Next Article in Journal
The Synthesis and Biological Evaluation of Aloe-Emodin-Coumarin Hybrids as Potential Antitumor Agents
Next Article in Special Issue
Comparison of Different Volatile Extraction Methods for the Identification of Fishy Off-Odor in Fish By-Products
Previous Article in Journal
Mycobacterium Time-Series Genome Analysis Identifies AAC2′ as a Potential Drug Target with Naloxone Showing Potential Bait Drug Synergism
Previous Article in Special Issue
In Vivo Detection of Tetrodotoxin in Takifugu obscurus Based on Solid-Phase Microextraction Coupled with Ultrahigh-Performance Liquid Chromatography–Tandem Mass Spectrometry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Progress on Techniques in the Detection of Aflatoxin B1 in Edible Oil: A Mini Review

1
School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Road, Binhu District, Wuxi 214122, China
2
School of Life Sciences, Guangzhou University, Guangzhou 510006, China
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(19), 6141; https://doi.org/10.3390/molecules27196141
Submission received: 10 August 2022 / Revised: 12 September 2022 / Accepted: 15 September 2022 / Published: 20 September 2022

Abstract

:
Contamination of agricultural products and foods by aflatoxin B1 (AFB1) is becoming a serious global problem, and the presence of AFB1 in edible oil is frequent and has become inevitable, especially in underdeveloped countries and regions. As AFB1 results from a possible degradation of aflatoxins and the interaction of the resulting toxic compound with food components, it could cause chronic disease or severe cancers, increasing morbidity and mortality. Therefore, rapid and reliable detection methods are essential for checking AFB1 occurrence in foodstuffs to ensure food safety. Recently, new biosensor technologies have become a research hotspot due to their characteristics of speed and accuracy. This review describes various technologies such as chromatographic and spectroscopic techniques, ELISA techniques, and biosensing techniques, along with their advantages and weaknesses, for AFB1 control in edible oil and provides new insight into AFB1 detection for future work. Although compared with other technologies, biosensor technology involves the cross integration of multiple technologies, such as spectral technology and new nano materials, and has great potential, some challenges regarding their stability, cost, etc., need further studies.

1. Introduction

Food security has always been an issue of concern in the international community, and, in recent years, food contamination has become a major factor affecting food security. Contaminated food can not only adversely influence human health (poisoning events, chronic diseases, etc.) but also affect and slow down the economy. When people consume contaminated food, they need to spend a lot of money and time on treatment. There are many factors causing food contamination, such as biological, chemical, and physical factors. Among these, microbial contamination is common and mainly includes contamination by bacteria, fungi, molds, viruses, or their toxins and by-products [1,2]. Mycotoxins are common food contaminants, which can cause changes in the appearance, flavor, smell, and other characteristics of food [3,4,5,6,7]. Mycotoxins are secondary metabolites produced by fungi (e.g., Fusarium, Aspergillus, and Penicillium) that have multiple toxic effects on organisms and contaminate agricultural products (cereals, milk, etc.). More than 400 kinds of mycotoxins have been identified. Among them, aflatoxins (AFs) have become one of the major concerns due to their high toxicity and carcinogenicity, causing approximately 25% of animal deaths [8,9,10,11,12].
Edible vegetable oil plays an irreplaceable role in the human diet. The world oil crop output has increased year by year and had reached 635.5 million tons by 2021 [13]. From the growth of oil crops to the final product, i.e., oil, each link may be affected by external factors (such as mycotoxins), which may affect the quality and safety of edible vegetable oil [14]. This is because most oil crops, such as corn, peanut, soybean, rapeseed, sunflower seeds, olives, and nuts, are seasonal. During the growth process, they will be affected by climate, pests, and other factors and can be easily be infected by Aspergillus flavus. After harvest, the oil may deteriorate or be affected by mildew due to storage conditions (such as temperature and humidity, etc.) and storage methods [15]. At the same time, during the production of edible oil, fresh-pressed edible oil is vulnerable to contamination of raw materials infected with Aspergillus by aflatoxin B1 (AFB1) [16,17,18,19,20,21,22]. Therefore, contamination of edible vegetable oil products by AFB1 is a serious food safety problem (Figure 1) [20,23,24,25].
The presence of aflatoxin is usually detected by using precision instruments, such as high-performance liquid chromatography–mass spectrometry (HPLC–MS), high-performance liquid chromatography–fluorescence detection (HPLC–FD), or other molecular techniques, while rapid detection is mainly realized by enzymatic immunoassay ELISA [26,27]. Although different methods are available for the detection of AFB1 toxicity, these methods require expensive equipment and complex sample pretreatment or can only be performed at relatively high concentrations [28]. Therefore, simple, sensitive, efficient, economical, rapid, and stable AFB1 detection methods are required. Recently, new technologies, such as biosensors, have been applied in many fields, such as health care and food detection. Because of their key advantages, such as convenient operation, rapid response, and excellent portability, these technologies can detect harmful substances in food sensitively and accurately, hel** effectively avoid their harmful effects. They have attracted increasing attention of researchers and also promoted the rapid development of biosensors. With progress in nanotechnology, scientists are paying special attention to biosensors based on nanomaterials. These new biosensors or detection systems are sensitive, rapid, consistent, and cost-effective and can be used to detect AFB1 in food [29,30,31,32,33].
Regarding the increased importance of biosensors for accurate detection of AFB1 in edible oil, we have summarized the recent advances in biosensors for AFB1 analysis, specifically from the points of view of the development of novel bioinspired recognition elements and nanomaterials-based electrochemical biosensors.
Therefore, we searched PubMed and web of science for publications describing the detection technology of aflatoxin B1 in edible oil. Search terms were as follows: aflatoxin B1 OR AFB1 OR Aspergillus OR mycotoxins OR AFB2 OR AFG1 OR AFG2 OR AFM1 OR AFM2 OR AFs OR AFBO OR CYP450 OR edible oil OR vegetable oil OR corn oil OR peanut oil OR soybean oil OR sesame oil OR rapeseed oil OR sunflower seeds oil OR olives oil OR nuts oil OR maize oil OR canola oil OR blend oil OR coconut oil OR almond oil OR rice oil OR palm oil OR tea oil OR chromatographic technology OR spectroscopic technology OR immunological technology OR biosensor technology OR QuEChERS OR Fluorescence spectrophotometry OR Infrared spectroscopy OR Terahertz spectroscopy OR surface-enhanced raman spectroscopy (SERS) OR enzyme-linked immunosorbent assay (ELISA) OR amperometric OR impedometric OR electrochemical impedance spectroscopy (EIS) OR voltammetry (potentiometric) OR Conductometric OR LOD OR chromogenic OR Luminogenic OR Chemiluminescence OR Gravimetric OR Piezoelectric OR Magnetoelastic OR Acoustic OR electrodes (SPEs)OR SRP OR biosensors OR Nanomaterial-based biosensors OR electrochemical biosensors OR bioinspired recognition elements OR antibodies OR aptamers OR molecularly imprinted polymers OR Phylogenetic Evolution of Ligands for Exponential Enrichment (SELEX) OR fluorescence resonance energy transfer (FRET).
Publications until 29 August 2022 were included. This review only had the detection technology targeted at aflatoxin B1 in edible oil, and that had not included other types of toxins or other food carriers. After 4692 publications were searched, 596 full-text articles were reviewed and 132 articles were finally identified to meet our requirements.

2. Importance of Aflatoxins

Aflatoxins are a type of mycotoxins. They are highly toxic metabolites of fungi, produced in food and agricultural products. They have severe toxic effects, such as immunosuppressive, nephrotoxic, teratogenic, carcinogenic, and mutagenic, on human and animal health [34,35,36,37,38].
Aflatoxins can be divided into aflatoxin B1 (AFB1), aflatoxin B2 (AFB2), aflatoxin G1 (AFG1), and aflatoxin G2 (AFG2) according to their fluorescence properties and chromatographic mobility (Figure 1) [39,40,41]. Aflatoxin M1 (AFM1) and aflatoxin M2 (AFM2) are hydroxylated metabolites of AFB1 and AFB2, respectively. AFB1 is the most toxic among all AF species, with a high incidence rate and the most complex detection mechanism (Figure 2) [42].
AFB1 is a powerful carcinogenic, teratogenic, mutagenic, immunotoxic, hepatotoxic, and reproducible poison. Previous studies have shown that the toxicity of AFB1 is 10, 68, and 416 times that of KCN, arsenic and melamine, respectively [43,44] (Figure 2). Therefore, AFB1 has been classified as a class 1 carcinogen by many international authoritative organizations or institutions [45,46]. Due to the structural double bonds in the furan ring, AFB1 has high carcinogenicity and toxicity [17,47]. The lipophilic structure of atrial fibrillation promotes its entry into the blood through gastrointestinal and respiratory tracts [48,49]. Once AFB1 enters blood, it is distributed in various tissues and accumulates in the liver or other organs, resulting in liver cancer (Figure 3). In the liver, AFB1 produces a variety of metabolites through the hydroxylation and demethylation of the first-stage drug metabolism enzymes (for example, cytochrome P450 oxidase and CYP450 superfamily members, such as CYP1A2, CYP3A4, and CYP2A6) [50]. Metabolic reaction (internal and external) activates the final carcinogen AFB1 -8,9-epoxy metabolite, which covalently binds to cellular macromolecules (DNA, RNA, or protein) and plays a key role in acute and chronic poisoning. AFB1 residues also destroy the function of tumor suppressor genes (p53 and Rb) in the liver, which affects normal cells and leads to liver injury, increasing the probability of tumor and liver cirrhosis [51,52,53,54,55]. It is estimated that about 30% of liver cancers in the world are caused by AFB1. Its toxicity increases the infection rate of hepatitis B virus (HBV) and the risk of liver cancer [56]. A recent study found that the synergistic effect of AFB1 and HBV leads to liver cancer [50]. The reason is that HBV infection directly or indirectly exposes hepatocytes to AFB1 sensitive to tumors. The toxic effect of AFB1 is also related to dose, age, sex, nutrition, exposure time, and type [57]. In addition, AFB1 can be transmitted to the fetus through the placenta and affect the health of infants [58]. AFB1 exposure also inhibits immunity, thereby increasing the susceptibility to immunodeficiency virus attack and the probability of infection with other infectious diseases [59,60,61,62,63].

3. AFB1 Regulations on Edible Oil

Because AFB1 poses many hazards to the human body, many governments and international research institutions have made many efforts to control AFB1 pollution in different foods. For example, the FAO and the European Commission and Codex Alimentarius Commission have formulated regulations regarding the content of AFB1 in various foods to ensure consumer safety [64,65,66,67,68,69].
As for edible oil, most countries have no legislative restrictions and only a few countries, such as China, have effective regulations, laws, and standards for the highest level of AFB1 in different edible oils (Table 1). Due to some adverse conditions in the traditional oil processing process, AFB1 is usually degraded to the normal level in the extraction and refining process [17,70]. The EU has strict regulatory norms. The total amount of AFB1 and AF allowed in oilseeds is restricted to 2 and 4 μg kg−1, respectively. However, the maximum limit of AFs in oils has not been determined. The corresponding regulations in China, the United States, Kenya, and Thailand clearly stipulate the maximum level of total AFs in all edible oils, but the maximum level required is different. It is worth mentioning that in China, the AFB1 limit in corn and peanut oil is stipulated to be 20 μg kg−1, which may be because corn and peanut are most vulnerable to aflatoxin pollution [71,72]. See Table 1 for specific differences.

4. Methods for Detecting AFB1 in Edible Oil

The matrix is too complex for edible oil, and the mycotoxin content is relatively low, making it difficult to detect AFB1. Therefore, researchers have developed various traditional and modern methods to detect AFB1 in oil. AFB1 detection technology is mainly divided into chromatographic technology, spectroscopic technology, immunological technology, and biosensor technology [16,77].
Figure 4 briefly summarizes the LOD timelines for AFB1 detection in edible oils published from 2007 to 2022 mentioned in this review. It can be seen from the figure that with the advancement of time, no matter what type of detection technology or which specific detection method is used, the LOD of AFB1 in edible oil tends to be lower. This shows that people have a great interest in the detection of AFB1 in edible oil. At the same time, the wide use of new materials represented by nanomaterials highlights the interdisciplinary characteristics of new sensors. Next is a brief introduction of the identification method of AFB1, including its advantages and disadvantages, combined with actual cases.

4.1. Chromatographic Technology

4.1.1. High Performance Liquid Chromatography (HPLC)

High-performance liquid chromatography is a common official detection method. Many countries and institutions have used it, such as China’s national standard, the European Committee for Standardization (CEN), and the association of analytical organizations (AOAC). One characteristic of the HPLC method is that it can measure multiple targets with high sensitivity [78]. In recent years, researchers have developed new detection strategies combining HPLC with other sensors, such as fluorescence detection (FLD), ultraviolet (UV) detection, diode array detection, and mass spectrometry (MS) [79,80]. Compared to traditional HPLC, this further improves the reliability, sensitivity, and accuracy of target analytes and is widely used to detect harmful substances in food. For example, HPLC combined with FLD is the standard method for detecting AFB1 in edible vegetable oil [81,82,83,84,85,86]. HPLC–FLD was able to detect AFB1 levels as low as 0.01–0.04 μg kg−1 [81] and 0.005–0.03 μg L−1 [82].
Recently, liquid chromatography–tandem mass spectrometry (LC–MS–MS) methods are being increasingly used for the analysis of mycotoxins [85]. They have the advantages of not having a sample purification limitation during extraction, high resolution, high sensitivity, and suitability for various edible vegetable oils [19,87,88,89,90,91,92,93,94,95,96,97,98,99]. GC analysis is mostly used for volatile substances, and most mycotoxins are non-volatile, further limiting the application of GC in mycotoxin detection. A similar procedure to HPLC, UHPLC or UPLC is also used on the column to improve the resolution of AFB1. Hidalgo et al. [100] developed a new analytical method by coupling UHPLC to a triple quadrupole analyzer (UHPLC–QqQ–MS/MS), which was well validated and applied to monitor mycotoxins, including AFB1, in 194 samples of edible vegetable oil.
Many commonly used methods require sample preparation due to the different matrices of edible oil. Currently, a variety of methods are available for the extraction and isolation of mycotoxins from oil, such as liquid–liquid extraction or partitioning (LLE), frequently reported in the literature [101,102,103,104,105]; solid–phase extraction (SPE) [105,106,107,108,109]; immune affinity columns (IACs) [81,94]; IAC combined with dispersive liquid–liquid microextraction (DLLME) [91]; multifunctional cleanup columns [110]; the QuEChERS system [90]; gel permeation chromatography (GPC) [111]; immune assay extraction; and low-temperature cleanup (LTC) [112,113,114,115]. However, each method has its advantages and limitations. Thus, which method to choose still depends on the type of food matrix, mycotoxin characterization, and detection techniques [116].

4.1.2. Thin-Layer Chromatography (TLC)

Thin-layer chromatography (TLC) is an adsorption thin-layer chromatographic separation method suitable for complex mixed samples [117,118]. Since its development in the 1950s, thin-layer chromatography has been widely used in, for example, biology, medicine, and the chemical industry. It has recently been used in food analysis and quality control and has become a conventional technology in laboratories. Many reports have shown that TLC can be applied to all stages of the food industry, such as the stage of traditional substances, represented by food raw materials, ingredients, and additives, and the stage of unconventional substances, represented by harmful substances and pollutants. The detection and determination of compounds cover almost all substance categories [119,120,121,122].
Thin-layer chromatography uses the different adsorption capacities of each component to the same adsorbent so that when the mobile phase (the solvent) is flowing through the stationary phase (the adsorbent), there is continuous adsorption, desorption, readsorption, and redesorption to achieve the mutual separation of each component [123].
Although the TLC method has matured, it still has shortcomings, such as a low detection accuracy, volatility during the experiment being harmful to the experimental operators and the environment, and complex sample pretreatment [124,125]. In recent years, an interdisciplinary approach, such as the combination of TLC with image analysis and with new technologies, such as surface-enhanced Raman spectroscopy, mass spectrometry, and nuclear magnetic resonance, has further promoted the development of thin-layer chromatography and enhanced the practicability of this method in food analysis [126,127,128,129]. TLC is used to detect harmful substances in various foods, such as AF in edible oil, making it an effective analytical tool in food science methods [124,130].

4.2. Spectroscopic Technology

4.2.1. Fluorescence Spectrophotometry

Spectrum-based sensing technology has been developed and used to assess AFs contamination in food [131]. Among many spectral techniques, fluorescence spectrometry shows certain potential in determining AFs in a variety of agricultural products and foods [125,132,133]. Fluorescence spectrometry uses the target molecules in the sample to absorb ultraviolet or visible light to produce fluorescence and determine its molecular structure. It has excellent detection sensitivity and specificity in the study of AFs and other chemical components [134,135]. The study found that the fluorescence phenomenon is conducive to the characterization and monitoring of target detection objects. For example, AFB1 can emit a specific range of fluorescence (425–500 nm) under the excitation of UV light source (340–400 nm), which provides the possibility of using fluorescence spectroscopy to analyze AFB1 in different foods [135,136]. In recent years, laser-induced fluorescence (LIF) technology has developed rapidly and attracted more attention because it uses a certain wavelength of laser light source and has better specificity and detection sensitivity. The advantage of LIF is that it can realize online, rapid and nondestructive direct detection according to the characteristic fluorescence peak of AFB1. Researchers have developed a detection model based on LIF, which can quickly and accurately screen AFB1 in different edible oils. The information and conclusions obtained in the study further show that LIF technology can be used for rapid and nondestructive detection of AFB1 in different edible oils [19,137]. However, LIF is also vulnerable to the interference of external factors, such as the power and accuracy of the instrument, the environmental factors of temperature and humidity, and the physical and chemical index factors of the detected object. Although this limits the wide application of LIF technology, researchers are still trying and exploring.

4.2.2. Infrared (IR) Spectroscopy

Infrared spectroscopy (IRs) has the characteristics of rapid detection, simple sample preparation process and strong adaptability. It has been widely proven to be an effective food safety detection and control technology. Because IR covers a wide range of electromagnetic spectra (780 to 2500 nm), IR can be applied to the detection of a variety of foods including edible oil, meat, aquatic products, fruits and vegetables [138,139,140,141,142,143,144]. When IRs radiation penetrates the sample, the radiation is reflected, absorbed or transmitted by molecular bonds, resulting in the energy change of light, which can reflect some characteristic chemical bonds, thus reflecting the characteristics of the tested product [145,146]. In the application of edible oil, IR shows many abilities, such as distinguishing different kinds of oil, grading the quality of oil, detecting harmful substances in oil, etc. [138,143,147,148,149,150]. Using near infrared (NIR) technology to detect mold in edible oil has also been a research hotspot in recent years. Researchers have promoted the further application and development of IR technology by establishing qualitative and quantitative analysis models for AFB1 pollution in edible oil [151,152,153].

4.2.3. Terahertz (THz) Spectroscopy

With the development of optical and electronic technology, terahertz spectroscopy (THz) has been a revolutionary development, and shows great potential as a new technology tool for nondestructive food testing [154,155,156,157]. As a technical information link between microwave spectroscopy and infrared spectroscopy, THz has the characteristics of both, making it widely used in basic research and industrial practice [158,159]. Like other spectral technologies, thanks to the development of chemometrics methods, THz has become a powerful technical tool in the food industry, due to its strong detection and quantification capabilities [156,157,160]. Through the combination of THz and chemometrics methods, researchers have constructed a rapid nondestructive detection model for AFB1 in edible oil. Although the accuracy is slightly lower than other conventional analysis methods, it provides a possibility for THz in food safety detection [161]. In a recent study, researchers further improved the accuracy of THz in detecting AFB1 in edible oil by adding pretreatment and other methods on the basis of predecessors, and reduced the LOD of AFB1 to 1 μg kg1, and the accuracy is improved to more than 90% [161,162]. The cross integration of THz and chemometrics and other disciplines is conducive to promoting its application and development in the detection of AFS in the edible oil industry. At the same time, the limitations of THz should also be clear, such as the low detection limit and sensitivity advantage are not obvious, the penetration of the detected object is limited, there is scattering effect, the technology is expensive, the database is lack, etc. [163].

4.2.4. Surface-Enhanced Raman Spectroscopy (SERS)

As a complementary analysis technology of IR, the Raman spectroscopy (RS) technology is sensitive to the symmetrical vibration of covalent bonds of non-polar groups (such as C=O, C-C and S-S) [164,165,166]. Therefore, RS has the advantages of being fast, sensitive and simple in the detection and evaluation system of food [165,167,168]. However, traditional RS has some limitations, such as Raman scattering. Therefore, researchers have developed SERS signal enhancement technology represented by electromagnetic field enhancement and chemical enhancement [165]. At present, the application of SERS technology in the detection of AFs is still challenging, and the intersection of technology development and multidisciplinarity (such as materials science, stoichiometry, etc.) is the focus of researchers [165]. In recent years, researchers have reported a variety of SERS schemes for AFB1 detection in edible oil, such as SERS tag detection using antibodies and aptamers, sandwich immunoassay based on SERS, etc. [169,170,171,172,173,174]. The growing research results show that SERS technology is becoming a powerful tool to ensure the safety development of the food industry, especially in the safety supervision of AFs. However, it cannot be denied that challenges still exist, such as the development of targeted new materials, the optimization of key core technologies, and the practical application of research results [175,176,177].

4.3. Immunological Technology

Enzyme-Linked Immunosorbent Assay (ELISA)

In recent years, researchers have often used immunochemical methods to determine mycotoxins in food, in addition to traditional chromatographic techniques. The core of immunochemistry is the specific interaction between immunoglobulin (Igs) and antigen (Ag). Several immunochemical methods have been applied to detect mycotoxins in edible vegetable oils, such as enzyme-linked immunosorbent assay (ELISA) and biosensors based on immunoassay.
ELISA is one of the most commonly used methods for detecting mycotoxins [24]. It has been designed and developed on the basis of the principle of specific immune responses between Igs and Ags. The specificity of this immunoassay is due to the use of enzyme-labeled Igs or Ags and solid-matrix-restricted immunoglobulins to capture unlabeled silver in the analyte and detect it with labeled immunoglobulins. Although ELISA is well developed and widely used in food analysis, clinical practice, biotechnology, environmental, chemical, and other industries, it still has several deficiencies, such as excessive dependence on the matrix caused by the interaction between the target antigen and matrix components. The standard ELISA is composed of four main parts (immune-recognition element, sorbent substrate, enzyme label, and chromogenic reagent), and the deficiency of the central part is the root cause of the limitation of ELISA. In recent years, researchers have used the cross-fusion of multiple technologies to drive the performance of one of the components or the whole ELISA, especially in terms of sensitivity, accuracy, and stability [27].
For mycotoxins, due to the high singularity of ELISA, the developed kit has specific recognition ability and has been widely used in the detection of mycotoxins [70]. For example, Qi et al. [20] used ELISA and UPLC–MS/MS to detect AFB1 in peanut oil, although the LOD was only 1.08 μg kg−1, much higher than the LOD of UPLC–MS/MS (the LOD is 0.01 μg kg−1) [20]. It has been affirmed because of its accuracy, rapidity, and other advantages. For the actual detection of other harmful substances, such as AFB1, AFB2, AFG1 and AFG2, in different edible oils (oils of soybean, coconut, peanut, fennel, melon, and palm kernel), ELISA showed satisfactory results and the concentration was lower than the legislative limit [178,179,180]. On this basis, the researchers developed a commercial ELISA kit that can detect AFB1, which can be applied to a variety of samples including edible oil, and the detection limit can be as low as 3 ppb. Although the current ELISA technology or kit still has problems such being as time-consuming, high cost, and cumbersome operation, with the advancement of technology, ELISA technology shows strong application potential [27].

4.4. Electrochemical Biosensing Technology

Due to rapidity, small footprint, economy, sensitivity, and unique capabilities, electrochemical biosensing devices have received particular attention in assessing food quality, mainly reflecting AFB1 levels in food samples [181]. The AFB1 electrochemical biosensor can produce various types of analytical signals, such as voltage, current, and impedance [182,183]. The standard transduction methods are amperometric, electrochemical impedance spectroscopy (EIS), and voltammetry (potentiometry).

4.4.1. Amperometric Biosensors

The amperometric biosensor is an electrochemical device with high selectivity and sensitivity that takes the change in the measuring current as the analysis signal. Because the change in the current is closely related to the concentration of AFB1 in food samples and the change can be achieved by maintaining a stable potential, an amperometric biosensor is relatively perfect. A typical amperometric biosensor consists of two- or three-electrode systems (containing a functional electrode, a reference electrode, and an auxiliary electrode), and the analytical performance of the latter is significantly higher than that of the former (Figure 5) [181]. This is because the additional auxiliary electrode not only increases the area of the detection surface but also increases the current between it and the functional electrode, as well as the operating potential between the functional electrode and the reference electrode, thereby enhancing the changes in the detection process of AFB1 in food in electronic dynamics. On the contrary, the dual-electrode system does not include auxiliary electrodes, which may lose their function at high temperatures. Therefore, amperometric biosensors with dual-electrode systems are not used to analyze the quality of food samples [181].
Even functional electrodes are usually made of inert metal materials (such as platinum, gold) or carbon (graphite, glassy carbon). The main drawback is reproducibility of measurements. Currently, printed electrodes have become a good substitute because their cost and mass production can be controlled [70,78].
Researchers used two kinds of nanomaterials with different charges to deposit on the electrode alternately, obtaining a multilayer electrode with a sandwich structure with excellent conductivity and rich electrochemical active sites [184]. Such a biosensor has good selectivity, reproducibility, and stability. In the subsequent optimization test, the optimized electrochemical biosensor was found to have significant stability and even after being placed for a period of time, it showed good LOD (0.002 ng mL−1). This sensor is believed to be one of the best biosensors for detecting mycotoxins. Researchers applied the electrochemical biosensor to detect AFB1 in real oil samples and found that it has a good recovery rate (98.11–103.36%).
Xuan et al. [185] developed an integrated AFB1 detection platform that uses disposable screen-printed electrodes (SPEs), allowing routine detection without electrode modification. According to the SPE used, the platform can simplify the tedious sample processing process through high-throughput processing, reduce operating errors, and improve experimental reproducibility, which can benefit large-scale sample processing. The detectable concentration range of AFB1 was 0.08–800 μg kg−1 with a LOD of 0.05 μg kg−1. Analysis of real samples and verification of the method showed the results of the new sensor to be consistent with those of the classical method (LC–MS/MS), indicating that the developed method has the potential to monitor AFB1 in peanut oil.
Another study reported a new aflatoxin biosensor based on the AFB1 inhibition of acetylcholinesterase (AChE) [186]. The core of this method is to immobilize choline oxidase on a screen-printed electrode modified with Prussian blue (PB). The electrode used in the biosensor can detect H2O2 at low potential. As per the results, the linear operating range of the biosensor is estimated to be 10–60 ppb and the LOD is 2 ppb. On using real olive oil samples to evaluate the sensor, the recovery rate was found to be 78 ± 9% at 10 ppb.

4.4.2. Electrochemical Impedance Spectroscopy (EIS)

Electrochemical impedance spectroscopy (EIS) technology is an effective monitoring tool for identifying and monitoring changes in mycotoxins at the interface between electrode surface modifications. When the target analyte is combined with a biometric element, it generates an electrochemical response by changing conductivity and capacitance through an impedance biosensor [187]. These biosensors monitor the impedance changes caused by the interaction between the target detection object, such as AFB1, and the biometric element fixed on the working electrode, and display the detection results in the form changed electron flow on the working electrode [188,189]. Typical potentiometric sensors are also suitable for the three-electrode system.
The main parameter of the EIS is the charge transfer resistance value (RCT), which is closely related to the reaction between immobilized mycotoxins and the antibody antigen and is also proportional to the target detector/concentration of the target [65,66]. For determining AFB1, Yu et al. [190] reported a sensitive and convenient EIS method involving MWCNT/RTIL/Ab-modified electrodes coated in bare GCE. The experimental results show that the resistance of the MWCNT/RTIL/Ab-modified electrode (605.6 Ω) is higher than that of bare GCE (151.9 Ω). When AFB1 was immobilized on MWCNT/RTIL/Ab-modified electrodes, the increase in the electron transfer resistance (Ret) value was found to be directly related to the AFB1 amount. The specific interaction between AFB1 and Ab causes an increase in the Ret value, which leads to the production of electrically insulating biological conjugates, which will prevent the electron transfer process of redox probes. Therefore, the EIS measurement results are consistent with the above cyclic voltammetry results. Because of its simple characteristics, this method can be widely used to detect various agricultural products and edible oils.
For many researchers exploring mycotoxin detection methods, aptamer-based EIS has become a hot research topic. Aptamer-based impedance biosensors have achieved satisfactory results in detecting mycotoxins in food and have great potential for practical application in edible oils.

4.4.3. Voltammetry Biosensors

Voltammetric biosensors solve the problem of obtaining analytical data using ion-selective electrodes. Similar to amperometric biosensors, voltammetry also requires a two- or three-electrode system. When the current is constant, it can detect target analytes, such as AFB1, in food samples by evaluating the change in circuit potential between the functional electrode and the reference electrode [60,191].
Biosensors have also shown promising results in detecting the AFB1 content in edible oils. For example, Wang et al. [192] developed a new disposable electrochemical biosensor based on strip** voltammetry to detect copper ions released from copper apatite. The biosensor uses copper ions as a signal label to immobilize AFB1 antibody on a screen-printed carbon electrode (SPCE) modified by gold nanoparticles. The detection is performed by the voltammetric signal of the dissolution of copper ions released from acid hydrolysis of copper apatite, and copper apatite increases the number of loaded copper ions. The electrochemical signal is further amplified. Peanut oil was used to evaluate the reliability and application potential of biosensors. Researchers believe that this new method will be applied to many fields in the near future because of its many excellent characteristics (low cost, rapidity, accuracy, and high sensitivity).

4.4.4. Nanomaterial-Based Biosensors

Recently, different nanomaterials, such as carbon and metal, have been used to modify the active surfaces of macroelectrodes and microelectrodes to design electrochemical biosensors for the detection of AFB1 [193,194,195]. This is because new biosensors directly use nanomaterials or other materials containing nanoparticles that show significant characteristics, such as high sensitivity and specificity for detecting targets, reliability, and consistency of products [181,196,197]. Nanomaterials significantly increase the effective surface area of biosensors and further improve the analytical performance [60,194]. Nanomaterials also enhance some characteristics of biometric elements in biosensor devices in terms of electrical, catalytic, optical, and thermal properties [198]. According to previous studies, some of the key functional enhancements are the enhanced immobilization of biomolecules, generation and expansion of analytical signals, and enhanced usability of fluorescent labels.

Characteristic of Nanomaterials Based Electrochemical Biosensors

The role of nanomaterials in biosensors is mainly reflected in the immobilization of biomolecules, signal generator, fluorescent labeling, and signal amplification.
Nanomaterials not only immobilize biomolecules but also increase the interaction between different molecular materials. In addition, nanomaterials enhance the stability of biomolecular immobilization, thereby increasing the signal strength of the immunoassay [31]. Metal nanomaterial particles, such as AgNPs and MOFs, can increase the surface area and biocompatibility of biomolecules bound to the detection target. However, non-metallic nanomaterials show negatively charged functional groups, which can be used as an effective carrier to bind and fix with positively charged targets.

Signal Generator

Xue et al. [31] reported that, when the photoelectric signal changes, nanoparticles such as gold and silver can act as a signal generator. By adjusting the fluorescence signal generated by nanomaterials, a new AFB1 nanoprobe can be constructed. In addition, because these nanoparticles can be prepared in different sizes according to need, they have good functionality, stability, and scalability [133,199].

Fluorescent Label

Nanomaterials have unique optical properties that enable them to be widely used in a variety of disciplines, especially in the detection of hazardous substances in food. Nanomaterials can detect AFB1 by sensing optical signals (absorbance, chemiluminescence, fluorescence, etc.) [31]. Some nanomaterials, such as metal nano-ions and quantum dots, have been used as fluorescence quenching agents because of the ability of AFB1 to directly quench or reduce the fluorescence intensity. In addition, quantum dots have transformed fluorescein into a marker element that binds to aptamers or antibodies.

Signal Amplification

Nanomaterials can also be used as functional materials for various electrodes, signal components, etc., to amplify signals in various ways. For example, on the electrode surface of electrochemical sensors, nanomaterials such as gold and silver can amplify the analytical signal by enhancing the redox reaction. Some metal nanoparticles, such as gold, can amplify signals related to their characteristics, such as unique catalytic activity, biocompatibility, and multiple absorption sites. Carbon, graphene, and other non-metallic nanomaterials improve the analytical performance by increasing the surface area.

4.5. Bioinspired Recognition Elements for Biosensors

A biosensor is independent quantitative analysis equipment used to study the analytes required in different types of food samples. A biosensor consists of many parts [29,30] (Figure 6). Biometric elements are the core components of biosensors and can detect specific target analytes. The quality of biometric elements usually determines the specificity and sensitivity of analysis [200,201]. Biorecognition elements, including antibodies, aptamers, molecularly imprinted polymers, and enzymes, have been used to manufacture biosensors [34,64,202]. These elements show increased sensitivity and selectivity for target analytes. Critical biometric elements for develo** biosensors to detect AFB1 in edible vegetable oil are elaborated below.

4.5.1. Antibody

Antibodies have been used as recognition elements for develo** biosensors because of their specificity and sensitivity [200]. Biosensors that use antibodies as recognition elements are called immunosensors, and their mechanism relies on the specific recognition of aflatoxin epitopes by antibodies.
The first batch of polyclonal antibodies, developed in 1976, became the basis for most mycotoxin detection methods. In the following decades, polyclonal and monoclonal antibodies were the basis for most mycotoxin detection methods [192,200,203]. Today, in addition to monoclonal and polyclonal antibodies, various other types of antibodies are being used to detect target analytes. Researchers have developed an antibody-based immunosensor that can directly recognize AFB1 and is used in peanut oil with a concentration range of 0.001 to 100 ng mL−1, with a detection limit of 0.2 pg mL−1 [192].
However, the production of monoclonal antibodies and polyclonal antibodies is complex and the antibodies degrade, denature, and aggregate easily [204,205]. In recent years, with the development of protein- and DNA-based new engineering technology, it has become possible to develop modified and recombinant antibodies (RAbs). RAbs integrate many advantages of biosensors, such as simple operation and a high degree of automation, high throughput screening, low requirements for configuration attributes, and the trend of more miniaturization [200]. Zhao et al. [206] developed a novel method of MB-dcELISA for AFB1 based on the mimotope of an RAb and nanobody. This study effectively proved that compared with monoclonal antibodies, an RAb is more economical and easier to prepare. Compared with chemically synthesized toxic antigens, immunoassay is safer and performs better in validation studies. In real samples (corn germ oil and peanut oil), the LOD of AFB1 is as low as 0.13 ng mL−1. Other researchers also designed an RAb with increased sensitivity to low-molecular-weight haptens, and this RAb was validated in olive oil with a lower LOD (0.03 ng mL−1) for AFB1 [190].
Researchers recently found that, by increasing the immobilization of antibodies and giving full play to the characteristics of specific antibodies, the performance of the sensor can be effectively improved, and on this basis, some immunosensors have been developed for detecting AFB1 in edible oil [133,196]. For example, to determine AFB1, Shi et al. [207] proposed a novel immobilized immunosensor based on graphene supported with hybrid gold nanoparti-cles-poly4-aminobenzoic acid. In the study, after the reduction in graphene oxide by PABA via an epoxy ring opening reaction, the nanocomposite PABA-r-GO was obtained. Then, gold nanoparticles (AuNPs) were prepared on this basis to form a Au-PPABA-r-GO nanohybrid. The final sensor was obtained by the covalent binding of the COOH group of functional nanocomposites with an AFB1-specific antibody. The sensor has good performance (linear range 0.01–25 ng mL−1 and LOD 0.001 ng mL−1) and has been successfully applied to detect real vegetable oil. This sensor also has good reproducibility and selectivity, especially stability, and can be stored at a low temperature for a long time.

4.5.2. Aptamers

Aptamers are single-stranded RNA or DNA (20–90 oligonucleotide sequences with specific sequences) that can bind to various targets, such as ions, antibodies, proteins, cells, and organic molecules [208]. The particular recognition ability of aptamers relies on the three-dimensional structure of a high-affinity target-induced DNA three-dimensional structure. The researcher procured specific targets for aptamers by screening oligonucleotides using the Phylogenetic Evolution of Ligands for Exponential Enrichment (SELEX) program. Aptamer sensors are biosensors integrated with aptamers developed in the 1990s [200,208,209,210,211].
Recently, aptamers have attracted significant attention in food contamination analysis and are used for various sensing applications due to their inherent benefits: (1) aptamers are obtained from in vitro synthesis, so animals are not necessary; (2) aptamers have lower toxicity, immuno-genicity, and production cost; (3) aptamers have enhanced chemical and thermal stability; (4) aptamers have excellent batch-to-batch reproducibility; (5) aptamers have a smaller size and show a remarkable ability to penetrate the tissue and adhere to target molecules; and (6) it is possible to change their structure [193,212,213].
Notably, the immobilization of aptamers is a critical step in biosensor design because it can affect the affinity of aptamers for their targets and their long-term stability in fundamental sample analysis.
Therefore, researchers have developed many strategies to immobilize aptamers: (1) adsorption or π–π stacking interactions between DNA bases and modified graphene oxide (GO) interface [214], (2) aptamers with carboxylic acids on surfaces or nanomaterial covalent bonding of groups [184,215], (3) binding of sulfide aptamer with CdTe quantum dots (QDs) or gold-based materials [216], (4) binding with avidin or other affinity interactions based on biotin streptomycin affinity [200,217,218], and (5) hybridization with partially complementary single-stranded DNA previously fixed on the surface of nanoparticles [219,220,221,222].
Table 2 describes some examples of aptasensors recently reported for the detection of AFB1 in edible oils. About half of the previous reports have been based on fluorescent mycotoxin aptamer sensors. Some of them use metal or nanostructured materials, such as gold nanoparticles (AuNPs), GO, single-walled carbon nanotubes, or TiO2 tubes, and are used to prepare aptamer sensors.
Nanometer material has always been the focus of research, and its applications in biosensors are also diverse. Black phosphorus nanosheets (BPNSs) have great application prospects in biosensors due to their unique characteristics [223]. Wu et al. [224] developed a highly specific and sensitive aptamer sensor (UCNPs-BPNSs) based on the team’s research on upconversion nanoparticles (UCNPs) [196]. The research team attached UCNPs to the surface of BPNSs at a very small space distance (less than 10 nm) through glutaraldehyde crosslinking method and π-π stacking effect method, and then constructed the fluorescence resonance energy transfer (FRET) system. This aptamer sensor can effectively detect AFB1 in peanut oil and other foods quantitatively with good linear range (0.2–500 ng mL−1) and LOD (0.028 ng mL−1).
** improved MIPs [257,258,259]. The key to the success of the sensor of an MIP is whether the MIP is effectively attached to the transducer. Three commonly used immobilization methods are in situ polymerization, electropolymerization, and physical coating. Additionally, the number of applications of MIP sensors for detecting AFB1 in edible vegetable oil is limited [200,202,260].

5. Conclusions and Perspectives

Mycotoxin contamination, especially AFB1 contamination in edible oil, is usually unavoidable. A more sensitive and rapid sensor-based early warning tool for AFB1 detection would help to reduce risk. Various traditional, modern, and biosensing technologies have been used to detect toxins in contaminated food. Spectroscopic techniques, chromatographic techniques are general methods for the detection of AFB1 in edible oils. In recent years, based on the cross-integration of multiple disciplines, the innovation, progress and development of general methods have also been promoted. Although traditional chromatographic techniques can effectively detect mycotoxins, their performance in all aspects cannot achieve satisfactory results. Combined use with other sensor equipment can effectively improve reliability, sensitivity and accuracy. However, due to the high cost of equipment, on-site inspection cannot be performed, and sample pretreatment is required, which limits the use of chromatography technology in the detection of AFB1 in edible oil. The development of spectroscopic techniques has become increasingly diverse and can effectively detect mycotoxins, especially AFB1 in edible oils. However, these methods are not suitable for on-site detection, because they still have many shortcomings, such as low sensitivity and reliability, and the need for professional personnel to operate.
Unlike conventional detection techniques, novel biosensors show high accuracy, sensitivity, and specificity; better cost controllability and portability; and reliability and simplicity in operation.
This review also discusses the development of important recognition elements in sensors. The recognition element of the sensor should have sensitivity and specificity sufficient enough to detect small amounts of target toxins, even in samples with complex matrix systems. The development and use of nanomaterials further improve the efficiency of biosensor conversion systems, but these require further improvements in their sensitivity, selectivity, and reproducibility. Of course, the stability and cost will also affect the selection of identification elements, which can improve the practicability.
Despite significant progress in biosensors for the detection of AFB1, there are some problems and challenges in the future. (1) The recognition elements of biosensors (such as metal nanoparticles, quantum dots, and graphene) improve the efficiency of sensing systems, but all these require further improvements in terms of sensitivity, selectivity, and reproducibility. (2) Future studies can perform AFB1 toxicity measurements and develop advanced nanomaterial-integrated biosensors to improve the overall detection of harmful substances, such as AFB1, in contaminated food samples. (3) When detecting AFB1 in contaminated food samples, researchers can focus on combining biosensing systems with microarray technology to fabricate more portable devices. (4) Reagent-free, clean-free, calibration-free, or nonbiological contamination biosensors for aflatoxin analysis require more effort and will reduce the possible future hazards.

Author Contributions

S.Y. Conceptualization, investigation, writing, review and editing of the manuscript; L.N. conceptualization and investigation of the manuscript; Y.L. review of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially supported by Key R&D project of Jiangsu Province (BE2021306); and Development Project (BE2021306) and the Shandong Province Key Research and Development Program (No. 2021CXGC010808).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AChEAcetylcholinesterase
Adanrs Aunr@DNTB@Ag Nanorods
AFB1 Aflatoxin B1
AFB2 Aflatoxin B2
AFBO Aflatoxin B1–8,9-Epoxide
AFG1 Aflatoxin G1
AFG2 Aflatoxin G2
AFM1Aflatoxin M1
AFM2 Aflatoxin M2
Afs Aflatoxins
AIE Induced Emission
AOAC Association of Analytical Communities
AuNPsGold Nanoparticles
BPNSsBlack phosphorus nanosheets
CBNs Carbon-Based Nanomaterials
CEN European Committee for Standardization
CODEX European Commission and The Codex Alimentarius Commission
CV Cyclic Voltammetry
CQDSCarbon Quantum Dots
CSNPsCore-Shell Nanoparticles
CYP Cytochrome P
DLLME DispersiveLiquid-Liquid Microextraction
DNTB 5,5′-Dithiobis (2-Nitrobenzoic Acid)
DS dual-terminal stemmed
DTNsDNA tetrahedral nanostructures
ECLElectrochemiluminescence
EIS Electrochemical Impedance Spectroscopy
ELISA Enzyme-Linked Immunoassay
EU European Union
EXO IExonuclease I
FAO Food and Agriculture Organization
FRETFluorescence Resonance Energy Transfer
GCEs Glassy Carbon Electrodes
Gdadnts Gold Nanotriangles (Gnts)-DTNB@Ag-DTNB Nanotriangles
GO Graphene Oxide
GODGraphene Quantum Dots
GPC Gel Permeation Chromatography
GST Glutathione-S-Transferase
HCRHybridization Chain Reaction
HBV Hepatitis B Virus
HPLC-FD High-Performance Liquid Chromatography Coupled with A Fluorescence Detector
HPLC-MS High-Performance Liquid Chromatography Coupled with Mass Spectrometry
IacsImmunoaffinity Columns
IMSPEImmuno magnetic solid phase extraction
IRsInfrared spectroscopy
ITC Isothermal Titration Calorimetry
KCN Editpotassium Cyanide
LC-MS-MS Liquid Chromatography-Tandem Mass Spectrometry
LIFLaser-Induced Fluorescence
LLE Liquid-Liquid Extraction or Partitioning
LOD Limits of Detection
LTCImmune Assay Extraction and Low Temperature Cleanup
MIPs Molecularly Imprinted Polymers
MNPsMetal Nanoparticles
MST Microthermophoresis
NH2-DNA1Amino-Terminal AFB1 Aptamer
NH2-Rh5-aminotetramethylrhodamine
NIRNear Infrared
PABA-R-GOHybrid 4-Aminobenzoic Acid-Reduced Graphene Oxide
PBPrussian Blue
QDsQuantum Dots
QuEChERS Quick, Easy, Cheap, Effective, Rugged, and Safe
RAbsRecombinant Antibodies
RSRaman spectroscopy
SELEXSystematic Evolution of Ligand by the Exponential Enrichment Process
SERS Surface-Enhanced Raman Spectroscopy
SH-DNA2Complementary Aptamer
SPCE Screen-Printed Carbon Electrode
SPESolid Phase Extraction
THzTerahertz
TLCThin-Layer Chromatography
TMB3,3,5,5-Tetramethylbenzidine
UCNPsUpconversion Nanoparticles
UHPLC/UPLCUltra-High Performance Liquid Chromatography
UHPLC-Qqq-MS/MSUltra-High Performance Liquid Chromatography Coupled to A Triple Quadrupole Analyzer
US-FDAUS Food and Drugs Administration
WHOThe World Health Organization
β-CDβ-cyclodextrin

References

  1. Shabeer, S.; Asad, S.; Jamal, A.; Ali, A. Aflatoxin Contamination, Its Impact and Management Strategies: An Updated Review. Toxins 2022, 14, 307. [Google Scholar] [CrossRef] [PubMed]
  2. Skrzydlewski, P.; Twaruzek, M.; Grajewski, J. Cytotoxicity of Mycotoxins and Their Combinations on Different Cell Lines: A Review. Toxins 2022, 14, 244. [Google Scholar] [CrossRef]
  3. Holban, A.M.; Grumezescu, A.M. Microbial Contamination and Food Degradation; Academic Press: London, UK, 2018; Volume 10. [Google Scholar]
  4. Pickova, D.; Ostry, V.; Malir, F. A Recent Overview of Producers and Important Dietary Sources of Aflatoxins. Toxins 2021, 13, 186. [Google Scholar] [CrossRef] [PubMed]
  5. Anater, A.; Manyes, L.; Meca, G.; Ferrer, E.; Luciano, F.B.; Pimpão, C.T.; Font, G. Mycotoxins and their consequences in aquaculture: A review. Aquaculture 2016, 451, 1–10. [Google Scholar] [CrossRef]
  6. Wang, Y.; Nie, J.; Yan, Z.; Li, Z.; Cheng, Y.; Chang, W. Occurrence and co-occurrence of mycotoxins in nuts and dried fruits from China. Food Control 2018, 88, 181–189. [Google Scholar] [CrossRef]
  7. Lee, H.J.; Ryu, D. Worldwide Occurrence of Mycotoxins in Cereals and Cereal-Derived Food Products: Public Health Perspectives of Their Co-occurrence. J. Agric. Food Chem. 2017, 65, 7034–7051. [Google Scholar] [CrossRef]
  8. Azam, M.S.; Ahmed, S.; Islam, M.N.; Maitra, P.; Islam, M.M.; Yu, D. Critical Assessment of Mycotoxins in Beverages and Their Control Measures. Toxins 2021, 13, 323. [Google Scholar] [CrossRef]
  9. Mahato, D.K.; Lee, K.E.; Kamle, M.; Devi, S.; Dewangan, K.N.; Kumar, P.; Kang, S.G. Aflatoxins in Food and Feed: An Overview on Prevalence, Detection and Control Strategies. Front. Microbiol. 2019, 10, 2266. [Google Scholar] [CrossRef]
  10. Ünüsan, N. Systematic review of mycotoxins in food and feeds in Turkey. Food Control 2019, 97, 1–14. [Google Scholar] [CrossRef]
  11. Ahlberg, S.H.; Joutsjoki, V.; Korhonen, H.J. Potential of lactic acid bacteria in aflatoxin risk mitigation. Int. J. Food. Microbiol. 2015, 207, 87–102. [Google Scholar] [CrossRef]
  12. Jard, G.; Liboz, T.; Mathieu, F.; Guyonvarc’h, A.; Lebrihi, A. Review of mycotoxin reduction in food and feed: From prevention in the field to detoxification by adsorption or transformation. Food Addit. Contam. Part A 2011, 28, 1590–1609. [Google Scholar] [CrossRef] [PubMed]
  13. OECD/FAO. OECD-FAO Agricultural Outlook 2022–2031; OECD Publishing: Paris, France, 2022. [Google Scholar] [CrossRef]
  14. Mao, X.; Yan, A.; Wan, Y.; Luo, D.; Yang, H. Dispersive Solid-Phase Extraction Using Microporous Sorbent UiO-66 Coupled to Gas Chromatography-Tandem Mass Spectrometry: A QuEChERS-Type Method for the Determination of Organophosphorus Pesticide Residues in Edible Vegetable Oils without Matrix Interference. J. Agric. Food Chem. 2019, 67, 1760–1770. [Google Scholar] [CrossRef] [PubMed]
  15. Pitt, J.I.; Taniwaki, M.H.; Cole, M.B. Mycotoxin production in major crops as influenced by growing, harvesting, storage and processing, with emphasis on the achievement of Food Safety Objectives. Food Control 2013, 32, 205–215. [Google Scholar] [CrossRef]
  16. Vasseghian, Y.; Moradi, M.; Dragoi, E.-N.; Khaneghah, A.M. A review on mycotoxins detection techniques in edible oils. Int. J. Environ. Anal. Chem. 2020, 102, 2125–2139. [Google Scholar] [CrossRef]
  17. Javanmardi, F.; Khodaei, D.; Sheidaei, Z.; Bashiry, M.; Nayebzadeh, K.; Vasseghian, Y.; Mousavi Khaneghah, A. Decontamination of Aflatoxins in Edible Oils: A Comprehensive Review. Food Rev. Int. 2020, 38, 1–17. [Google Scholar] [CrossRef]
  18. Shavakhi, F.; Rahmani, A.; Piravi-Vanak, Z. A global systematic review and meta-analysis on prevalence of the aflatoxin B1 contamination in olive oil. J. Food. Sci. Technol. 2022, 1–10. [Google Scholar] [CrossRef]
  19. He, X.; Zhang, Y.; Yang, X.; Chen, M.; Pang, Y.; Shen, F.; Fang, Y.; Liu, Q.; Hu, Q. Estimating bulk optical properties of AFB1 contaminated edible oils in 300-900 nm by combining double integrating spheres technique with laser induced fluorescence spectroscopy. Food Chem. 2022, 375, 131666. [Google Scholar] [CrossRef]
  20. Qi, N.; Yu, H.; Yang, C.; Gong, X.; Liu, Y.; Zhu, Y. Aflatoxin B1 in peanut oil from Western Guangdong, China, during 2016-2017. Food Addit. Contam. Part B 2019, 12, 45–51. [Google Scholar] [CrossRef]
  21. Li, S.; Li, X.; Zhang, Q. Advances in the development of detection techniques for mycotoxins in vegetable oil. Chin. J. Chromatogr. 2019, 37, 569–580. [Google Scholar] [CrossRef]
  22. Ji, J.; Jiang, M.; Zhang, Y.; Hou, J.; Sun, S. Co-occurrence of aflatoxins in plant oil products from China. Food Addit. Contam. Part B 2022, 1–8. [Google Scholar] [CrossRef]
  23. Einolghozati, M.; Talebi-Ghane, E.; Ranjbar, A.; Mehri, F. Concentration of aflatoxins in edible vegetable oils: A systematic meta-analysis review. Eur. Food Res. Technol. 2021, 247, 2887–2897. [Google Scholar] [CrossRef]
  24. Bordin, K.; Sawada, M.M.; Rodrigues, C.E.d.C.; da Fonseca, C.R.; Oliveira, C.A.F. Incidence of Aflatoxins in Oil Seeds and Possible Transfer to Oil: A Review. Food Eng. Rev. 2014, 6, 20–28. [Google Scholar] [CrossRef]
  25. Shephard, G.S. Aflatoxins in peanut oil: Food safety concerns. World Mycotoxin J. 2018, 11, 149–158. [Google Scholar] [CrossRef]
  26. Wu, L.X.; Ding, X.X.; Li, P.W.; Du, X.H.; Zhou, H.Y.; Bai, Y.Z.; Zhang, L.X. Aflatoxin contamination of peanuts at harvest in China from 2010 to 2013 and its relationship with climatic conditions. Food Control 2016, 60, 117–123. [Google Scholar] [CrossRef]
  27. Wu, L.; Li, G.; Xu, X.; Zhu, L.; Huang, R.; Chen, X. Application of nano-ELISA in food analysis: Recent advances and challenges. TrAC, Trends Anal. Chem. 2019, 113, 140–156. [Google Scholar] [CrossRef]
  28. Hayashi, Y.; Matsuda, R.; Maitani, T.; Imai, K.; Nishimura, W.; Ito, K.; Maeda, M. Precision, limit of detection and range of quantitation in competitive ELISA. Anal. Chem. 2004, 76, 1295–1301. [Google Scholar] [CrossRef]
  29. Bhalla, N.; Jolly, P.; Formisano, N.; Estrela, P. Introduction to biosensors. Essays Biochem. 2016, 60, 1–8. [Google Scholar] [CrossRef]
  30. Naresh, V.; Lee, N. A Review on Biosensors and Recent Development of Nanostructured Materials-Enabled Biosensors. Sensors 2021, 21, 1109. [Google Scholar] [CrossRef]
  31. Xue, Z.; Zhang, Y.; Yu, W.; Zhang, J.; Wang, J.; Wan, F.; Kim, Y.; Liu, Y.; Kou, X. Recent advances in aflatoxin B1 detection based on nanotechnology and nanomaterials-A review. Anal. Chim. Acta. 2019, 1069, 1–27. [Google Scholar] [CrossRef]
  32. Goud, K.Y.; Reddy, K.K.; Satyanarayana, M.; Kummari, S.; Gobi, K.V. A review on recent developments in optical and electrochemical aptamer-based assays for mycotoxins using advanced nanomaterials. Mikrochim. Acta 2019, 187, 29. [Google Scholar] [CrossRef]
  33. Bhardwaj, H.; Sumana, G.; Marquette, C.A. Gold nanobipyramids integrated ultrasensitive optical and electrochemical biosensor for Aflatoxin B1 detection. Talanta 2021, 222, 121578. [Google Scholar] [CrossRef] [PubMed]
  34. Danesh, N.M.; Bostan, H.B.; Abnous, K.; Ramezani, M.; Youssefi, K.; Taghdisi, S.M.; Karimi, G. Ultrasensitive detection of aflatoxin B1 and its major metabolite aflatoxin M1 using aptasensors: A review. TrAC Trends Anal. Chem. 2018, 99, 117–128. [Google Scholar] [CrossRef]
  35. Castillo, G.; Poturnayová, A.; Šnejdárková, M.; Hianik, T.; Spinella, K.; Mosiello, L. Development of electrochemical aptasensor using dendrimers as an immobilization platform for detection of Aflatoxin B1 in food samples. In Proceedings of the 2015 XVIII AISEM Annual Conference, Trento, Italy, 3–5 February 2015; pp. 1–4. [Google Scholar]
  36. Zheng, M.Z.; Richard, J.L.; Binder, J. A review of rapid methods for the analysis of mycotoxins. Mycopathologia 2006, 161, 261–273. [Google Scholar] [CrossRef]
  37. Bennett, J.W.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516. [Google Scholar] [CrossRef] [PubMed]
  38. Robbins, C.A.; Swenson, L.J.; Nealley, M.L.; Gots, R.E.; Kelman, B.J. Health effects of mycotoxins in indoor air: A critical review. Appl. Occup. Environ. Hyg. 2000, 15, 773–784. [Google Scholar] [CrossRef] [PubMed]
  39. Catanante, G.; Rhouati, A.; Hayat, A.; Marty, J.L. An Overview of Recent Electrochemical Immunosensing Strategies for Mycotoxins Detection. Electroanalysis 2016, 28, 1750–1763. [Google Scholar] [CrossRef]
  40. Yao, H.; Hruska, Z.; Di Mavungu, J.D. Developments in detection and determination of aflatoxins. World Mycotoxin J. 2015, 8, 181–191. [Google Scholar] [CrossRef]
  41. Liu, D.; Li, W.; Zhu, C.; Li, Y.; Shen, X.; Li, L.; Yan, X.; You, T. Recent progress on electrochemical biosensing of aflatoxins: A review. TrAC Trends Anal. Chem. 2020, 133, 115966. [Google Scholar] [CrossRef]
  42. Hui, Y.; Wang, B.; Ren, R.; Zhao, A.; Zhang, F.; Song, S.; He, Y. An electrochemical aptasensor based on DNA-AuNPs-HRP nanoprobes and exonuclease-assisted signal amplification for detection of aflatoxin B1. Food Control 2020, 109, 106902. [Google Scholar] [CrossRef]
  43. Luan, Y.; Chen, Z.; **e, G.; Chen, J.; Lu, A.; Li, C.; Fu, H.; Ma, Z.; Wang, J. Rapid Visual Detection of Aflatoxin B1 by Label-Free Aptasensor Using Unmodified Gold Nanoparticles. J. Nanosci. Nanotechnol. 2015, 15, 1357–1361. [Google Scholar] [CrossRef]
  44. Nguyen, B.H.; Tran, L.D.; Do, Q.P.; Nguyen, H.L.; Tran, N.H.; Nguyen, P.X. Label-free detection of aflatoxin M1 with electrochemical Fe3O4/polyaniline-based aptasensor. Mater. Sci. Eng. C Mater. Biol. Appl. 2013, 33, 2229–2234. [Google Scholar] [CrossRef]
  45. Taghdisi, S.M.; Danesh, N.M.; Ramezani, M.; Abnous, K. A new amplified fluorescent aptasensor based on hairpin structure of G-quadruplex oligonucleotide-Aptamer chimera and silica nanoparticles for sensitive detection of aflatoxin B1 in the grape juice. Food Chem. 2018, 268, 342–346. [Google Scholar] [CrossRef]
  46. Henry, S.H.; Bosch, F.X.; Troxell, T.C.; Bolger, P.M. Reducing liver cancer--global control of aflatoxin. Science 1999, 286, 2453–2454. [Google Scholar] [CrossRef] [PubMed]
  47. Mao, J.; He, B.; Zhang, L.; Li, P.; Zhang, Q.; Ding, X.; Zhang, W. A Structure Identification and Toxicity Assessment of the Degradation Products of Aflatoxin B(1) in Peanut Oil under UV Irradiation. Toxins 2016, 8, 332. [Google Scholar] [CrossRef]
  48. Mousavi Khaneghah, A.; Eş, I.; Raeisi, S.; Fakhri, Y. Aflatoxins in cereals: State of the art. J. Food Saf. 2018, 38, 12532. [Google Scholar] [CrossRef]
  49. Qureshi, H. Is Aflatoxin B1 A Biomarker for Pathogenic Potential of Aspergillus flavus? J. Cell Sci. 2014, 5, 1000188. [Google Scholar] [CrossRef]
  50. Ali, N. Aflatoxins in rice: Worldwide occurrence and public health perspectives. Toxicol. Rep. 2019, 6, 1188–1197. [Google Scholar] [CrossRef]
  51. Sun, C.; Liao, X.; Jia, B.; Shi, L.; Zhang, D.; Wang, R.; Zhou, L.; Kong, W. Development of a ZnCdS@ZnS quantum dots-based label-free electrochemiluminescence immunosensor for sensitive determination of aflatoxin B1 in lotus seed. Mikrochim. Acta 2020, 187, 236. [Google Scholar] [CrossRef]
  52. Bhardwaj, H.; Sumana, G.; Marquette, C.A. A label-free ultrasensitive microfluidic surface Plasmon resonance biosensor for Aflatoxin B1 detection using nanoparticles integrated gold chip. Food Chem. 2020, 307, 125530. [Google Scholar] [CrossRef]
  53. Ramalho, L.N.Z.; Porta, L.D.; Rosim, R.E.; Petta, T.; Augusto, M.J.; Silva, D.M.; Ramalho, F.S.; Oliveira, C.A.F. Aflatoxin B1 residues in human livers and their relationship with markers of hepatic carcinogenesis in Sao Paulo, Brazil. Toxicol. Rep. 2018, 5, 777–784. [Google Scholar] [CrossRef]
  54. Kensler, T.W.; Roebuck, B.D.; Wogan, G.N.; Groopman, J.D. Aflatoxin: A 50-year odyssey of mechanistic and translational toxicology. Toxicol. Sci. 2011, 120 (Suppl. 1), S28–S48. [Google Scholar] [CrossRef] [PubMed]
  55. Rushing, B.R.; Selim, M.I. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food Chem. Toxicol. 2019, 124, 81–100. [Google Scholar] [CrossRef] [PubMed]
  56. Chu, Y.J.; Yang, H.I.; Wu, H.C.; Liu, J.; Wang, L.Y.; Lu, S.N.; Lee, M.H.; Jen, C.L.; You, S.L.; Santella, R.M.; et al. Aflatoxin B1 exposure increases the risk of cirrhosis and hepatocellular carcinoma in chronic hepatitis B virus carriers. Int. J. Cancer 2017, 141, 711–720. [Google Scholar] [CrossRef] [PubMed]
  57. Marin, S.; Ramos, A.J.; Cano-Sancho, G.; Sanchis, V. Mycotoxins: Occurrence, toxicology, and exposure assessment. Food Chem. Toxicol. 2013, 60, 218–237. [Google Scholar] [CrossRef]
  58. Wild, C.P.; Turner, P.C. The toxicology of aflatoxins as a basis for public health decisions. Mutagenesis 2002, 17, 471–481. [Google Scholar] [CrossRef]
  59. Zhou, Q.; Tang, D. Recent advances in photoelectrochemical biosensors for analysis of mycotoxins in food. TrAC Trends Anal. Chem. 2020, 124, 115814. [Google Scholar] [CrossRef]
  60. Eivazzadeh-Keihan, R.; Pashazadeh, P.; Hejazi, M.; de la Guardia, M.; Mokhtarzadeh, A. Recent advances in Nanomaterial-mediated Bio and immune sensors for detection of aflatoxin in food products. TrAC Trends Anal. Chem. 2017, 87, 112–128. [Google Scholar] [CrossRef]
  61. Keenan, J.; Jolly, P.; Preko, P.; Baidoo, J.; Wang, J.S.; Phillips, T.D.; Williams, J.H.; McGwin, G., Jr. Association Between Aflatoxin B1 Albumin Adduct Levels and Tuberculosis Infection Among HIV+ Ghanaians. Arch. Clin. Microbiol. 2011, 2, 3. [Google Scholar]
  62. Williams, R.; Airey, M.; Baxter, H.; Forrester, J.; Kennedy-Martin, T.; Girach, A. Epidemiology of diabetic retinopathy and macular oedema: A systematic review. Eye 2004, 18, 963–983. [Google Scholar] [CrossRef]
  63. Gong, Y.; Hounsa, A.; Egal, S.; Turner, P.C.; Sutcliffe, A.E.; Hall, A.J.; Cardwell, K.; Wild, C.P. Postweaning exposure to aflatoxin results in impaired child growth: A longitudinal study in Benin, West Africa. Environ. Health Persp. 2004, 112, 1334–1338. [Google Scholar] [CrossRef]
  64. Abdolmaleki, K.; Khedri, S.; Alizadeh, L.; Javanmardi, F.; Oliveira, C.A.F.; Mousavi Khaneghah, A. The mycotoxins in edible oils: An overview of prevalence, concentration, toxicity, detection and decontamination techniques. Trends Food Sci. Technol. 2021, 115, 500–511. [Google Scholar] [CrossRef]
  65. Claeys, L.; Romano, C.; De Ruyck, K.; Wilson, H.; Fervers, B.; Korenjak, M.; Zavadil, J.; Gunter, M.J.; De Saeger, S.; De Boevre, M.; et al. Mycotoxin exposure and human cancer risk: A systematic review of epidemiological studies. Compr. Rev. Food Sci. Food Saf. 2020, 19, 1449–1464. [Google Scholar] [CrossRef] [PubMed]
  66. Adeyeye, S.A.O.; Yildiz, F. Fungal mycotoxins in foods: A review. Cogent Food Agric. 2016, 2, 1213127. [Google Scholar] [CrossRef]
  67. Selamat, J.; Iqbal, S.Z. Food Safety: Basic Concepts, Recent Issues, and Future Challenges; Springer: Berlin/Heidelberg, Germany, 2016. [Google Scholar]
  68. Directorate-General for Health and Consumers. The Rapid Alert System for Food and Feed (RASFF): Annual Report 2005; Publications Office: Luxembourg, 2006. [Google Scholar]
  69. Papachristou, A.; Markaki, P. Determination of ochratoxin A in virgin olive oils of Greek origin by immunoaffinity column clean-up and high-performance liquid chromatography. Food Addit. Contam. 2004, 21, 85–92. [Google Scholar] [CrossRef] [PubMed]
  70. Bhat, R.; Reddy, K.R. Challenges and issues concerning mycotoxins contamination in oil seeds and their edible oils: Updates from last decade. Food Chem. 2017, 215, 425–437. [Google Scholar] [CrossRef] [PubMed]
  71. Mazumder, P.M.; Sasmal, D. Mycotoxins–limits and regulations. Anc. Sci. Life 2001, 20, 1. [Google Scholar]
  72. Zinedine, A.; Mañes, J. Occurrence and legislation of mycotoxins in food and feed from Morocco. Food Control 2009, 20, 334–344. [Google Scholar] [CrossRef]
  73. Selvaraj, J.N.; Wang, Y.; Zhou, L.; Zhao, Y.; **ng, F.; Dai, X.; Liu, Y. Recent mycotoxin survey data and advanced mycotoxin detection techniques reported from China: A review. Food Addit. Contam. Part A 2015, 32, 440–452. [Google Scholar] [CrossRef]
  74. Anukul, N.; Vangnai, K.; Mahakarnchanakul, W. Significance of regulation limits in mycotoxin contamination in Asia and risk management programs at the national level. J. Food Drug Anal. 2013, 21, 227–241. [Google Scholar] [CrossRef]
  75. Ji, F.; He, D.; Olaniran, A.O.; Mokoena, M.P.; Xu, J.; Shi, J. Occurrence, toxicity, production and detection of Fusarium mycotoxin: A review. Food Prod. Process. Nutr. 2019, 1, 1–14. [Google Scholar] [CrossRef]
  76. Aiko, V.; Mehta, A. Occurrence, detection and detoxification of mycotoxins. J. Biosci. 2015, 40, 943–954. [Google Scholar] [CrossRef] [PubMed]
  77. Le, V.T.; Vasseghian, Y.; Dragoi, E.N.; Moradi, M.; Mousavi Khaneghah, A. A review on graphene-based electrochemical sensor for mycotoxins detection. Food Chem. Toxicol. 2021, 148, 111931. [Google Scholar] [CrossRef]
  78. Turner, N.W.; Bramhmbhatt, H.; Szabo-Vezse, M.; Poma, A.; Coker, R.; Piletsky, S.A. Analytical methods for determination of mycotoxins: An update (2009–2014). Anal. Chim. Acta. 2015, 901, 12–33. [Google Scholar] [CrossRef]
  79. Ingenbleek, L.; Sulyok, M.; Adegboye, A.; Hossou, S.E.; Kone, A.Z.; Oyedele, A.D.; Kisito, C.; Dembele, Y.K.; Eyangoh, S.; Verger, P.; et al. Regional Sub-Saharan Africa Total Diet Study in Benin, Cameroon, Mali and Nigeria Reveals the Presence of 164 Mycotoxins and Other Secondary Metabolites in Foods. Toxins 2019, 11, 54. [Google Scholar] [CrossRef] [PubMed]
  80. Zhu, C.; Liu, D.; Li, Y.; Shen, X.; Ma, S.; Liu, Y.; You, T. Ratiometric electrochemical aptasensor for ultrasensitive detection of Ochratoxin A based on a dual signal amplification strategy: Engineering the binding of methylene blue to DNA. Biosens. Bioelectron. 2020, 150, 111814. [Google Scholar] [CrossRef] [PubMed]
  81. Karunarathna, N.B.; Fernando, C.J.; Munasinghe, D.M.S.; Fernando, R. Occurrence of aflatoxins in edible vegetable oils in Sri Lanka. Food Control 2019, 101, 97–103. [Google Scholar] [CrossRef]
  82. Wang, N.; Duan, C.; Geng, X.; Li, S.; Ding, K.; Guan, Y. One step rapid dispersive liquid-liquid micro-extraction with in-situ derivatization for determination of aflatoxins in vegetable oils based on high performance liquid chromatography fluorescence detection. Food Chem. 2019, 287, 333–337. [Google Scholar] [CrossRef]
  83. Nabizadeh, S.; Shariatifar, N.; Shokoohi, E.; Shoeibi, S.; Gavahian, M.; Fakhri, Y.; Azari, A.; Mousavi Khaneghah, A. Prevalence and probabilistic health risk assessment of aflatoxins B1, B2, G1, and G2 in Iranian edible oils. Environ. Sci. Pollut. Res. Int. 2018, 25, 35562–35570. [Google Scholar] [CrossRef]
  84. Ma, F.; Chen, R.; Li, P.; Zhang, Q.; Zhang, W.; Hu, X. Preparation of an immunoaffinity column with amino-silica gel microparticles and its application in sample cleanup for aflatoxin detection in agri-products. Molecules 2013, 18, 2222–2235. [Google Scholar] [CrossRef]
  85. Afzali, D.; Ghanbarian, M.; Mostafavi, A.; Shamspur, T.; Ghaseminezhad, S. A novel method for high preconcentration of ultra trace amounts of B(1), B(2), G(1) and G(2) aflatoxins in edible oils by dispersive liquid-liquid microextraction after immunoaffinity column clean-up. J. Chromatogr. A 2012, 1247, 35–41. [Google Scholar] [CrossRef]
  86. Elzupir, A.O.; Suliman, M.A.; Ibrahim, I.A.; Fadul, M.H.; Elhussein, A.M. Aflatoxins levels in vegetable oils in Khartoum State, Sudan. Mycotoxin Res. 2010, 26, 69–73. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, K.; Xu, D. Application of Stable Isotope Dilution and Liquid Chromatography Tandem Mass Spectrometry for Multi-Mycotoxin Analysis in Edible Oils. J. AOAC Int. 2019, 102, 1651–1656. [Google Scholar] [CrossRef] [PubMed]
  88. Eom, T.; Cho, H.D.; Kim, J.; Park, M.; An, J.; Kim, M.; Kim, S.H.; Han, S.B. Multiclass mycotoxin analysis in edible oils using a simple solvent extraction method and liquid chromatography with tandem mass spectrometry. Food Addit. Contam. Part A 2017, 34, 2011–2022. [Google Scholar] [CrossRef] [PubMed]
  89. Huang, S.; Chen, X.; Wang, Y.; Zhu, F.; Jiang, R.; Ouyang, G. High enrichment and ultra-trace analysis of aflatoxins in edible oils by a modified hollow-fiber liquid-phase microextraction technique. Chem. Commun. 2017, 53, 8988–8991. [Google Scholar] [CrossRef] [PubMed]
  90. Sharmili, K.; **ap, S.; Sukor, R. Development, optimization and validation of QuEChERS based liquid chromatography tandem mass spectrometry method for determination of multimycotoxin in vegetable oil. Food Control 2016, 70, 152–160. [Google Scholar] [CrossRef]
  91. Pereira, V.L.; Fernandes, J.O.; Cunha, S.C. Comparative assessment of three cleanup procedures after QuEChERS extraction for determination of trichothecenes (type A and type B) in processed cereal-based baby foods by GC-MS. Food Chem. 2015, 182, 143–149. [Google Scholar] [CrossRef] [PubMed]
  92. Nathanail, A.V.; Syvahuoko, J.; Malachova, A.; Jestoi, M.; Varga, E.; Michlmayr, H.; Adam, G.; Sievilainen, E.; Berthiller, F.; Peltonen, K. Simultaneous determination of major type A and B trichothecenes, zearalenone and certain modified metabolites in Finnish cereal grains with a novel liquid chromatography-tandem mass spectrometric method. Anal. Bioanal. Chem. 2015, 407, 4745–4755. [Google Scholar] [CrossRef]
  93. Capriotti, A.L.; Cavaliere, C.; Foglia, P.; Samperi, R.; Stampachiacchiere, S.; Ventura, S.; Lagana, A. Multiclass analysis of mycotoxins in biscuits by high performance liquid chromatography-tandem mass spectrometry. Comparison of different extraction procedures. J. Chromatogr. A 2014, 1343, 69–78. [Google Scholar] [CrossRef]
  94. Malachova, A.; Sulyok, M.; Beltran, E.; Berthiller, F.; Krska, R. Optimization and validation of a quantitative liquid chromatography-tandem mass spectrometric method covering 295 bacterial and fungal metabolites including all regulated mycotoxins in four model food matrices. J. Chromatogr. A 2014, 1362, 145–156. [Google Scholar] [CrossRef]
  95. Tsiplakou, E.; Anagnostopoulos, C.; Liapis, K.; Haroutounian, S.A.; Zervas, G. Determination of mycotoxins in feedstuffs and ruminant’s milk using an easy and simple LC-MS/MS multiresidue method. Talanta 2014, 130, 8–19. [Google Scholar] [CrossRef]
  96. Yang, L.X.; Liu, Y.P.; Miao, H.; Dong, B.; Yang, N.J.; Chang, F.Q.; Yang, L.X.; Sun, J.B. Determination of aflatoxins in edible oil from markets in Hebei Province of China by liquid chromatography-tandem mass spectrometry. Food Addit. Contam. Part B 2011, 4, 244–247. [Google Scholar] [CrossRef] [PubMed]
  97. Bao, L.; Trucksess, M.W.; White, K.D. Determination of aflatoxins B1, B2, G1, and G2 in olive oil, peanut oil, and sesame oil. J. AOAC Int. 2010, 93, 936–942. [Google Scholar] [CrossRef] [PubMed]
  98. Junsai, T.; Poapolathep, S.; Sutjarit, S.; Giorgi, M.; Zhang, Z.; Logrieco, A.F.; Li, P.; Poapolathep, A. Determination of Multiple Mycotoxins and Their Natural Occurrence in Edible Vegetable Oils Using Liquid Chromatography-Tandem Mass Spectrometry. Foods 2021, 10, 2795. [Google Scholar] [CrossRef]
  99. Deng, H.; Su, X.; Wang, H. Simultaneous Determination of Aflatoxin B1, Bisphenol A, and 4-Nonylphenol in Peanut Oils by Liquid-Liquid Extraction Combined with Solid-Phase Extraction and Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry. Food Anal. Methods 2017, 11, 1303–1311. [Google Scholar] [CrossRef]
  100. Hidalgo-Ruiz, J.L.; Romero-Gonzalez, R.; Martinez Vidal, J.L.; Garrido Frenich, A. A rapid method for the determination of mycotoxins in edible vegetable oils by ultra-high performance liquid chromatography-tandem mass spectrometry. Food Chem. 2019, 288, 22–28. [Google Scholar] [CrossRef]
  101. Zhu, Z.; Feng, M.; Zuo, L.; Zhu, Z.; Wang, F.; Chen, L.; Li, J.; Shan, G.; Luo, S.Z. An aptamer based surface plasmon resonance biosensor for the detection of ochratoxin A in wine and peanut oil. Biosens. Bioelectron. 2015, 65, 320–326. [Google Scholar] [CrossRef]
  102. Idris, Y.M.; Mariod, A.A.; Elnour, I.A.; Mohamed, A.A. Determination of aflatoxin levels in Sudanese edible oils. Food Chem. Toxicol. 2010, 48, 2539–2541. [Google Scholar] [CrossRef]
  103. Majerus, P.; Graf, N.; Kramer, M. Rapid determination of zearalenone in edible oils by HPLC with fluorescence detection. Mycotoxin Res. 2009, 25, 117–121. [Google Scholar] [CrossRef]
  104. Wang, H.; Zhao, L.; Yang, H.; Guo, Q.; Shi, H.; Pan, H.; Zhao, L.; Qian, C. Determination of benzo(a)pyrene and aflatoxins (B1, B2, G1, G2) in vegetable oil by GPC-HPLC-FLD. Anal. Methods 2014, 6, 1545–1549. [Google Scholar] [CrossRef]
  105. **e, J.; Peng, T.; He, J.L.; Shao, Y.; Fan, C.L.; Chen, Y.; Jiang, W.X.; Chen, M.; Wang, Q.; Pei, X.Y.; et al. Preparation and characterization of an immunoaffinity column for the selective extraction of aflatoxin B1 in 13 kinds of foodstuffs. J. Chromatogr. B 2015, 998–999, 50–56. [Google Scholar] [CrossRef]
  106. Han, X.; Xu, W.; Zhang, J.; Xu, J.; Li, F. Co-Occurrence of Beauvericin and Enniatins in Edible Vegetable Oil Samples, China. Toxins 2019, 11, 100. [Google Scholar] [CrossRef] [Green Version]
  107. Drzymala, S.S.; Weiz, S.; Heinze, J.; Marten, S.; Prinz, C.; Zimathies, A.; Garbe, L.A.; Koch, M. Automated solid-phase extraction coupled online with HPLC-FLD for the quantification of zearalenone in edible oil. Anal. Bioanal. Chem. 2015, 407, 3489–3497. [Google Scholar] [CrossRef] [PubMed]
  108. Yu, X.; Yang, H. Pyrethroid residue determination in organic and conventional vegetables using liquid-solid extraction coupled with magnetic solid phase extraction based on polystyrene-coated magnetic nanoparticles. Food Chem. 2017, 217, 303–310. [Google Scholar] [CrossRef] [PubMed]
  109. Sheng, J.; Zuo, J.; Liu, K.; Ma, L.; Li, C.; Li, Y.; Kong, D. Highly selective enrichment of aflatoxin B1 from edible oil using polydopamine-modified magnetic nanomaterials. Food Sci. Technol. 2021, 41, 321–327. [Google Scholar] [CrossRef]
  110. Ferracane, R.; Tafuri, A.; Logieco, A.; Galvano, F.; Balzano, D.; Ritieni, A. Simultaneous determination of aflatoxin B1 and ochratoxin A and their natural occurrence in Mediterranean virgin olive oil. Food Addit. Contam. 2007, 24, 173–180. [Google Scholar] [CrossRef]
  111. Giménez, I.; Herrera, M.; Escobar, J.; Ferruz, E.; Lorán, S.; Herrera, A.; Ariño, A. Distribution of deoxynivalenol and zearalenone in milled germ during wheat milling and analysis of toxin levels in wheat germ and wheat germ oil. Food Control 2013, 34, 268–273. [Google Scholar] [CrossRef]
  112. Jiang, Y.; Li, Y.; Jiang, Y.; Li, J.; Pan, C. Determination of multiresidues in rapeseed, rapeseed oil, and rapeseed meal by acetonitrile extraction, low-temperature cleanup, and detection by liquid chromatography with tandem mass spectrometry. J. Agric. Food Chem. 2012, 60, 5089–5098. [Google Scholar] [CrossRef]
  113. Payanan, T.; Leepipatpiboon, N.; Varanusupakul, P. Low-temperature cleanup with solid-phase extraction for the determination of polycyclic aromatic hydrocarbons in edible oils by reversed phase liquid chromatography with fluorescence detection. Food Chem. 2013, 141, 2720–2726. [Google Scholar] [CrossRef]
  114. Urusov, A.E.; Petrakova, A.V.; Vozniak, M.V.; Zherdev, A.V.; Dzantiev, B.B. Rapid immunoenzyme assay of aflatoxin B1 using magnetic nanoparticles. Sensors 2014, 14, 21843–21857. [Google Scholar] [CrossRef]
  115. Yu, X.; Li, Z.; Zhao, M.; Lau, S.C.S.; Ru Tan, H.; Teh, W.J.; Yang, H.; Zheng, C.; Zhang, Y. Quantification of aflatoxin B1 in vegetable oils using low temperature clean-up followed by immuno-magnetic solid phase extraction. Food Chem. 2019, 275, 390–396. [Google Scholar] [CrossRef]
  116. Zhao, H.; Chen, X.; Shen, C.; Qu, B. Determination of 16 mycotoxins in vegetable oils using a QuEChERS method combined with high-performance liquid chromatography-tandem mass spectrometry. Food Addit. Contam. Part A 2017, 34, 255–264. [Google Scholar] [CrossRef] [PubMed]
  117. Qian, M.; Zhang, H.; Wu, L.; **, N.; Wang, J.; Jiang, K. Simultaneous determination of zearalenone and its derivatives in edible vegetable oil by gel permeation chromatography and gas chromatography-triple quadrupole mass spectrometry. Food Chem. 2015, 166, 23–28. [Google Scholar] [CrossRef] [PubMed]
  118. Alshannaq, A.; Yu, J.H. Occurrence, Toxicity, and Analysis of Major Mycotoxins in Food. Int. J. Environ. Res. Public. Health 2017, 14, 632. [Google Scholar] [CrossRef] [PubMed]
  119. Santos, A.; Vaz, A.; Rodrigues, P.; Veloso, A.; Venâncio, A.; Peres, A. Thin Films Sensor Devices for Mycotoxins Detection in Foods: Applications and Challenges. Chemosensors 2019, 7, 3. [Google Scholar] [CrossRef]
  120. Shephard, G.S. Current Status of Mycotoxin Analysis: A Critical Review. J. AOAC Int. 2016, 99, 842–848. [Google Scholar] [CrossRef] [PubMed]
  121. Schwack, W.; Pellissier, E.; Morlock, G. Analysis of unauthorized Sudan dyes in food by high-performance thin-layer chromatography. Anal. Bioanal. Chem. 2018, 410, 5641–5651. [Google Scholar] [CrossRef]
  122. Gauthier, M.S.; O’Brien, E.L.; Bigornia, S.; Mott, M.; Cacicedo, J.M.; Xu, X.J.; Gokce, N.; Apovian, C.; Ruderman, N. Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochem. Biophys. Res. Commun. 2011, 404, 382–387. [Google Scholar] [CrossRef]
  123. Sun, Y.; Wang, H.; Wang, W.; Hu, B.; Zhou, L.; Ye, H.; Zeng, X. Changes in molecular structure of chickpea starch during processing treatments: A thin layer chromatography study. Food Chem. 2018, 243, 186–191. [Google Scholar] [CrossRef]
  124. Villani, T.S.; Reichert, W.; Ferruzzi, M.G.; Pasinetti, G.M.; Simon, J.E.; Wu, Q. Chemical investigation of commercial grape seed derived products to assess quality and detect adulteration. Food Chem. 2015, 170, 271–280. [Google Scholar] [CrossRef]
  125. Wacoo, A.P.; Wendiro, D.; Vuzi, P.C.; Hawumba, J.F. Methods for Detection of Aflatoxins in Agricultural Food Crops. J. Appl. Chem. 2014, 2014, 706291. [Google Scholar] [CrossRef]
  126. Cabezudo, I.; Salazar, M.O.; Ramallo, I.A.; Furlan, R.L.E. Effect-directed analysis in food by thin-layer chromatography assays. Food Chem. 2022, 390, 132937. [Google Scholar] [CrossRef] [PubMed]
  127. Lin, L.; Zhang, J.; Wang, P.; Wang, Y.; Chen, J. Thin-layer chromatography of mycotoxins and comparison with other chromatographic methods. J. Chromatogr. A 1998, 815, 3–20. [Google Scholar] [CrossRef]
  128. Sereshti, H.; Poursorkh, Z.; Aliakbarzadeh, G.; Zarre, S. Quality control of saffron and evaluation of potential adulteration by means of thin layer chromatography-image analysis and chemometrics methods. Food Control 2018, 90, 48–57. [Google Scholar] [CrossRef]
  129. Li, Y.; Zhao, C.; Lu, C.; Zhou, S.; Tian, G.; He, L.; Bao, Y.; Fauconnier, M.L.; **ao, H.; Zheng, J. Simultaneous determination of 14 bioactive citrus flavonoids using thin-layer chromatography combined with surface enhanced Raman spectroscopy. Food Chem. 2021, 338, 128115. [Google Scholar] [CrossRef] [PubMed]
  130. Morlock, G.E.; Klingelhofer, I. Liquid chromatography-bioassay-mass spectrometry for profiling of physiologically active food. Anal. Chem. 2014, 86, 8289–8295. [Google Scholar] [CrossRef] [PubMed]
  131. Tao, F.; Yao, H.; Hruska, Z.; Burger, L.W.; Rajasekaran, K.; Bhatnagar, D. Recent development of optical methods in rapid and non-destructive detection of aflatoxin and fungal contamination in agricultural products. TrAC Trends Anal. Chem. 2018, 100, 65–81. [Google Scholar] [CrossRef]
  132. Wu, Q.; **e, L.; Xu, H. Determination of toxigenic fungi and aflatoxins in nuts and dried fruits using imaging and spectroscopic techniques. Food Chem. 2018, 252, 228–242. [Google Scholar] [CrossRef]
  133. Yadav, N.; Yadav, S.S.; Chhillar, A.K.; Rana, J.S. An overview of nanomaterial based biosensors for detection of Aflatoxin B1 toxicity in foods. Food Chem. Toxicol. 2021, 152, 112201. [Google Scholar] [CrossRef]
  134. Karoui, R.; Blecker, C. Fluorescence Spectroscopy Measurement for Quality Assessment of Food Systems—A Review. Food Bioprocess Technol. 2010, 4, 364–386. [Google Scholar] [CrossRef]
  135. Ma, H.; Ran, C.; Li, M.; Gao, J.; Wang, X.; Zhang, L.; Bian, J.; Li, J.; Jiang, Y. Graphene oxide-coated stir bar sorptive extraction of trace aflatoxins from soy milk followed by high performance liquid chromatography-laser-induced fluorescence detection. Food Addit. Contam. Part A 2018, 35, 772–781. [Google Scholar] [CrossRef]
  136. Hruska, Z.; Yao, H.; Kincaid, R.; Brown, R.; Cleveland, T.; Bhatnagar, D. Fluorescence Excitation–Emission Features of Aflatoxin and Related Secondary Metabolites and Their Application for Rapid Detection of Mycotoxins. Food Bioprocess Technol. 2014, 7, 1195–1201. [Google Scholar] [CrossRef]
  137. Chen, M.; He, X.; Pang, Y.; Shen, F.; Fang, Y.; Hu, Q. Laser induced fluorescence spectroscopy for detection of Aflatoxin B1 contamination in peanut oil. J. Food Meas. Charact. 2021, 15, 2231–2239. [Google Scholar] [CrossRef]
  138. Qu, J.H.; Liu, D.; Cheng, J.H.; Sun, D.W.; Ma, J.; Pu, H.; Zeng, X.A. Applications of near-infrared spectroscopy in food safety evaluation and control: A review of recent research advances. Crit. Rev. Food Sci. Nutr. 2015, 55, 1939–1954. [Google Scholar] [CrossRef] [PubMed]
  139. Rohman, A. Infrared spectroscopy for quantitative analysis and oil parameters of olive oil and virgin coconut oil: A review. Int. J. Food Prop. 2016, 20, 1447–1456. [Google Scholar] [CrossRef]
  140. Nicolaï, B.M.; Beullens, K.; Bobelyn, E.; Peirs, A.; Saeys, W.; Theron, K.I.; Lammertyn, J. Nondestructive measurement of fruit and vegetable quality by means of NIR spectroscopy: A review. Postharvest Biol. Technol. 2007, 46, 99–118. [Google Scholar] [CrossRef]
  141. Uddin, M.; Okazaki, E. Classification of Fresh and Frozen-thawed Fish by Near-infrared Spectroscopy. J. Food Sci. 2004, 69, C665–C668. [Google Scholar] [CrossRef]
  142. Kurz, C.; Leitenberger, M.; Carle, R.; Schieber, A. Evaluation of fruit authenticity and determination of the fruit content of fruit products using FT-NIR spectroscopy of cell wall components. Food Chem. 2010, 119, 806–812. [Google Scholar] [CrossRef]
  143. Kuligowski, J.; Carrión, D.; Quintás, G.; Garrigues, S.; de la Guardia, M. Direct determination of polymerised triacylglycerides in deep-frying vegetable oil by near infrared spectroscopy using Partial Least Squares regression. Food Chem. 2012, 131, 353–359. [Google Scholar] [CrossRef]
  144. Balabin, R.M.; Smirnov, S.V. Melamine detection by mid- and near-infrared (MIR/NIR) spectroscopy: A quick and sensitive method for dairy products analysis including liquid milk, infant formula, and milk powder. Talanta 2011, 85, 562–568. [Google Scholar] [CrossRef]
  145. Ravikanth, L.; Jayas, D.S.; White, N.D.G.; Fields, P.G.; Sun, D.-W. Extraction of Spectral Information from Hyperspectral Data and Application of Hyperspectral Imaging for Food and Agricultural Products. Food Bioprocess Technol. 2016, 10, 1–33. [Google Scholar] [CrossRef]
  146. Morsy, N.; Sun, D.W. Robust linear and non-linear models of NIR spectroscopy for detection and quantification of adulterants in fresh and frozen-thawed minced beef. Meat Sci. 2013, 93, 292–302. [Google Scholar] [CrossRef] [PubMed]
  147. Luo, J.; Liu, T.; Liu, Y. FT-NIR and Confocal Microscope Raman Spectroscopic Studies of Sesame Oil Adulteration. In Proceedings of the Computer and Computing Technologies in Agriculture V, Berlin, Heidelberg, 29 October 2011; pp. 24–31. [Google Scholar]
  148. Mignani, A.G.; Ciaccheri, L.; Ottevaere, H.; Thienpont, H.; Conte, L.; Marega, M.; Cichelli, A.; Attilio, C.; Cimato, A. Visible and near-infrared absorption spectroscopy by an integrating sphere and optical fibers for quantifying and discriminating the adulteration of extra virgin olive oil from Tuscany. Anal. Bioanal. Chem. 2011, 399, 1315–1324. [Google Scholar] [CrossRef] [PubMed]
  149. Sinelli, N.; Cerretani, L.; Egidio, V.D.; Bendini, A.; Casiraghi, E. Application of near (NIR) infrared and mid (MIR) infrared spectroscopy as a rapid tool to classify extra virgin olive oil on the basis of fruity attribute intensity. Food Res. Int. 2010, 43, 369–375. [Google Scholar] [CrossRef]
  150. Luna, A.S.; da Silva, A.P.; Pinho, J.S.; Ferre, J.; Boque, R. Rapid characterization of transgenic and non-transgenic soybean oils by chemometric methods using NIR spectroscopy. Spectrochim. Acta Part A 2013, 100, 115–119. [Google Scholar] [CrossRef]
  151. Yao, W.; Liu, R.; Xu, Z.; Zhang, Y.; Deng, Y.; Guo, H.; Cozzolino, D. Rapid Determination of Aflatoxin B1 Contamination in Peanut Oil by Fourier Transform Near-Infrared Spectroscopy. J. Spectro. 2022, 2022, 1–9. [Google Scholar] [CrossRef]
  152. Song, H.; Li, F.; Guang, P.; Yang, X.; Pan, H.; Huang, F. Detection of Aflatoxin B1 in Peanut Oil Using Attenuated Total Reflection Fourier Transform Infrared Spectroscopy Combined with Partial Least Squares Discriminant Analysis and Support Vector Machine Models. J. Food Prot. 2021, 84, 1315–1320. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, Y.; Zhang, Y.; He, C.; **e, M.; Luo, H.; Wang, Y.; Zhang, J. Rapid screen of aflatoxin-contaminated peanut oil using Fourier transform infrared spectroscopy combined with multivariate decision tree. Int. J. Food Sci. Technol. 2018, 53, 2386–2393. [Google Scholar] [CrossRef]
  154. Rawson, A.; Sunil, C.K. Recent Advances in Terahertz Time-Domain Spectroscopy and Imaging Techniques for Automation in Agriculture and Food Sector. Food Anal. Methods 2021, 15, 498–526. [Google Scholar] [CrossRef]
  155. Jan, A.; Pandiselvam, R.; Kothakota, A.; Sruthi, N.; Ramesh, S. Terahertz Spectroscopy Imaging Technique: Non-Destructive Tool For Evaluation Of Quality And Safety Of Food Products. In Handbook of Research on Food Processing and Preservation Technologies; Apple Academic Press: New York, NY, USA, 2021; pp. 141–157. [Google Scholar]
  156. Feng, C.H.; Otani, C. Terahertz spectroscopy technology as an innovative technique for food: Current state-of-the-Art research advances. Crit. Rev. Food Sci. Nutr. 2021, 61, 2523–2543. [Google Scholar] [CrossRef]
  157. Afsah-Hejri, L.; Hajeb, P.; Ara, P.; Ehsani, R.J. A Comprehensive Review on Food Applications of Terahertz Spectroscopy and Imaging. Compr. Rev. Food Sci. Food Saf. 2019, 18, 1563–1621. [Google Scholar] [CrossRef]
  158. Kawano, Y.; Ishibashi, K. An on-chip near-field terahertz probe and detector. Nat. Photonics 2008, 2, 618–621. [Google Scholar] [CrossRef]
  159. McIntosh, A.I.; Yang, B.; Goldup, S.M.; Watkinson, M.; Donnan, R.S. Terahertz spectroscopy: A powerful new tool for the chemical sciences? Chem. Soc. Rev. 2012, 41, 2072–2082. [Google Scholar] [CrossRef] [PubMed]
  160. Wang, K.; Sun, D.-W.; Pu, H. Emerging non-destructive terahertz spectroscopic imaging technique: Principle and applications in the agri-food industry. Trends Food Sci. Technol. 2017, 67, 93–105. [Google Scholar] [CrossRef]
  161. Chen, M.; Lijuan, X. A Preliminary Study of Aflatoxin B1 Detection in Peanut Oil by Terahertz Time-Domain Spectroscopy. Trans. ASABE 2014, 57, 1793–1799. [Google Scholar] [CrossRef]
  162. Liu, W.; Zhao, P.; Wu, C.; Liu, C.; Yang, J.; Zheng, L. Rapid determination of aflatoxin B1 concentration in soybean oil using terahertz spectroscopy with chemometric methods. Food Chem. 2019, 293, 213–219. [Google Scholar] [CrossRef]
  163. Wang, Q.; **e, L.; Ying, Y. Overview of imaging methods based on terahertz time-domain spectroscopy. Appl. Spectrosc. Rev. 2021, 57, 249–264. [Google Scholar] [CrossRef]
  164. Zhang, W.; Ma, J.; Sun, D.W. Raman spectroscopic techniques for detecting structure and quality of frozen foods: Principles and applications. Crit. Rev. Food Sci. Nutr. 2021, 61, 2623–2639. [Google Scholar] [CrossRef]
  165. Wu, Z.; Pu, H.; Sun, D.-W. Fingerprinting and tagging detection of mycotoxins in agri-food products by surface-enhanced Raman spectroscopy: Principles and recent applications. Trends Food Sci. Technol. 2021, 110, 393–404. [Google Scholar] [CrossRef]
  166. He, H.; Sun, D.W.; Pu, H.; Chen, L.; Lin, L. Applications of Raman spectroscopic techniques for quality and safety evaluation of milk: A review of recent developments. Crit. Rev. Food Sci. Nutr. 2019, 59, 770–793. [Google Scholar] [CrossRef]
  167. Deng, J.; Zhang, X.; Li, M.; Jiang, H.; Chen, Q. Feasibility study on Raman spectra-based deep learning models for monitoring the contamination degree and level of aflatoxin B1 in edible oil. Microchem. J. 2022, 180, 107613. [Google Scholar] [CrossRef]
  168. Zhu, C.; Jiang, H.; Chen, Q. High Precisive Prediction of Aflatoxin B1 in Pressing Peanut Oil Using Raman Spectra Combined with Multivariate Data Analysis. Foods 2022, 11, 1565. [Google Scholar] [CrossRef] [PubMed]
  169. Yang, M.; Liu, G.; Mehedi, H.M.; Ouyang, Q.; Chen, Q. A universal SERS aptasensor based on DTNB labeled GNTs/Ag core-shell nanotriangle and CS-Fe3O4 magnetic-bead trace detection of Aflatoxin B1. Anal. Chim. Acta. 2017, 986, 122–130. [Google Scholar] [CrossRef] [PubMed]
  170. Chen, Q.; Yang, M.; Yang, X.; Li, H.; Guo, Z.; Rahma, M. A large Raman scattering cross-section molecular embedded SERS aptasensor for ultrasensitive Aflatoxin B1 detection using CS-Fe3O4 for signal enrichment. Spectrochim. Acta Part A 2018, 189, 147–153. [Google Scholar] [CrossRef] [PubMed]
  171. Guo, M.; Hou, Q.; Waterhouse, G.I.N.; Hou, J.; Ai, S.; Li, X. A simple aptamer-based fluorescent aflatoxin B1 sensor using humic acid as quencher. Talanta 2019, 205, 120131. [Google Scholar] [CrossRef]
  172. Chen, Q.; Jiao, T.; Yang, M.; Li, H.; Ahmad, W.; Hassan, M.M.; Guo, Z.; Ali, S. Pre etched Ag nanocluster as SERS substrate for the rapid quantification of AFB1 in peanut oil via DFT coupled multivariate calibration. Spectrochim. Acta Part A 2020, 239, 118411. [Google Scholar] [CrossRef]
  173. Li, J.; Wang, Q.; **ong, C.; Deng, Q.; Zhang, X.; Wang, S.; Chen, M.M. An ultrasensitive CH3NH3PbBr3 quantum dots@SiO2-based electrochemiluminescence sensing platform using an organic electrolyte for aflatoxin B1 detection in corn oil. Food Chem. 2022, 390, 133200. [Google Scholar] [CrossRef]
  174. He, H.; Sun, D.W.; Pu, H.; Huang, L. Bridging Fe3O4@Au nanoflowers and Au@Ag nanospheres with aptamer for ultrasensitive SERS detection of aflatoxin B1. Food Chem. 2020, 324, 126832. [Google Scholar] [CrossRef]
  175. Jiang, Y.; Sun, D.-W.; Pu, H.; Wei, Q. A simple and sensitive aptasensor based on SERS for trace analysis of kanamycin in milk. J. Food Meas. Charact. 2020, 14, 3184–3193. [Google Scholar] [CrossRef]
  176. Toh, S.Y.; Citartan, M.; Gopinath, S.C.; Tang, T.H. Aptamers as a replacement for antibodies in enzyme-linked immunosorbent assay. Biosens. Bioelectron. 2015, 64, 392–403. [Google Scholar] [CrossRef]
  177. Jiang, Y.; Sun, D.W.; Pu, H.; Wei, Q. Ultrasensitive analysis of kanamycin residue in milk by SERS-based aptasensor. Talanta 2019, 197, 151–158. [Google Scholar] [CrossRef]
  178. Azadniya, E.; Goldoni, L.; Bandiera, T.; Morlock, G.E. Same analytical method for both (bio)assay and zone isolation to identify/quantify bioactive compounds by quantitative nuclear magnetic resonance spectroscopy. J. Chromatogr. A 2020, 1628, 461434. [Google Scholar] [CrossRef] [PubMed]
  179. Malu, S.P.; Donatus, R.B.; Ugye, J.T.; Imarenezor, E.P.K.; Leubem, A. Determination of Aflaxtoxin in Some Edible Oils Obtained from Makurdi Metropolis, North Central Nigeria. Am. J. Chem. Appl. 2017, 4, 36–40. [Google Scholar]
  180. Zhang, Y.; Li, M.; Cui, Y.; Hong, X.; Du, D. Using of Tyramine Signal Amplification to Improve the Sensitivity of ELISA for Aflatoxin B1 in Edible Oil Samples. Food Anal. Methods 2018, 11, 2553–2560. [Google Scholar] [CrossRef]
  181. Pundir, C.S.; Yadav, N.; Chhillar, A.K. Occurrence, synthesis, toxicity and detection methods for acrylamide determination in processed foods with special reference to biosensors: A review. Trends Food Sci. Technol. 2019, 85, 211–225. [Google Scholar] [CrossRef]
  182. Wang, X.; Shan, Y.; Gong, M.; **, X.; Lv, l.; Jiang, M.; Xu, J. A novel electrochemical sensor for ochratoxin A based on the hairpin aptamer and double report DNA via multiple signal amplification strategy. Sens. Actuators B 2019, 281, 595–601. [Google Scholar] [CrossRef]
  183. Wang, Z.; Ma, B.; Shen, C.; Cheong, L.Z. Direct, selective and ultrasensitive electrochemical biosensing of methyl parathion in vegetables using Burkholderia cepacia lipase@MOF nanofibers-based biosensor. Talanta 2019, 197, 356–362. [Google Scholar] [CrossRef]
  184. Lin, T.; Shen, Y. Fabricating electrochemical aptasensors for detecting aflatoxin B1 via layer-by-layer self-assembly. J. Electroanal. Chem. 2020, 870, 114247. [Google Scholar] [CrossRef]
  185. Xuan, Z.; Liu, H.; Ye, J.; Li, L.; Tian, W.; Wang, S. Reliable and disposable quantum dot-based electrochemical immunosensor for aflatoxin B1 simplified analysis with automated magneto-controlled pretreatment system. Anal. Bioanal. Chem. 2020, 412, 7615–7625. [Google Scholar] [CrossRef]
  186. Ben Rejeb, I.; Arduini, F.; Arvinte, A.; Amine, A.; Gargouri, M.; Micheli, L.; Bala, C.; Moscone, D.; Palleschi, G. Development of a bio-electrochemical assay for AFB1 detection in olive oil. Biosens. Bioelectron. 2009, 24, 1962–1968. [Google Scholar] [CrossRef]
  187. Kim, M.; Iezzi, R., Jr.; Shim, B.S.; Martin, D.C. Impedimetric Biosensors for Detecting Vascular Endothelial Growth Factor (VEGF) Based on Poly(3,4-ethylene dioxythiophene) (PEDOT)/Gold Nanoparticle (Au NP) Composites. Front. Chem. 2019, 7, 234. [Google Scholar] [CrossRef]
  188. Lu, Y.; Zhang, B.; Tian, Y.; Guo, Q.; Yang, X.; Nie, G. An enhanced photoelectrochemical sensor for aflatoxin B1 detection based on organic-inorganic heterojunction nanomaterial: Poly(5-formylindole)/NiO. Mikrochim. Acta 2020, 187, 467. [Google Scholar] [CrossRef] [PubMed]
  189. Sharma, P.; Panchal, A.; Yadav, N.; Narang, J. Analytical techniques for the detection of glycated haemoglobin underlining the sensors. Int. J. Biol. Macromol. 2020, 155, 685–696. [Google Scholar] [CrossRef] [PubMed]
  190. Yu, L.; Zhang, Y.; Hu, C.; Wu, H.; Yang, Y.; Huang, C.; Jia, N. Highly sensitive electrochemical impedance spectroscopy immunosensor for the detection of AFB1 in olive oil. Food Chem. 2015, 176, 22–26. [Google Scholar] [CrossRef] [PubMed]
  191. Perumal, V.; Hashim, U. Advances in biosensors: Principle, architecture and applications. J. Appl. Biomed. 2014, 12, 1–15. [Google Scholar] [CrossRef]
  192. Wang, H.; Zhang, Y.; Chu, Y.; Ma, H.; Li, Y.; Wu, D.; Du, B.; Wei, Q. Disposable competitive-type immunoassay for determination of aflatoxin B1 via detection of copper ions released from Cu-apatite. Talanta 2016, 147, 556–560. [Google Scholar] [CrossRef]
  193. Farka, Z.; Jurik, T.; Kovar, D.; Trnkova, L.; Skladal, P. Nanoparticle-Based Immunochemical Biosensors and Assays: Recent Advances and Challenges. Chem. Rev. 2017, 117, 9973–10042. [Google Scholar] [CrossRef]
  194. Wang, X.; Wu, X.; Lu, Z.; Tao, X. Comparative Study of Time-Resolved Fluorescent Nanobeads, Quantum Dot Nanobeads and Quantum Dots as Labels in Fluorescence Immunochromatography for Detection of Aflatoxin B1 in Grains. Biomolecules 2020, 10, 575. [Google Scholar] [CrossRef]
  195. Perez-Fernandez, B.; de la Escosura-Muniz, A. Electrochemical biosensors based on nanomaterials for aflatoxins detection: A review (2015–2021). Anal. Chim. Acta. 2022, 1212, 339658. [Google Scholar] [CrossRef]
  196. Sun, C.; Li, H.; Koidis, A.; Chen, Q. Quantifying Aflatoxin B1 in peanut oil using fabricating fluorescence probes based on upconversion nanoparticles. Spectrochim. Acta Part A 2016, 165, 120–126. [Google Scholar] [CrossRef]
  197. **e, G.; Zhu, M.; Liu, Z.; Zhang, B.; Shi, M.; Wang, S. Development and evaluation of the magnetic particle-based chemiluminescence immunoassay for rapid and quantitative detection of Aflatoxin B1 in foodstuff. Food Agric. Immunol. 2017, 29, 564–576. [Google Scholar] [CrossRef]
  198. Neelam; Chhillar, A.K.; Rana, J.S. Enzyme nanoparticles and their biosensing applications: A review. Anal. Biochem. 2019, 581, 113345. [Google Scholar] [CrossRef] [PubMed]
  199. Ding, L.; Bond, A.M.; Zhai, J.; Zhang, J. Utilization of nanoparticle labels for signal amplification in ultrasensitive electrochemical affinity biosensors: A review. Anal. Chim. Acta. 2013, 797, 1–12. [Google Scholar] [CrossRef] [PubMed]
  200. Peltomaa, R.; Benito-Pena, E.; Moreno-Bondi, M.C. Bioinspired recognition elements for mycotoxin sensors. Anal. Bioanal. Chem. 2018, 410, 747–771. [Google Scholar] [CrossRef] [PubMed]
  201. Miklos, G.; Angeli, C.; Ambrus, A.; Nagy, A.; Kardos, V.; Zentai, A.; Kerekes, K.; Farkas, Z.; Jozwiak, A.; Bartok, T. Detection of Aflatoxins in Different Matrices and Food-Chain Positions. Front. Microbiol. 2020, 11, 1916. [Google Scholar] [CrossRef]
  202. Wang, Q.; Yang, Q.; Wu, W. Progress on Structured Biosensors for Monitoring Aflatoxin B1 From Biofilms: A Review. Front. Microbiol. 2020, 11, 408. [Google Scholar] [CrossRef]
  203. Langone, J.J.; Van Vunakis, H. Aflatoxin B1: Specific antibodies and their use in radioimmunoassay. J. Natl. Cancer Inst. 1976, 56, 591–595. [Google Scholar] [CrossRef] [PubMed]
  204. Bazin, I.; Tria, S.A.; Hayat, A.; Marty, J.L. New biorecognition molecules in biosensors for the detection of toxins. Biosens. Bioelectron. 2017, 87, 285–298. [Google Scholar] [CrossRef]
  205. Ma, H.; Sun, J.; Zhang, Y.; Bian, C.; **a, S.; Zhen, T. Label-free immunosensor based on one-step electrodeposition of chitosan-gold nanoparticles biocompatible film on Au microelectrode for determination of aflatoxin B1 in maize. Biosens. Bioelectron. 2016, 80, 222–229. [Google Scholar] [CrossRef] [PubMed]
  206. Zhao, F.; Tian, Y.; Shen, Q.; Liu, R.; Shi, R.; Wang, H.; Yang, Z. A novel nanobody and mimotope based immunoassay for rapid analysis of aflatoxin B1. Talanta 2019, 195, 55–61. [Google Scholar] [CrossRef]
  207. Shi, L.; Wang, Z.; Yang, G.; Yang, H.; Zhao, F. A novel electrochemical immunosensor for aflatoxin B1 based on Au nanoparticles-poly 4-aminobenzoic acid supported graphene. Appl. Surf. Sci. 2020, 527, 146934. [Google Scholar] [CrossRef]
  208. Pfeiffer, F.; Mayer, G. Selection and Biosensor Application of Aptamers for Small Molecules. Front. Chem. 2016, 4, 25. [Google Scholar] [CrossRef] [PubMed]
  209. Kong, H.Y.; Byun, J. Nucleic Acid aptamers: New methods for selection, stabilization, and application in biomedical science. Biomol. Ther. 2013, 21, 423–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Kleinjung, F.; Klussmann, S.; Erdmann, V.; Scheller, F.; Fürste, J.; Bier, F. High-affinity RNA as a recognition element in a biosensor. Anal. Chem. 1998, 70, 328–331. [Google Scholar] [CrossRef]
  211. Ma, X.; Wang, W.; Chen, X.; **a, Y.; Wu, S.; Duan, N.; Wang, Z. Selection, identification, and application of Aflatoxin B1 aptamer. Eur. Food Res. Technol. 2014, 238, 919–925. [Google Scholar] [CrossRef]
  212. Vidal, J.C.; Bonel, L.; Ezquerra, A.; Hernandez, S.; Bertolin, J.R.; Cubel, C.; Castillo, J.R. Electrochemical affinity biosensors for detection of mycotoxins: A review. Biosens. Bioelectron. 2013, 49, 146–158. [Google Scholar] [CrossRef]
  213. Rivas, L.; Mayorga-Martinez, C.C.; Quesada-Gonzalez, D.; Zamora-Galvez, A.; de la Escosura-Muniz, A.; Merkoci, A. Label-free impedimetric aptasensor for ochratoxin-A detection using iridium oxide nanoparticles. Anal. Chem. 2015, 87, 5167–5172. [Google Scholar] [CrossRef]
  214. McKeague, M.; Bradley, C.R.; De Girolamo, A.; Visconti, A.; Miller, J.D.; Derosa, M.C. Screening and initial binding assessment of fumonisin b(1) aptamers. Int. J. Mol. Sci. 2010, 11, 4864–4881. [Google Scholar] [CrossRef]
  215. Sabet, F.S.; Hosseini, M.; Khabbaz, H.; Dadmehr, M.; Ganjali, M.R. FRET-based aptamer biosensor for selective and sensitive detection of aflatoxin B1 in peanut and rice. Food Chem. 2017, 220, 527–532. [Google Scholar] [CrossRef]
  216. Chen, X.; Bai, X.; Li, H.; Zhang, B. Aptamer-based microcantilever array biosensor for detection of fumonisin B-1. RSC Adv. 2015, 5, 35448–35452. [Google Scholar] [CrossRef]
  217. Guo, X.; Wen, F.; Zheng, N.; Li, S.; Fauconnier, M.L.; Wang, J. A qPCR aptasensor for sensitive detection of aflatoxin M1. Anal. Bioanal. Chem. 2016, 408, 5577–5584. [Google Scholar] [CrossRef]
  218. Samokhvalov, A.V.; Safenkova, I.V.; Eremin, S.A.; Zherdev, A.V.; Dzantiev, B.B. Use of anchor protein modules in fluorescence polarisation aptamer assay for ochratoxin A determination. Anal. Chim. Acta 2017, 962, 80–87. [Google Scholar] [CrossRef] [PubMed]
  219. Li, Q.; Lu, Z.; Tan, X.; **ao, X.; Wang, P.; Wu, L.; Shao, K.; Yin, W.; Han, H. Ultrasensitive detection of aflatoxin B1 by SERS aptasensor based on exonuclease-assisted recycling amplification. Biosens. Bioelectron. 2017, 97, 59–64. [Google Scholar] [CrossRef] [PubMed]
  220. Abnous, K.; Danesh, N.M.; Alibolandi, M.; Ramezani, M.; Sarreshtehdar Emrani, A.; Zolfaghari, R.; Taghdisi, S.M. A new amplified pi-shape electrochemical aptasensor for ultrasensitive detection of aflatoxin B1. Biosens. Bioelectron. 2017, 94, 374–379. [Google Scholar] [CrossRef]
  221. Wang, S.; Zhang, Y.; Pang, G.; Zhang, Y.; Guo, S. Tuning the Aggregation/Disaggregation Behavior of Graphene Quantum Dots by Structure-Switching Aptamer for High-Sensitivity Fluorescent Ochratoxin A Sensor. Anal. Chem. 2017, 89, 1704–1709. [Google Scholar] [CrossRef] [PubMed]
  222. Zhang, S.; Shen, Y.; Shen, G.; Wang, S.; Shen, G.; Yu, R. Electrochemical immunosensor based on Pd-Au nanoparticles supported on functionalized PDDA-MWCNT nanocomposites for aflatoxin B1 detection. Anal. Biochem. 2016, 494, 10–15. [Google Scholar] [CrossRef]
  223. Ren, S.; Li, Y.; Guo, Q.; Peng, Y.; Bai, J.; Ning, B.; Gao, Z. Turn-on fluorometric immunosensor for diethylstilbestrol based on the use of air-stable polydopamine-functionalized black phosphorus and upconversion nanoparticles. Mikrochim. Acta 2018, 185, 429. [Google Scholar] [CrossRef] [PubMed]
  224. Wu, J.; Ali, S.; Ouyang, Q.; Wang, L.; Rong, Y.; Chen, Q. Highly specific and sensitive detection of aflatoxin B1 in food based on upconversion nanoparticles-black phosphorus nanosheets aptasensor. Microchem. J. 2021, 171, 106847. [Google Scholar] [CrossRef]
  225. **a, X.; Wang, H.; Yang, H.; Deng, S.; Deng, R.; Dong, Y.; He, Q. Dual-Terminal Stemmed Aptamer Beacon for Label-Free Detection of Aflatoxin B1 in Broad Bean Paste and Peanut Oil via Aggregation-Induced Emission. J. Agric. Food Chem. 2018, 66, 12431–12438. [Google Scholar] [CrossRef]
  226. Yang, Y.; Yin, Y.; Li, X.; Wang, S.; Dong, Y. Development of a chimeric aptamer and an AuNPs aptasensor for highly sensitive and specific identification of Aflatoxin B1. Sens. Actuators B 2020, 319, 128250. [Google Scholar] [CrossRef]
  227. Zhong, T.; Li, S.; Li, X.; JiYe, Y.; Mo, Y.; Chen, L.; Zhang, Z.; Wu, H.; Li, M.; Luo, Q. A label-free electrochemical aptasensor based on AuNPs-loaded zeolitic imidazolate framework-8 for sensitive determination of aflatoxin B1. Food Chem. 2022, 384, 132495. [Google Scholar] [CrossRef]
  228. Wang, N.; Liu, Q.; Hu, X.; Wang, F.; Hu, M.; Yu, Q.; Zhang, G. Electrochemical immunosensor based on AuNPs/Zn/Ni-ZIF-8-800@graphene for rapid detection of aflatoxin B1 in peanut oil. Anal. Biochem. 2022, 650, 114710. [Google Scholar] [CrossRef] [PubMed]
  229. Zhao, Y.; Chen, F.; Li, Q.; Wang, L.; Fan, C. Isothermal Amplification of Nucleic Acids. Chem. Rev. 2015, 115, 12491–12545. [Google Scholar] [CrossRef] [PubMed]
  230. Hu, Y.; Cecconello, A.; Idili, A.; Ricci, F.; Willner, I. Triplex DNA Nanostructures: From Basic Properties to Applications. Angew. Chem. Int. Ed. Engl. 2017, 56, 15210–15233. [Google Scholar] [CrossRef] [PubMed]
  231. Bai, Y.; Zhang, H.; Zhao, L.; Wang, Y.; Chen, X.; Zhai, H.; Tian, M.; Zhao, R.; Wang, T.; Xu, H.; et al. A novel aptasensor based on HCR and G-quadruplex DNAzyme for fluorescence detection of Carcinoembryonic Antigen. Talanta 2021, 221, 121451. [Google Scholar] [CrossRef]
  232. Zhang, Q. Application of Hybridization Chain Reaction (HCR) in Electrochemical Analysis. Int. J. Electrochem. Sci. 2022, 17, 220227. [Google Scholar] [CrossRef]
  233. Dirks, R.M.; Pierce, N.A. Triggered amplification by hybridization chain reaction. Proc. Natl. Acad. Sci. USA 2004, 101, 15275–15278. [Google Scholar] [CrossRef]
  234. Evanko, D. Hybridization chain reaction. Nat. Methods 2004, 1, 186. [Google Scholar] [CrossRef]
  235. Zhou, R.; Zeng, Z.; Sun, R.; Liu, W.; Zhu, Q.; Zhang, X.; Chen, C. Traditional and new applications of the HCR in biosensing and biomedicine. Analyst 2021, 146, 7087–7103. [Google Scholar] [CrossRef]
  236. Wang, Q.; Zhao, F.; Yang, Q.; Wu, W. Graphene oxide quantum dots based nanotree illuminates AFB1: Dual signal amplified aptasensor detection AFB1. Sens. Actuators B 2021, 345, 130387. [Google Scholar] [CrossRef]
  237. Zou, L.; Zhang, M.; Li, M.; **ao, Z.; Ling, L. Hybridization chain reaction and DNAzyme-based dual signal amplification strategy for sensitive fluorescent sensing of aflatoxin B1 by using the pivot of triplex DNA. Food Res. Int. 2022, 158, 111538. [Google Scholar] [CrossRef]
  238. Molaei, M.J. Principles, mechanisms, and application of carbon quantum dots in sensors: A review. Anal. Methods 2020, 12, 1266–1287. [Google Scholar] [CrossRef]
  239. Lim, S.Y.; Shen, W.; Gao, Z. Carbon quantum dots and their applications. Chem. Soc. Rev. 2015, 44, 362–381. [Google Scholar] [CrossRef]
  240. Tajik, S.; Dourandish, Z.; Zhang, K.; Beitollahi, H.; Le, Q.V.; Jang, H.W.; Shokouhimehr, M. Carbon and graphene quantum dots: A review on syntheses, characterization, biological and sensing applications for neurotransmitter determination. RSC Adv. 2020, 10, 15406–15429. [Google Scholar] [CrossRef] [PubMed]
  241. Ye, J.; Zheng, M.; Ma, H.; Xuan, Z.; Tian, W.; Liu, H.; Wang, S.; Zhang, Y. Development and Validation of an Automated Magneto-Controlled Pretreatment for Chromatography-Free Detection of Aflatoxin B1 in Cereals and Oils through Atomic Absorption Spectroscopy. Toxins 2022, 14, 454. [Google Scholar] [CrossRef]
  242. Lu, Z.; Chen, X.; Wang, Y.; Zheng, X.; Li, C.M. Aptamer based fluorescence recovery assay for aflatoxin B1 using a quencher system composed of quantum dots and graphene oxide. Microchim. Acta 2014, 182, 571–578. [Google Scholar] [CrossRef]
  243. Hussain, A.; Sun, D.W.; Pu, H. Bimetallic core shelled nanoparticles (Au@AgNPs) for rapid detection of thiram and dicyandiamide contaminants in liquid milk using SERS. Food Chem. 2020, 317, 126429. [Google Scholar] [CrossRef]
  244. Jiao, T.; Ahmad, W.; Zhu, J.; Hassan, M.M.; Wang, J.; Rong, Y.; Guo, Z.; Li, H.; Ding, Z.; Lv, C.; et al. Aggregation triggered aflatoxin B1 determination in foodstuff employing 5-aminotetramethylrhodamine decorated gold–silver core–shell nanoparticles in surface enhanced Raman scattering. Sens. Actuators B 2021, 331, 129424. [Google Scholar] [CrossRef]
  245. **ong, Z.; Wang, Q.; **e, Y.; Li, N.; Yun, W.; Yang, L. Simultaneous detection of aflatoxin B1 and ochratoxin A in food samples by dual DNA tweezers nanomachine. Food Chem. 2021, 338, 128122. [Google Scholar] [CrossRef] [PubMed]
  246. Wu, S.S.; Wei, M.; Wei, W.; Liu, Y.; Liu, S. Electrochemical aptasensor for aflatoxin B1 based on smart host-guest recognition of β-cyclodextrin polymer. Biosens. Bioelectron. 2019, 129, 58–63. [Google Scholar] [CrossRef] [PubMed]
  247. Ahmad, O.S.; Bedwell, T.S.; Esen, C.; Garcia-Cruz, A.; Piletsky, S.A. Molecularly Imprinted Polymers in Electrochemical and Optical Sensors. Trends Biotechnol. 2019, 37, 294–309. [Google Scholar] [CrossRef]
  248. Uzun, L.; Turner, A.P. Molecularly-imprinted polymer sensors: Realising their potential. Biosens. Bioelectron. 2016, 76, 131–144. [Google Scholar] [CrossRef] [PubMed]
  249. BelBruno, J.J. Molecularly Imprinted Polymers. Chem. Rev. 2019, 119, 94–119. [Google Scholar] [CrossRef] [PubMed]
  250. Li, S.; Ge, Y.; Piletsky, S.A.; Lunec, J. Molecularly Imprinted Sensors: Overview and Applications; Elsevier: Amsterdam, The Netherlands, 2012. [Google Scholar]
  251. Haupt, K. Molecular Imprinting; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012; Volume 325. [Google Scholar]
  252. Mirsky, V.M.; Yatsimirsky, A. Artificial Receptors for Chemical Sensors; John Wiley & Sons: Hoboken, NJ, USA, 2010. [Google Scholar]
  253. Sergey, A.; Piletsky, M.J.W. Designing Receptors for the Next Generation of Biosensors, 1st ed.; Springer: Berlin/Heidelberg, Germany, 2013; Volume 12, p. 264. [Google Scholar]
  254. Lee, S.-W.; Kunitake, T. Handbook of Molecular Imprinting: Advanced Sensor Applications; CRC Press: Boca Raton, FL, USA, 2012. [Google Scholar]
  255. Wang, S.; Zhu, X.; Zhao, M.; Li, S.; Le, Y.; Li, H. Optical sensors based on molecularly imprinted nanomaterials. In Smart Nanomaterials for Sensor Application; Bentham: Oak Park, IL, USA, 2012; pp. 60–75. [Google Scholar]
  256. Alvarez-Lorenzo, C. Handbook of Molecularly Imprinted Polymers; Smithers Rapra: Shawbury, UK, 2013. [Google Scholar]
  257. Chen, L.; Wang, X.; Lu, W.; Wu, X.; Li, J. Molecular imprinting: Perspectives and applications. Chem. Soc. Rev. 2016, 45, 2137–2211. [Google Scholar] [CrossRef]
  258. Sergeyeva, T.; Yarynka, D.; Piletska, E.; Linnik, R.; Zaporozhets, O.; Brovko, O.; Piletsky, S.; El’skaya, A. Development of a smartphone-based biomimetic sensor for aflatoxin B1 detection using molecularly imprinted polymer membranes. Talanta 2019, 201, 204–210. [Google Scholar] [CrossRef]
  259. Gu, Y.; Wang, Y.; Wu, X.; Pan, M.; Hu, N.; Wang, J.; Wang, S. Quartz crystal microbalance sensor based on covalent organic framework composite and molecularly imprinted polymer of poly(o-aminothiophenol) with gold nanoparticles for the determination of aflatoxin B1. Sens. Actuators B 2019, 291, 293–297. [Google Scholar] [CrossRef]
  260. Gui, R.; **, H.; Guo, H.; Wang, Z. Recent advances and future prospects in molecularly imprinted polymers-based electrochemical biosensors. Biosens. Bioelectron. 2018, 100, 56–70. [Google Scholar] [CrossRef] [PubMed]
  261. Cavaliere, C.; Foglia, P.; Guarino, C.; Nazzari, M.; Samperi, R.; Lagana, A. Determination of aflatoxins in olive oil by liquid chromatography-tandem mass spectrometry. Anal. Chim. Acta 2007, 596, 141–148. [Google Scholar] [CrossRef]
  262. Tan, H.; Ma, L.; Guo, T.; Zhou, H.; Chen, L.; Zhang, Y.; Dai, H.; Yu, Y. A novel fluorescence aptasensor based on mesoporous silica nanoparticles for selective and sensitive detection of aflatoxin B1. Anal. Chim. Acta 2019, 1068, 87–95. [Google Scholar] [CrossRef]
  263. **a, X.; Wang, Y.; Yang, H.; Dong, Y.; Zhang, K.; Lu, Y.; Deng, R.; He, Q. Enzyme-free amplified and ultrafast detection of aflatoxin B1 using dual-terminal proximity aptamer probes. Food Chem. 2019, 283, 32–38. [Google Scholar] [CrossRef]
  264. **ao, M.W.; Bai, X.L.; Liu, Y.M.; Yang, L.; Liao, X. Simultaneous determination of trace Aflatoxin B1 and Ochratoxin A by aptamer-based microchip capillary electrophoresis in food samples. J. Chromatogr. A 2018, 1569, 222–228. [Google Scholar] [CrossRef]
  265. Li, R.; Wang, X.; Zhou, T.; Yang, D.; Wang, Q.; Zhou, Y. Occurrence of four mycotoxins in cereal and oil products in Yangtze Delta region of China and their food safety risks. Food Control 2014, 35, 117–122. [Google Scholar] [CrossRef]
  266. Chen, Y.; Yang, Y.; Wang, Y.; Peng, Y.; Nie, J.; Gao, G.; Zhi, J. Development of an Escherichia coli-based electrochemical biosensor for mycotoxin toxicity detection. Bioelectrochemistry 2020, 133, 107453. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Harmful effects of different types aflatoxins contaminated edible oil.
Figure 1. Harmful effects of different types aflatoxins contaminated edible oil.
Molecules 27 06141 g001
Figure 2. Main mechanisms of toxicity of aflatoxin B1 for humans.
Figure 2. Main mechanisms of toxicity of aflatoxin B1 for humans.
Molecules 27 06141 g002
Figure 3. Illustration of the mechanism of hepatocellular carcinoma caused by ingestion of AFB1-contaminated foods.
Figure 3. Illustration of the mechanism of hepatocellular carcinoma caused by ingestion of AFB1-contaminated foods.
Molecules 27 06141 g003
Figure 4. Timeline of the limit of detection on AFB1 in edible oil by different (a) detection technology and (b) detection method. CI: Chemiluminescence immunoassay.
Figure 4. Timeline of the limit of detection on AFB1 in edible oil by different (a) detection technology and (b) detection method. CI: Chemiluminescence immunoassay.
Molecules 27 06141 g004
Figure 5. Scheme of the two or three-electrode setup used in electrochemical methods.
Figure 5. Scheme of the two or three-electrode setup used in electrochemical methods.
Molecules 27 06141 g005
Figure 6. Schematic diagram of typical biosensor. Including analyzer, bioreceptor, transducer, electronic system (amplifier and processor), detector (for data processing).
Figure 6. Schematic diagram of typical biosensor. Including analyzer, bioreceptor, transducer, electronic system (amplifier and processor), detector (for data processing).
Molecules 27 06141 g006
Table 1. The maximum limits (μg kg−1) established for major AFB1 in some countries/regions for edible oils.
Table 1. The maximum limits (μg kg−1) established for major AFB1 in some countries/regions for edible oils.
Countries/AgenciesFood ProductsEdible Vegetable OilTotal of AFs
(μg kg−1)
AFB1
(μg kg−1)
Refs.
EUOil seeds-158[32,69]
EU-Peanut oil
Others oil
42[32,69]
China-Maize oil
Peanut oil
Others oils
-
-
-
20
20
10
[73]
Greece-Olive oil--[69]
Russia-Vegetable oil--[64]
France-Vegetable oil-5[64]
Kenya-Vegetable oil20-[64,71]
Taiwan-Edible oil10-[65]
Morocco-Vegetable oil-5[72]
ThailandAll foodsOil and fats20-[64,65,74]
USAAll foods-20-[64,71,75]
BrazilAll foods--15[76]
IndiaAll foods--30[65]
ChileAll foods---[76]
IndonesiaAll foods-3520[65]
Singapore
Australia
All foods-5-[64,65]
MalaysiaAll foods-35-[65]
Japan
Vietnam
All foods-10-[65]
Sri LankaAll foods-30-[65]
Table 2. Techniques used for the detection of AFB1 in different types of edible oil.
Table 2. Techniques used for the detection of AFB1 in different types of edible oil.
MatrixAnalytical MethodSample Preparation MethodLinear RangeRecoveryLODRef.
Peanut oilELISAImmunoaffinity column cleanup-84.40–92.60%1.08 μg kg−1[20]
Coconut oilHPLC-FLDImmunoaffinity chromatography--0.01–0.04 μg kg−1[81]
Peanut oil
Corn oil
Soybean oil
HPLC-FLDDLLME with in situ derivatization0.1–100 ng mL−1106.90–121.50%0.03 ng mL−1[82]
Sunflower oil
Olive oil
Canola oil
Frying oil
Blend oil
HPLC-FLDImmunoaffinity column cleanup0.04–0.16 ng g−195.56–102.13%0.16 ng g−1[83]
Vegetable oilHPLC-FLDImmunoaffinity chromatography
- Solid phase extraction
-95.20–99.00%0.25 μg kg−1[84]
Coconut oil Almond oil
Soybean oil
Olive oil
HPLC -FLDImmuno Affinity chromatography combined with DLLME0.005–10.00 ng mL−196.00–109.90%830 ng mL−1[85]
Sesame oil
Sunflower oil
Peanut oil
Mixed oil
HPLC-FLDLiquid-Liquid Extraction0.34–109.2 µg kg−183.00–96.00%0.09–1.5 µg kg−1[86]
Canola oil
Corn oil
Olive oil
Peanut oil
Soybean oil
LC-MS/MSLiquid-Partitioning-101.00–111.00%0.030 μg kg−1[87]
Soybean oil
Corn oil
Rice bran oil
LC-MS/MSQuEChERS × DLLME-70.70–76.00%[88]
Blend oil
Peanut oil
Maize oil
LC-MS/MSHollow fiber liquid phase microextraction0.1–500 μg kg−178.59–80.61%0.02 μg kg−1[89]
Sunflower oil
Palm oil
Corn oil
LC-ESI-MS/MSQuEChERS0.04–2000 ng g−187.90–106.60%0.01 ng kg−1[90]
Soybean oil
Corn oil
Peanut oil
Blended oil
LC–MS/MSImmunoaffinity chromatography0.16 μg kg−187.40–97.30%0.05 μg kg−1[96]
Olive oil
Peanut oil
Sesame oil
LC-MS/MSImmunoaffinity column cleanup2–20 mg kg−187.70–102.20%0.1 μg kg−1[97]
Olive oilLC/ESI-MS/MSMatrix Solid Phase Dispersion0.2–0.4 (pg inj)95.00–98.00%0.2 pg inj[261]
Olive oil Sunflower oil
Soybean oil
Corn oil
UHPLC-QqQ-MS/MSQuEChERS0.5–25 μg kg−196.00–107.90%0.5 μg kg−1[100]
Sesame oil Groundnut oil Cottonseed oilHPLCLiquid-Liquid extraction0.2–0.8 μg kg−1-0.1 μg kg−1[102]
Vegetable oilsGPC-HPLC-FLDLiquid-Liquid Extraction1.0–30.0 μg kg−182.60–90.60%1.0 μg kg−1[104]
Peanut oil
Sunflower oil
Olive oil
IAC-LC-ESI–MS/MSLiquid-Liquid Extraction0.02–10 μg kg−184.00–99.00%0.02 μg kg−1[105]
Virgin olive oilHPLC-FLDSolid Phase
Extraction
65.50–87.50%0.25 ng g−1[110]
Canola oil
Soybean oil
Corn oil
Olive oil
Peanut oil
FLLTC-IMSPE0.0048–0.0126 ng·g−179.60–117.90%0.0048 ng·g−1[115]
Rapeseed oil
Peanut oil
Blended oil
Blended olive oil
Sunflower oil
Tea oil
Rice oil
Corn oil
Sesame oil
Soybean oil
HPLC-MS/MSQuEChERS0.2–20 ng mL−187.80–98.60%0.05 ng g−1[116]
Soya bean oil
Groundnut oil
Beniseed oil
Palm kernel oil
Melon oil
Coconut oil
ELISAImmunoaffinity column cleanup (226 Aflatoxin clean-up Column)--≤0.8352 μg L−1[179]
Peanut oil
Virgin olive oil
ELISA& TSA-ELISALiquid-Liquid Extraction-81.40–118.80%0.004 ng mL−1[180]
Olive oilAmperometric biosensor coupled with AChE enzymeLiquid-Liquid Extraction10–60 ppb76.00–78.00%2 ppb[186]
Olive oilEIS based on MWCNTs/RTIL composite filmsLiquid-partitioning0.1–10 ng mL−196.00–116.00%0.03 ng mL−1[190]
Peanut oilDisposable electrochemical immunosensor with Au NPs modified SPCE-0.001–100 ng mL−190.00–102.00%0.2 pg mL−1[192]
Peanut oilFluorescence spectroscopy based on UCNPs
upconversion nanoparticles (UCNPs)
Liquid-partitioning0.2–100 ng mL−192.80–113.40%0.2 ng mL−1[196]
OilChemiluminescence immunoassay combined with the magnetic particles (MPCLIA)Liquid-partitioning0.1–100 ng mL−185.67–108.67%0.05 ng mL−1[197]
Corn germ oil
Peanut oil
An immunoassay based on both recombinant antibody and its mimotopeLiquid-Liquid extraction0.242.21 ng mL−186.70–116.20%0.13 ng mL−1[206]
Vegetable oilImmobilized immunosensor based on the hybrid gold nanoparticles-poly 4-aminobenzoic acid supported graphene-0.01–25 ng mL−1-0.001 ng mL−1[207]
Peanut oilUCNPs-BPNSs aptamer-0.2–500 ng mL−192.89–99.24%0.028 ng mL−1[224]
Peanut oilDual-terminal stemmed aptamer beacon, aggregation-induced emissionLiquid-Liquid Extraction40–300 ng mL−193.59–109.30%27.3 ng mL−1[225]
Corn oilA chimeric aptamer-based gold nanoparticles aptasensor-5–5120 nM91.50–117.60%1.88 nM[226]
Corn oil
Peanut oil
An electrochemical aptasensor base on an AuNPs/ZIF-8 nanocomposite-10.0–1.0 × 105 pg mL−193.49–106.90%1.82 pg mL−1[227]
Peanut oilAn electrochemical aptasensor base on an AuNPs/Zn/Ni-ZIF-8–800@ graphene nanocomposite-0.18–100 ng mL−180.26–109.60%0.18 ng mL−1[228]
OilAn aptasensor of hybridization chain reaction and Zn2+-dependent DNAzyme catalyzed cleavage-0.4–16 nmol L−192.20–107.80%0.22 nmol L−1[237]
OilFabricating electrochemical aptasensors-0.04–0.10 ng m L-194.5–103.3%0.002 ng m L−1[184]
Peanut oilElectrochemical immunosensor base on AFB1-BSA-QDs-0.08–80 μg kg−1102.70–113.30%0.05 μg kg−1[185]
Peanut oilSERS aptasensor-0.0001–100 ng·mL−196.60–115.00%0.40 pg·mL−1[174]
Peanut oilSERS aptasensor-0.001–10 ng mL−194.70–109.00%0.54 pg mL−1[169]
Peanut oilQ-dots-aptamer-GO fluorescence quenching system-1.6–160 μM-1.4 nM[242]
Peanut oilAtomic absorption spectroscopy-2.5–240 μg kg−1-0.04 μg kg−1[241]
Peanut oilSERS aptasensor0.01–100 ng mL−191.09–105.73%0.0036 ng mL−1[170]
Peanut oilSERS aptasensor with NH2-Rh-Au@Ag CSNPsSolid Phase
Extraction
0.1–5.0 ng mL−1-0.03 ng mL−1[244]
Olive oil
Peanut oil
Dual DNA tweezers nanomachine-0.08–10 ppb90.00–110.00%0.035 ppb[245]
Peanut oilElectrochemical aptasensor based on smart host-guest recognition of β-cyclodextrin polymer-0.1 × 10−4–10 ng mL−194.50–106.70%0.049 pg mL−1[246]
Peanut oilA dual signal amplified aptasensor based on
DNA walker, (DTNs) and network (HCR)
-1−1000 pg mL−187.56–105.28%0.492 pg mL−1[236]
Corn oilA novel fluorescence aptasensor based on mesoporous silica nanoparticles-0.5–50 ng mL−190.30–92.40%0.13 ng mL−1[262]
Peanut oilDual-terminal proximity aptamer probes-1.0–200 ng mL−190.30–102.91%0.9 ng mL−1[263]
Sesame oil
Olive oil
Peanut oil
Soybean oil
An aptamer-based MCE-LIF-0.05–5.0 ng mL−195.29–109.19%0.026 ng mL−1[264]
Peanut oilA simple fluorescent AFB1 sensor based on a humic acid/carbon dots system-0.1–0.8 ng mL−1103.80–108.00%70 pg mL−1[171]
Peanut oilSERS aptasensor-0.01–100 ng mL−190.40–113.10%5.0 ng mL−1[172]
Edible oilImmunoaffinity chromatography fluorometerImmunoaffinity column clean-up1.0–32.2 μg kg−1 -1 μg kg−1[265]
Corn oil An MIP-ECP-ECL sensing platform based on CH3NH3PbBr3 quantum dots (MAPB QDs)@SiO2-10−5–10 ng mL−1102.00–110.00%8.5 fg mL−1 [173]
Soybean oilTerahertz spectroscopy (photoelectric techniques)---2 μg kg−1[162]
Peanut oil
Corn oil
ELC based on Escherichia coli-0.01–0.3 μg mL−190.00–112.00%1 μg mL−1[266]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yin, S.; Niu, L.; Liu, Y. Recent Progress on Techniques in the Detection of Aflatoxin B1 in Edible Oil: A Mini Review. Molecules 2022, 27, 6141. https://doi.org/10.3390/molecules27196141

AMA Style

Yin S, Niu L, Liu Y. Recent Progress on Techniques in the Detection of Aflatoxin B1 in Edible Oil: A Mini Review. Molecules. 2022; 27(19):6141. https://doi.org/10.3390/molecules27196141

Chicago/Turabian Style

Yin, Shipeng, Liqiong Niu, and Yuanfa Liu. 2022. "Recent Progress on Techniques in the Detection of Aflatoxin B1 in Edible Oil: A Mini Review" Molecules 27, no. 19: 6141. https://doi.org/10.3390/molecules27196141

Article Metrics

Back to TopTop