
  142. 142. Attaallah, R., Antonacci, A., Mazzaracchio, V., et al. (2020). Carbon black nanoparticles to sense algae oxygen evolution for herbicides detection: Atrazine as a case study. Biosensors & Bioelectronıcs, 159, 112203
  143. 143. Avelino, K. Y. P. S., Oliveira, L. S., Lucena-Silva, N., et al. (2020). Metal-polymer hybrid nanomaterial for impedimetric detection of human papillomavirus in cervical specimens. Journal of Pharmaceutical and Biomedical Analysis, 185, 113249
  144. 144. Lu, J., Hu, Y., Wang, P., et al. (2020). Electrochemical biosensor based on gold nanoflowers-encapsulated magnetic metal-organic framework nanozymes for drug evaluation with in-situ monitoring of H2O2 released from H9C2 cardiac cells. Sensors and Actuators B-Chemıcal, 311, 127909
  145. 145. Fang, B., Xu, S., Huang, Y., et al. (2020). Gold nanorods etching-based plasmonic immunoassay for qualitative and quantitative detection of aflatoxin M1 in milk. Food Chemistry, 329, 127160
  146. 146. Kumar, V., Kaur, I., Arora, S., et al. (2020). Graphene nanoplatelet/graphitized nanodiamond-based nanocomposite for mediator-free electrochemical sensing of urea. Food Chemistry, 303, 125375
  147. 147. Guo, W., Pi, F., Zhang, H., Sun, J., Zhang, Y., Sun, X. (2017). A Novel Molecularly Imprinted Electrochemical Sensor Modified with Carbon Dots, Chitosan, Gold Nanoparticles for the Determination of Patulin. Biosens. Bioelectron, 98, 299-304

Written By

Kübra Gençdağ Şensoy and Mihrican Muti

Submitted: 07 June 2020 Reviewed: 06 November 2020 Published: 07 December 2020