Next Article in Journal
Exploring the Interaction of Biotinylated FcGamma RI and IgG1 Monoclonal Antibodies on Streptavidin-Coated Plasmonic Sensor Chips for Label-Free VEGF Detection
Next Article in Special Issue
Spermine Enhances the Peroxidase Activities of Multimeric Antiparallel G-quadruplex DNAzymes
Previous Article in Journal
Development of RT h-CLAT, a Rapid Assessment Method for Skin Sensitizers Using THP-1 Cells as a Biosensor
Previous Article in Special Issue
Construction of Metal–Organic Framework as a Novel Platform for Ratiometric Determination of Cyanide
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitigating Antibiotic Resistance: The Utilization of CRISPR Technology in Detection

1
Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
2
College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, China
3
Inner Mongolia Institute of Metrology and Testing, Hohhot 010030, China
4
Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, China Jiliang University, Hangzhou 310018, China
*
Authors to whom correspondence should be addressed.
Biosensors 2024, 14(12), 633; https://doi.org/10.3390/bios14120633
Submission received: 23 October 2024 / Revised: 7 December 2024 / Accepted: 17 December 2024 / Published: 20 December 2024
(This article belongs to the Special Issue Advanced Nanozyme for Biosensors)

Abstract

:
Antibiotics, celebrated as some of the most significant pharmaceutical breakthroughs in medical history, are capable of eliminating or inhibiting bacterial growth, offering a primary defense against a wide array of bacterial infections. However, the rise in antimicrobial resistance (AMR), driven by the widespread use of antibiotics, has evolved into a widespread and ominous threat to global public health. Thus, the creation of efficient methods for detecting resistance genes and antibiotics is imperative for ensuring food safety and safeguarding human health. The clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (Cas) systems, initially recognized as an adaptive immune defense mechanism in bacteria and archaea, have unveiled their profound potential in sensor detection, transcending their notable gene-editing applications. CRISPR/Cas technology employs Cas enzymes and guides RNA to selectively target and cleave specific DNA or RNA sequences. This review offers an extensive examination of CRISPR/Cas systems, highlighting their unique attributes and applications in antibiotic detection. It outlines the current utilization and progress of the CRISPR/Cas toolkit for identifying both nucleic acid (resistance genes) and non-nucleic acid (antibiotic micromolecules) targets within the field of antibiotic detection. In addition, it examines the current challenges, such as sensitivity and specificity, and future opportunities, including the development of point-of-care diagnostics, providing strategic insights to facilitate the curbing and oversight of antibiotic-resistance proliferation.

1. Introduction

1.1. Antibiotics

Antibiotics are mainly used to treat a range of bacterially induced bacterial infections. They are undoubtedly one of the most effective medications ever found in the history of human drug therapy. Most antibiotic chemical scaffolds currently in clinical use, such as β-lactams (penicillins and cephalosporins), macrolides, aminoglycosides, tetracyclines, glycopeptides, and fluoroquinolones, were discovered by scientists during the two decades between the clinical use of penicillin in the early 1940s and the discovery of fluoroquinolone antibiotics in 1962 [1]. Currently, in addition to their widespread use as therapeutic agents in humans, livestock, and aquaculture, antibiotics are also used as feed additives in animal husbandry. The use of antibiotic drugs has dramatically reduced the mortality rate caused by bacterial infections. However, the overuse of antibiotics has been recognized as a serious global public health problem due to the increase in the number of antibiotic-resistant bacteria in recent years, the decrease in the efficacy of antibiotics, and the multiplicity of infections [2].
Earlier this year, Chen et al. released a research survey showing that total antibiotic use in the Yangtze River Basin alone exceeded 1600 tons/year between 2013 and 2021 [3]. Grand View Research, a market research firm, recently published a market study on the antibiotics market, showing a global antimicrobial additives market size of USD 3.11 billion in 2023 [4], with increasing consumption of pharmaceuticals and medical devices and other end-use industries such as healthcare, personal care, and electronics, demand for antibiotics is growing at a significant rate. The global antimicrobial additive market size is expected to reach an estimated USD 5.63 billion by 2030 [4].
Antibiotics cannot be completely metabolized in living organisms and are often excreted into the environment as parent compounds or metabolites. Due to the limited removal capacity of conventional wastewater-treatment processes, antibiotics are continuously introduced into the environment, causing environmental pollution [5]. The most significant consequence of antibiotic release in the natural environment is the generation of antibiotic-resistance genes (ARGs) and -resistant bacteria (ARB), which not only causes pollution of the natural environment but also disrupt the structure and functionality of environmental microbial communities, with possible implications for human health and the evolution of ecological microbial populations [6].
Globally, several hundreds of thousands of people die each year from antibiotic-resistant bacteria [7], and it is estimated that by 2050, the global economic cost of ARB infections will reach USD 100 trillion [8]. In 2008, Rice et al. identified six “antibiotic failure” pathogens [9], which, according to a report published by the World Health Organization (WHO), can be considered the most common pathogens. According to the WHO 2024 Catalogue of Priority Bacterial Pathogens [10], a total of 15 drug-resistant bacteria are listed, and the existence of these drug-resistant bacteria poses a serious threat to global public health. It requires practical public health approaches and new antibiotic research to deal with it. However, it highlights that the development of new antibiotics is significantly outpaced by the evolution of bacterial resistance [11]. In the 21st century, only a handful of new antibiotic classes are available for clinical use. Without new antibiotics, by 2050, the number of deaths from drug-resistant infections is expected to reach 10 million per year globally [12].
Antibiotic resistance has become one of the most significant challenges [8]. A cross-national analysis of studies published in 2022 showed that high levels of resistance to pathogens are strongly associated with high mortality rates due to these pathogens [7]. Furthermore, the evolution and spread of antimicrobial resistance have been accelerated by the epidemic of pneumococcal pneumonia [13], and bacterial resistance poses a severe threat to human life. Therefore, in order to ensure food safety and human health, it is of great practical importance to establish efficient methods for detecting antibiotics in the environment or biological samples [14].

1.2. Rapid Antibiotic-Detection Methods

Antibiotic resistance is mainly caused by the misuse, abuse, and overuse of antibiotics across various sectors, including poultry, agriculture, livestock, healthcare, industry, and environmental management. This poses a serious threat to human, animal, and ecological health. Therefore, antibiotics come from different sources, including hospitals [15], livestock [16], aquaculture [17], agriculture [18], wastewater [18], and more.
The liquid chromatography mass spectrometry/mass spectrometry (LC-MS/MS) method for the detection of penicillin-group antibiotic residues in food of animal origin (GB/T 21315-2007) was promulgated and implemented by the China Academy of Inspection and Quarantine Science (CAIQS) in 2008, and is a standard detection method in laboratories. Other methods include LC-MS/MS [19,20], high-performance liquid chromatography (HPLC) [21], ultraviolet liquid chromatography (HPLC-UV) [22], field-effect transistor (FET) [23,24], surface-enhanced Raman spectroscopy (SERS) [25], enzyme-linked immunosorbent assay (ELISA) [26], etc. Current state-of-the-art antibiotic-monitoring methods are achieved by optical/electrochemical/colorimetric/biological sensors with driver modules such as SERS, metal–organic frameworks (MOFs), localized surface plasmon resonance (LSPR), CRISPR/Cas, and fluorescent materials [27,28]. The physicochemical properties of these sensors can be adapted by optimizing the sensor platform design scheme, spiking concentration, and synthesis parameters with relatively short response time, ease of use, portability, and adequate sensitivity and accuracy. The detection efficiency of these sensors can be further improved by integrating modern information technologies such as artificial intelligence (AI) [29]. Used as a monitoring tool, they can effectively prevent the overuse of antibiotics in the agricultural sector and production sectors such as poultry farms.
In recent studies, optical and electrochemical sensing platforms have also been coupled with aptamers (Apt) to design aptasensors [30,31] that use ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) aptamers for specific binding of targeted antibiotics [32,33]. Aptamer sensors are expected to detect specific antibiotics in a wide range of matrices with minimal sample pretreatment, have the advantages of rapidity, sensitivity, adjustability, versatility, and low cost, and provide miniaturized and portable sensing platforms [31], which overcomes the problems of traditional detection platforms such as large time consumption and costly, expensive, and complicated instrumentation, and low detection sensitivity and specificity; they are expected to promote the further antibiotic detection sensor. Table 1 summarizes the different methods used to detect antibiotics and compares the sensitivity of the different detection methods.

2. CRISPR/Cas Detection System

2.1. Introduction to the CRISPR/Cas System

The CRISPR/Cas are adaptive immune defense mechanisms in bacteria and archaea. The CRISPR/Cas system can be classified into Class-I and Class-II based on the types of effector proteins, which contain six isoforms: Type I, Type II, Type III, Type IV, Type V, and Type VI isoforms [45,46]. The Cas effector proteins of Class-I are multi-subunit proteins composed of complexes, including Type I, Type III, and Type IV isoforms. The Cas effector proteins of Class-II are composed of single subunits, including the Cas9 protein in Type II, the Cas12 protein in Type V, and the Cas13 protein in Type VI [47,48,49]. The Cas effector proteins of Class-II have a single structure that can efficiently achieve target recognition and cleavage, making them widely used for genome editing [50,51]. In these systems, the recognition of different target nucleic acids can be easily achieved by altering the crRNA sequence.
The first gene-editing tool to be developed was the Cas9 protein. In 2020, the Nobel Prize in Chemistry was awarded to French scientist Emmanuelle Charpentier and American scientist Jennifer A. Doudna for their discovery of one of the biggest tools in gene-editing technology, CRISPR/Cas9. Since then, research on the CRISPR/Cas9 system has been in full swing.
The CRISPR/Cas9 system consists of the Cas9 protein and guide RNA (gRNA). The Cas9 protein contains two major nuclease domains—the RuvC domain, which cuts noncomplementary DNA strands, and the HNH domain, which cuts complementary DNA strands; the gRNA serves a scaffolding function. The gRNA is a chimeric RNA formed by the combination of transactivated CRISPR RNA (tracrRNA), which functions as a scaffold, and specific crRNA, used to direct Cas9 to its target dsDNA. In this process, 5′-NGG-3′, the protospacer adjacent motif (PAM) sequences must be contained upstream of the targeting sequences [52]. Since PAM sequences are frequently found in most DNA sequences, Cas9 can target almost any gene with high specificity.
Currently, CRISPR/Cas-based nucleic acid assays are classified into two types in terms of principle: the first utilizes the property of Cas proteins such as Cas9 to recognize and bind dsDNA with high specificity, and the second utilizes the property of Cas proteins such as Cas13, Cas12, and other Cas proteins that activate trans-cleavage activity to cleave ssDNA or ssRNA nonspecifically after specifically recognizing the nucleic acid [53]. In 2016, Collins first introduced the CRISPR/Cas9 system to nucleic acid molecular diagnostics, developing a new technique to identify Zika virus lineages [54]. The method can provide precise genotypic information in hours and has attracted the attention of scientists. Researchers have developed a series of nucleic acid-detection protocols based on Cas9′s particular recognition and combined with the properties of dsDNA.
In 2018, Jennifer Doudna’s team published a related study on the CRISPR/Cas12 system in Science [55]. The CRISPR/Cas12 system consists of a single RuvC structural domain that mediates DNA cleavage at the far end of the PAM, labeled with the Cas12 gene, and some of them are mediated by single crRNAs such as CRISPR/Cas12a; some are co-mediated by crRNA and tracrRNA (or fused sgRNA alone), such as CRISPR/Cas12b [56,57]. As many as 11 isoforms (Cas12a-k) have been identified for the labeling genes of the CRISPR/Cas12 system, with recently discovered CasX classified as Cas12e, CasY classified as Cas12d, Cas14 categorized as Cas12f [58]; the most widely studied of these are Cas12a, Cas12b, and Cas12f [55,56,57,58,59].
Cas12a, also known as Cpf1, is a class of nucleic acid endonucleases mediated by a single crRNA that recognizes explicitly and shears dsDNA targets with PAM (5-TTTN-3′ or 5′-TTN-3), causing dsDNA targets to break and generate sticky ends. The recognition and shearing of ssDNA targets can be independent of PAM sequences. PAM sites are rich in thymines under the guidance of crRNA, activating CRISPR/Cas12a for targeted dsDNA cleavage (cis-cleavage) and activating CRISPR/Cas12a for non-targeted cleavage (trans-cleavage) [60]. To date, CRISPR/Cas12a-based tools have been widely used in many fields. Studies have shown that CRISPR/Cas12a has highly efficient trans-cleavage activity on ssDNA [61], a unique property that has been widely used in nucleic acid detection and has contributed to the improvement of CRISPR/Cas-based diagnostics (CRISPR-Dx) platforms by providing a strategy to improve specificity, sensitivity, and reaction speed [62].
The research related to CRISPR/Cas13 originated from a report published by Feng Zhang in Molecular Cell in 2015 [63], and his subsequent related report further elucidated that Cas13 has the function of cutting RNA, which formally unveiled the research and application of the CRISPR/Cas13 system.
The Cas13 protein does not require trans-activating RNA (tracrRNA) [49]. It has two distinct RNase activities: preprocessing of pre-crRNA into mature crRNA and shearing of the target RNA [64,65,66]. Cas13 is guided by a single crRNA, which catalyzes the maturation of pre-crRNA into mature crRNA, which recognizes specific pre-spacer sequence flanking sites (PFS, equivalent to the PAM sequence of DNA recognized by Cas9) in the non-target strand when the crRNA-Cas13a complex is formed. When the Cas13-crRNA complex recognizes PFS, the crRNA directs it to activate the CRISPR/Cas13 system by base complementary pairing with the target RNA. The target RNA is specifically sheared in cis, and nearby RNAs are trans-cleavage in a non-specific manner. At the time, this could be a promising tool for the detection of single-stranded RNA (ssRNA) virus detection [64,67].
Doudna’s group studied the Cas systems available in nature (Cas9, Cas12, and Cas13). They searched for uncharacterized Cas genes by creating a macrogenomic database of bacterial genomes, which led to the discovery of the Cas14 protein [68]. The Cas14 protein has been reported to target ssDNA and cleave ssDNA without PAM. The cas14 protein recognizes ssDNA, mediates the interaction of the target sequence with the target ssDNA, and cleaves ssDNA but not dsDNA or ssRNA. Similar to Cas9, the Cas14 protein requires tracrRNA and crRNA to target ssDNA [68]. Cas14 proteins are more specific in their cleavage efficiency than Cas9, Cas12, and Cas13 proteins that do not have a PAM region [58]. Therefore, this system meets all the criteria for high-fidelity genome editing.

2.2. CRISPR/Cas System for Detection of Nucleic Acid Targets

Because of its remarkable specificity and programmability in identifying and cutting particular DNA sequences, the CRISPR/Cas system has completely transformed the area of gene editing. Numerous studies have shown that the CRISPR/Cas system includes DNase/RNase characteristics in addition to its gene-editing ability, which makes it ideal for usage in the detection sector [69,70]. The nucleic acid signals of the target to be tested can be accurately recognized and targeted by gRNA/crRNA and Cas proteins. After recognizing ssDNA, dsDNA, or ssRNA targets, Cas12a/Cas13a/Cas14a proteins can exhibit nonspecific “trans-cleavage activity” towards nucleic acid sequences in the surrounding environment [61,71]. Numerous CRISPR/Cas-based nucleic acid-detection platforms, such as DNA endonuclease-targeted CRISPR trans reporter (DETECTR) [51], specific high-sensitivity enzymatic reporter unlocking (SHERLOCK) [72], one-hour low-cost multipurpose highly efficient system (HOLMES) [73], CRISPR/Cas-only amplification network (CONAN) [74], and others, have been successfully developed on the basis of these theoretical underpinnings.
The effector proteins of CRISPR/Cas12a (Cpf1) are RNA-directed enzymes that cleave both dsDNA and ssDNA. Similar to Cas9, Cas12a has been used for genome editing due to its ability to cleave targeted dsDNA. However, Cas12 (especially LbCas12a) stimulates nonspecific trans-cleavage activity by binding to its target DNA, which can completely degrade ssDNA molecules in the system. This target-activated transcutting activity is suitable for the molecular diagnosis of DNA and has contributed to the development of DETECTR methods [51]. For example, the trans-cutting activity of Cas12 was used as a diagnostic platform to diagnose Mycobacterium tuberculosis in high-sensitivity clinical samples [51]. Xu et al. also identified a DETECTR assay platform based on the recombinase polymerase amplification (RPA)-based DETECTR assay platform [75]. This method is rapid (<40 min), easy to implement, and accurate for identifying B. anthracis nucleic acids with a sensitivity close to 2 copies. Recent studies have also developed the DETECTR assay platform based on LtCas12a for precise and rapid detection of the human papillomavirus (HPV) 16/18 gene. Unlike AsCas12a and LbCas12a, which exert their cleavage activity by recognizing TTTV-containing PAM sites, limiting targeting, this LtCas12a utilizes unique TTNA PAM sites with equivalent cleavage capacity and specificity, thus providing new research directions for the development of new therapeutics and diagnostics.
Researchers also continue to improve the CRISPR/Cas system’s ability to detect nucleic acids at the molecular level rapidly. For example, HOLMES was developed to detect target DNA and RNA rapidly. Interestingly, HOLMES was further improved (HOLMESv2.0) to not only perform simple nucleic acid detection but also specifically differentiate between single nucleotide polymorphisms (SNPs), conveniently quantify target nucleic acids by a one-step system coupled with loop-mediated isothermal amplification (LAMP) at constant temperature, and accurately quantify target DNA methylation by a combination of Cas12b detection and bisulfite treatment [56,76,77]. However, the method relies on pre-amplification, which limits its development.
SHERLOCK, a Cas13a-based nucleic acid-detection platform, was developed to detect target RNA rapidly. In the SHERLOCK platform, a quenched fluorophore is added to the substrate, which is released and, therefore, fluoresces once the substrate cleaves, allowing target RNA detection [64]. Current CRISPR diagnostics typically combine isothermal amplification with CRISPR/Cas-mediated signal amplification and detection via RPA or cyclic LAMP. This combination dramatically improves diagnostic specificity and sensitivity.
To speed up detection, the researchers introduced a pathogen sample-processing method, heating an unextracted diagnostic sample to eliminate nuclease (HUDSON), that allows detection directly from body fluids without nucleic acid extraction. On this basis, SHERLOCK can detect Zika virus (ZIKV) and Dengue virus (DENV) directly from patient samples in 2 h without instrumentation, with detection limits as low as 1 copy/ul [78]. Subsequent research has shown that SHERLOCKv2 may leverage the selectivity of several Cas proteins, such as Cas12 and Cas13, to perform multiplexed nucleic acid detection of at least four distinct targets in a single process [79].
Pena recently reported two novel Cas12 enzymes, SLK9 and SLK5-2 [80], which exhibit high activity at 60 °C. Real-time nucleic acid-detection methods (real-time SLK) can be achieved using LAMP for detection. This method can simultaneously detect SARS-CoV-2 and human-based controls, with a detection limit of 5 copies/mL and a detection time of 30 min [81].
Impressive advances in the field have led scientists to utilize CRISPR/Cas technology creatively as a susceptible diagnostic platform. Quan et al. developed FLASH (Finding Low Abundance Sequences by Hybridization) by exploiting the efficiency, specificity, and programmability of Cas9 [82]. The target sequence is enhanced in the background and utilized for further sequencing using targeted amplification.
CRISPR/Cas-based methods have achieved unprecedented precision in gene editing, and it is important to consider the risks of their potential misuse [83,84]. This is because such misuse may raise ethical and social issues. Sound regulations can prevent misuse, but overly strict measures may hinder the development of CRISPR/Cas technology. Therefore, it is important to strike a balance between avoiding misuse and promoting scientific progress [85].

2.3. CRISPR/Cas System for Non-Nucleic Acid Target Detection

The CRISPR/Cas system has revolutionized gene editing. However, ions, cells, proteins, and small molecules are also biomarkers closely related to life and health. Traditional detection methods are time-consuming, costly, and have low sensitivity and specificity. In contrast, CRISPR/Cas-based sensors can provide inexpensive, rapid, and accurate detection solutions. Introducing this technology not only expands the application field of CRISPR gene-editing technology but also provides a new idea for the susceptible and specific detection of non-nucleic acid targets.
Non-nucleic acid target-detection strategies based on the CRISPR-Dx system typically utilize the trans-cutting activity of CRISPR/Cas proteins for detection. During the detection process, non-nucleic acid targets must be converted into recognizable nucleic acid signals by biotransduction elements that can be taken up by CRISPR/Cas [86]. The use of nucleic acid sequences to activate CRISPR/Cas cleavage of target DNA, along with activation of CRISPR/Cas trans-cleavage activity to produce detectable output signals [87], enables the detection of non-nucleic acid targets (Figure 1).
In 2019, Liang et al. published a small molecule-detection platform named CaT-SMelor, CRISPR/Cas12a and small molecule detector mediated by bacterial ectopic transcription factor (aTF) [88]. By combining the ssDNA cleavage ability of CRISPR/Cas12a and the competitive binding activity of aTFs on small molecules and dsDNA, this high-throughput small-molecule-detection platform can detect small molecules, including uric acid and p-hydroxybenzoic acid and their structurally similar analogs, with a limit of detection down to the nanomolar level.
In 2023, Yee et al. introduced a CRISPR/Cas-based aptasensor for the susceptible and specific detection of the antibiotic agent ampicillin [41]. They developed the aptamer sensor using the CRISPR/Cas system using three different ampicillin-specific aptamers. An ssDNA activator designed according to the aptamers binds to the aptamers by complementary base pairing, and the CRISPR/Cas system is activated by the release of bound ssDNA after the aptamers are attracted to the ampicillin target during detection. It activates the trans-cleavage activity of Cas12a, cleaves the probe, and outputs a fluorescent signal. The detection limit reaches the picomolar level, and the detection time is less than 30 min.
In addition, Wu et al. [89] investigated a new strategy called Nazyme-Activated CRISPR/Cas12a with Circular CRISPR RNA (NA3C), which utilizes cyclic topology of crRNAs to “lock” the CRISPR/Cas12a cis-cleavage and trans-cleavage activity and activates CRISPR/Cas12a cleavage by linearizing the cyclic current in a reaction system using a nuclease with RNA cleavage activity. The use of NA3C for the detection of E. coli in the urine of clinical patients eliminates the need for a culture step. It provides a diagnostic sensitivity of up to 100% and a specificity of 90%.
In addition to metabolites and antibiotics, CRISPR-Dx has also been developed to detect ions, polysaccharides, proteins, cells, transcription factors, etc. (Figure 1) [86]. CRISPR-Dx has the advantages of high specificity, high sensitivity, and programmability; however, unlike nucleic acid targets, non-nucleic acid target detection is currently in the early stages of development, and it still faces many challenges.

3. Application of CRISPR Technology for Antibiotic Detection

3.1. CRISPR/Cas for the Detection of Antibiotic-Resistance Genes

The misuse and improper disposal of antibiotics accelerate the formation of mutant and multidrug-resistant bacteria (MDR) (also known as “superbugs”) [90,91]. The mechanism of antibiotic resistance in bacteria can be attributed to horizontal transfer and vertical transmission of resistance genes on mobile genetic elements (MGEs), such as plasmids, between bacteria [92,93]. Normal bacteria can develop antibiotic resistance through changes in their chromosomes or genetic material [94]. Serious infections and molecular alterations in the host genome may result from this [95]. Effective infection prevention relies on the early identification of antibiotic-resistant bacteria, and rapid detection of specific drug-resistant genes using straightforward techniques is crucial for promptly addressing antibiotic resistance.
Traditional culture methods are the gold standard for bacterial detection, including bacterial culture and plate-counting processes for standard drug sensitivity testing [96]. However, these assays are complex, time-consuming, labor-intensive, and have relatively low sensitivity. With the discovery of an increasing number of antibiotic-resistance genes in both Gram-negative and Gram-positive bacteria, gene-based assays have emerged [97,98]. Commonly used assays are primarily based on polymerase chain reaction (PCR) [99]. Although sensitive and fast, PCR is prone to nonspecific amplification during thermal cycling, leading to false positive results and low specificity. Therefore, there is an urgent need to develop a method that is simple, rapid, and specific for detecting antibiotic-resistance genes.

3.1.1. Cas9 for the Detection of Drug-Resistance Genes

The use of CRISPR/Cas for gene editing has undergone exponential growth over the past few years. It has been rapidly used for genome editing in a variety of various cell types and experimental platforms [100], with some studies suggesting a greater potential for targeting ARGs. The rapid increase in antibiotic-resistant bacteria poses a significant threat to human health. It has the potential to undermine most of the gains of modern medicine in the near future [101]. Rapid and sensitive diagnosis of resistance can help healthcare professionals administer appropriate treatment directly, use existing antibiotics more effectively, and avoid the use of “last resort” antibiotics [102]. Therefore, a method for rapid and accurate detection of drug-resistance genes is important for the development of modern medicine.
Since the characterization of the Cas9 protein from the Streptococcus pyogenes in 2012 [103], the exquisite and programmable specificity of the CRISPR system has inspired many new uses of its enzyme beyond genome engineering. There are also more applications in antibiotic detection, which is in full swing in detecting drug-resistant genes and their use in detecting bacterial pathogens (Table 2).
Müller et al. introduced a method for optical DNA mapping using a single plasmid within a nanofluidic channel [104]. This approach uses the cleavage capability of Cas9 to convert the circular plasmid into a linear form to identify resistance genes (Figure 2A). CRISPR/Cas9 is equipped with a gRNA to improve gene identification, which can be linearized at a particular location on the circular plasmid and subsequently identified by optical DNA mapping. In the research process, the researchers discussed optimizing this detection method for future clinical applications by combining multiple gRNAs that target different genes associated with resistance to the same class of antibiotics.
The assay is suitable for low sample concentrations, and can reveal as much information as possible in a single experiment. At the same time, the assay can measure the size of each plasmid, provide a fingerprint that can be used to identify and track the plasmid and determine the presence or absence of the resistance gene of interest on the plasmid. However, the comprehensive information obtained would require several different techniques and take up to a week to complete, so the method needs to be further developed for future applications.
Using the principles of this technique, Nyblom et al. identified Escherichia coli and Klebsiella pneumoniae directly from patient samples at the strain level [105]. They identified strains or subtypes simultaneously and characterized the corresponding plasmids by restricting antibiotic-resistance genes from the targeted plasmids with Cas9. This optical DNA-profiling assay can rapidly provide comprehensive diagnostic information to optimize early antibiotic treatment regimens and open the way for future precision medicine management.
In 2019, Quan et al. developed a CRISPR/Cas9-based detection technology called FLASH. Due to the high sensitivity and specificity of Cas9, this technology can identify antimicrobial resistance genes in various clinical samples, with a detection limit of 1.9aM [82]. FLASH can reveal the sequence identity of the target DNA, which is an advantage over other detection platforms.
In 2023, Qin et al. developed a novel technique for identifying antibiotic-resistance genes using the CRISPR/Cas9-induced isothermal exponential amplification reaction (IEXPAR) [106]. In the reaction system, antibiotic-resistance genes can be identified by CRISPR/Cas9 and cleaved into two short fragments with free 3ʹ-OH end. The utilization of a cleaved DNA template initiates efficient exponential amplification of IEXPAR, which directly identifies antibiotic-resistance genes under isothermal conditions (Figure 2C). This detection is rapid and precise. As for antibiotic-sensitive bacteria, due to the absence of antibiotic-resistant genes, the CRISPR/Cas9 system cannot cleave any DNA, and no subsequent amplification reaction occurs, ensuring the high specificity of the method. In the experiment, after about 30 min of amplification, antibiotic-resistant genes as low as 100 fM could be sensitively detected, with a detection limit of 81 fM. The method was also applied to the detection of antibiotic-resistant bacteria in actual biological samples. Antibiotic-resistant and antibiotic-sensitive bacteria could be identified under isothermal conditions, and the operation was simple.

3.1.2. Cas12 for the Detection of Drug-Resistance Genes

CRISPR/Cas12a is widely used for genome editing due to its ability to perform precise double-stranded DNA cleavage. Curti et al. constructed a CRISPR/Cas12a-based assay platform [107] in which DNA target sequences correspond to carbapenemase-resistance genes. The system detected each target on a picomolar scale in 10 min. Furthermore, considering that the background of genomic DNA (gDNA) and low levels of free targets in blood can lead to inhibition of reaction, the researchers’ strategy of combining RPA and CRISPR/Cas12 allowed the detection of carbapenem-resistance genes, specifically Klebsiella pneumoniae carbapenemase (KPC), New Delhi metallo β-lactamase (NDM), in less than an hour. The sensitivity and accuracy were comparable to that of RT- qPCR. Validation was carried out using portable test strips with a 100% correlation between the fluorescence test and the portable test strips.
The development of superbugs, a serious threat to the health of all living things, may be accelerated by the accumulation and dissemination of ARB in the environment. Chen et al. created a colorimetric method for identifying ARGs by combining the Au-Fe3O4 nanozyme with the CRISPR/Cas12a nucleic acid-specific recognition capability [108]. The trans-cleavage activity of the CRISPR/Cas12a system in the research system is triggered by the recognition of the target resistance gene. This results in the release of the Au-Fe3O4 nanozyme, the oxidation of 3,3,5,5-tetramethylbenzidine (TMB), and a change in the color of the solution from transparent to blue (Figure 2B). Diagnostic signals can be recorded and analyzed using a smartphone and the signal recognition terminal. Chloramphenicol, ampicillin, and kanamycin-resistance genes can be found using this technique. Due to its high sensitivity (less than 0.1 CFU/μL) and speed (less than 1 h), this detection technique enables the quick and precise identification of ARB or ARG in the field, enabling flexible and efficient monitoring and management of antibiotic contamination [109].
In 2023, Kasputis et al. also developed a CRISPR/Cas12a-based assay [110] to detect ARGs in wash water collected from food-processing plants. DNA-functionalized AuNPs were crosslinked during the assay with ssDNA cross-linker. Degradation of the cross-linker using Cas12a trans-cleavage activity alters the optical properties of AuNPs to produce a simple visual readout (Figure 2D). This assay can still efficiently detect three representative ARGs without DNA amplification with a detection limit of 5 nM or less.
Mao et al. combined CRISPR/Cas12a and LAMP to construct a portable biosensor [111]. For this purpose, primer sets and detection systems were also specially designed, and finally, the output signals were analyzed using fluorescence and lateral flow. Typical ARG ermB was used as the target in the study, and the detection limit was as low as 2.75 × 103 copies/μL. This simple-to-operate and low-cost biosensor is of significant use for large urban areas with many wastewater-treatment plants and rural regions with limited resources. It also provides a new treatment method for detecting ARGs in wastewater.
ARB pose a significant threat to global health, with bacteria producing NDM being particularly problematic because they are resistant to most beta-lactam antibiotics. In this regard, Shin et al. proposed a fluorescent assay based on PCR-coupled CRISPR/Cas12a that can detect NDM genes (blaNDM) produced in bacteria [112]. Thanks to its self-designed gRNA, this CRISPR/Cas12a system can efficiently cleave both the PCR amplification product and the fluorescent probe simultaneously, thus generating a fluorescent signal. The detection performance of this method is excellent, with a detection limit of 2.7 CFU/mL, which is 100 times higher than that of the conventional gel electrophoresis PCR method. Furthermore, this assay detects ARGs in food samples and performs better than previously published quantitative fluorescent quantitative PCR assays.

3.1.3. Cas13 and Others

In recent years, the CRISPR/Cas system has been rapidly developed for pathogen nucleic acid detection. In 2017, Zhang’s team established the SHERLOCK molecular diagnostic platform for the first time by combining the CRISPR/Cas13a system with isothermal amplification technology [113]. On the basis of this, researchers have established a series of highly sensitive and specific nucleic acid-detection methods for various viruses and bacteria by combining different amplification and visualization detection techniques.
In response to the fact that current drug-resistance gene detection usually relies on specialized testing facilities and equipment, rapid detection technology for drug-resistance genes still needs to be developed. Qiang Hu et al. developed a CRISPR/Cas13-based rapid detection technique for the rapid identification of the mecA-resistance gene in Staphylococcus aureus [114]. The researchers developed the recombinase-aided amplification (RAA) primers and crRNA, subsequently employing the easy read-out and sensitive enhanced (ERASE) technology to identify the mecA gene. The method demonstrates a minimum detection limit of 10 copies/μL and achieves 100% concordance with the results of drug susceptibility tests and qPCR detection. The detection platform established by Yan et al. using the same method, named SHIELD (easy-read H. pylori easy-read dual detection) [115], was used to detect the clarithromycin-resistance gene. The detection limit can reach 50 copies/μL.
Both of the above are based on CRISPR technology and, simultaneously, combined with ERASE nucleic acid-detection test strips, reducing dependence on specialized testing instruments and equipment and allowing for more convenient and quicker detection of bacterial drug-resistance genes. It has excellent development prospects to prevent the spread of ARB and to monitor the progress of bacterial drug resistance in real-time. However, this method is the same as most current bacterial nucleic acid-detection methods, which require the extraction of nucleic acids from bacterial samples prior to detection, and the current rapid extraction of bacterial nucleic acids usually relies on equipment such as centrifuges and magnetic racks or requires the purchase of the corresponding nucleic acid extraction kits. This strategy not only increases the complexity of the testing process but also increases the cost of testing. In contrast, Ortiz-Cartagena et al. used a new assay based on LAMP & CRISPR/Cas13a for the detection of carbapenem-resistant genes in clinical samples [116], which has high specificity and sensitivity, can be performed without the need for specialized methods for RNA extraction, and shows 100% accuracy for the detection of drug-resistant genes. Furthermore, it does not require specific equipment or trained personnel, and the sample per reaction is small, making it convenient and inexpensive. Therefore, this CRISPR/Cas-based assay kit will be highly competitive in the commercial market in the future.
Due to the widespread use of antibiotics, bacteria have developed various defense methods, such as restriction-modification systems and CRISPR/Cas mechanisms. As the innate and adaptive immune systems of bacteria, these two mechanisms have a significant impact on the spread of ARGs. They are considered inhibitors of horizontal gene transfer in bacteria [117,118,119]. Pathogens with the CRISPR/Cas system have been found to be less likely to carry ARGs than pathogens lacking this defense system [120], a finding that provides new strategies for combating the spread of ABR.
Figure 2. The CRISPR/Cas system-based detection platform for antibiotic-resistance genes. (A) Specific DNA was cut using the Cas9 enzyme, the fluorescent dye was added, and then the plasmid was stretched in the nano-channel, and the information was observed by fluorescence microscope. Copyright Vilhelm Müller. (B) The resistance gene fragment was amplified by RPA technology to activate the CRISPR/Cas12a system, followed by colorimetric detection using Au-Fe3O4 nanoenzymes.(* p < 0.05, ** p < 0.01) Reproduced with permission from Ref. [108]. Copyright 2023 Elsevier. (C) CRISPR/Cas9 recognizes and cleaves drug-resistant genes and activates IEXPAR amplification for rapid detection. Reproduced with permission from Ref. [106]. Copyright 2022 Elsevier. (D) The presence of target ARGs activates CRISPR/Cas12a to degrade the crosslinker, making the solution red. Without target genes, it turns purple due to the undegraded crosslinker. Reproduced with permission from Ref. [110]. Copyright 2023 American Chemical Society.
Figure 2. The CRISPR/Cas system-based detection platform for antibiotic-resistance genes. (A) Specific DNA was cut using the Cas9 enzyme, the fluorescent dye was added, and then the plasmid was stretched in the nano-channel, and the information was observed by fluorescence microscope. Copyright Vilhelm Müller. (B) The resistance gene fragment was amplified by RPA technology to activate the CRISPR/Cas12a system, followed by colorimetric detection using Au-Fe3O4 nanoenzymes.(* p < 0.05, ** p < 0.01) Reproduced with permission from Ref. [108]. Copyright 2023 Elsevier. (C) CRISPR/Cas9 recognizes and cleaves drug-resistant genes and activates IEXPAR amplification for rapid detection. Reproduced with permission from Ref. [106]. Copyright 2022 Elsevier. (D) The presence of target ARGs activates CRISPR/Cas12a to degrade the crosslinker, making the solution red. Without target genes, it turns purple due to the undegraded crosslinker. Reproduced with permission from Ref. [110]. Copyright 2023 American Chemical Society.
Biosensors 14 00633 g002
Table 2. CRISPR/Cas systems for the detection of drug-resistance genes.
Table 2. CRISPR/Cas systems for the detection of drug-resistance genes.
Cas EffectorsDetection PlatformWhether to Amplify or Not 1TargetLODTime
(min)
Ref.
CRISPR/Cas9CRISPR/Cas9 combined with optical DNA mapping NESBL gene family blaCTX-M;
The carbapenemase gene families blaNDM and blaKPC
NRNR[104,110]
FLASH Yantimicrobial-resistance genes1.9 aMNR[82]
IEXPARYmecA gene in real genomic DNA samples81 fMNR[106]
CRISPR/dCas9CRISPR/dCas9-SERSNmacrolide antibiotic-resistant
macB gene in milk
11.9 fMNR[121]
CRISPR/Cas12aRPA amplification Ycarbapenemases-resistance genes such as KPC, NDM and OXA100 aM<120[107]
Au-Fe3O4 nanozyme coupled with CRISPR/Cas12aNKana-resistance genes; AMPI-resistance genes;
Chloramphenicol-resistance genes
<0.1 CFU/μL<60[108]
Colorimetric detection based on CRISPR/Cas systemNermB; sul1; tetW5 nM50[110]
Portable biosensor combining CRISPR/Cas12a and LAMPYermB in wastewater2.75 × 103 copies/μL70[111]
RPA coupled with CRISPR/Cas12a platformYColistin
Resistance Gene mcr-1
1.6 × 103 CFU/mL60[122]
Cas12a/3D DNAzyme colorimetric paper sensorYNDM-1 gene encoding metallo-β-lactamase100 fM<90[123]
Cas12a dual detection platform (Cas12a-Ddp) Y (PCR&RPA)mcr-1 and invA genes33/214 fM45/75[124]
CRISPR/Cas12a cou-
pled with PCR
YblaNDM in Carbapenem-Resistant Enterobacterales2.7 CFU/mLNR[112]
CRISPR/Cas13aCRISPR/Cas system Combining ERASEYStaphylococcus aureus mecA-resistance gene10 copies/μLNR[114]
RAA-CRISPR/Cas13a Fluorescence Detection SystemYA2142G and A2143G mutant DNAs causing clarithromycin
resistance
50 copies/μLNR[115]
Combines RPA and CRISPR/Cas13a: one-tube and two-step reactionYmexX gene in P. aeruginosa10 aM/1 aM5/40[125]
RPA-Cas13a assayYblaKPC2.5 copies/μL60[126]
LAMP-CRISPR/Cas13a-based assayYOXA-48
and GES Carbapenemases
NR<120[116]
1 In the third column, Y = Amplify; N = Not Amplify.

3.2. CRISPR/Cas for the Detection of Antibiotic Molecules

Nature has recognized antiretroviral enzymes based on CRISPR/Cas as one of the top seven technologies to watch in 2022 [127]. CRISPR/Cas-based biosensing technology is indeed promising, but currently, it is mainly limited to detecting nucleic acid targets. The development of detection systems for non-nucleic acid targets is still in its infancy, especially in detecting antibiotic molecules with fewer applications.
An elemental probe-based CRISPR/Cas14 detection platform (Figure 3A) for non-nucleic acid targets was first proposed by Hu et al. in 2021 [128]. Metal isotope detection was combined with CRISPR/Cas14 biosensors to detect the non-nucleic acid target ampicillin sensitively. In this study, a fluorescence-quenching pair-labeled probe (FQ) was designed and optimized, and the target nucleic acid could be quantified using the fluorescence signal of the incidentally cleaved FQ fragment. This method quantifies trace ampicillin in aqueous solution in 45 min at room temperature (25 °C) with a detection limit as low as 2.06 nM, which is excellent in anti-interference testing and complex matrix detection.
At the same time, Lv et al. published a metal-labeled CRISPR/Cas12a bioassay, and applied it to an ultra-sensitive and highly selective assessment of antibiotic bioaccumulation in wild fish [43]. The methodology integrates an elementally labeled CRISPR/Cas12a reporter probe with incidental cleavage activity. The target kanamycin was recognized and captured by a “lock activate” system, followed by the release of an activator chain to activate the incidental cleavage activity of Cas12a, which cleaves the free metal reporter (Tm-Rep). Uncleaved probes and cleaved biotin-modified molecules were captured using streptavidin-coated magnetic beads, and the remaining probes were available for quantitative detection of metal isotopes by inductively coupled plasma mass spectrometry (ICPMS). In this study, kanamycin was used as the detection target, and the detection limit was as low as 4.06 pM in 30 min.
Both methods mentioned above utilize the rare elements terbium (159Tb) and thulium (169Tm). However, both 159Tb and 169Tm are rare earth elements with minimal reserves and are war-prepared materials. This limitation could have been more conducive to large-scale replication of the method. The researchers then turned their attention to aptamers, coupling optical and electrochemical sensing platforms with aptamers [19,20] and using RNA or DNA aptamers for specific binding to targeted antibiotics [21] as a way to detect particular antibiotics. Based on this idea, in 2022, Li et al. constructed two ultrasensitive biosensors (sensor-ss and sensor-ds) based on the CRISPR/Cas system [42], which consisted of heterologous aptamer probes integrated with the CRISPR/Cas12a system for the detection of antibiotic tobramycin. Both sensors utilize hairpin DNA containing tobramycin aptamer sequences as target recognition probes and further generate signal-transduction sequences.
The two sensors vary in their target-recognition and signal-amplification methodologies. In sensor-ss (Figure 3B), ssDNA activates the trans-cutting activity of CRISPR/Cas12a, resulting in the cleavage of the FQ probe and an enhanced fluorescence signal. The dsDNA with the PAM sites at the sensor ds functions as a signal-transduction sequence, identified by CRISPR/Cas12a, resulting in the cleavage of the FQ probe. In the signal-amplification strategy, sensor-ss utilized a strand displacement amplification (SDA) reaction for signal augmentation, while sensor-ds did not incorporate a DNA-amplification method. By optimizing the DNA sequence and reaction parameters, the fluorescence response of sensor ds exhibited a linear correlation with the tobramycin concentration (10–300 pM), achieving a limit of detection (LOD) as low as 3.719 pM. Despite sensor ss exhibiting superior sensitivity (LOD = 1.542 pM) compared to sensor ds, rapid attainment of fluorescence signal saturation led to a limited linear response range (5–30 pM). This CRISPR/Cas12a-based assay is visually interpretable under UV light for on-site detection. The fabricated sensor-ds were utilized to identify tobramycin in milk and lake water samples. This target-recognition and signal-amplification technology offers a promising approach for developing a multipurpose sensing platform to detect other non-nucleic acid targets.
With the idea of developing a sensor for the CRISPR/Cas-based on aptamers, in the same year, Mahas et al. introduced an aTF into the CRISPR/Cas system to detect antibiotics [129]. They developed a simple, rapid, sensitive, and field-deployable small molecule-detection platform based on the CRISPR/Cas12a-aTF biosensor for tetracycline. In this biosensor, aTF binds to an operon sequence that hinders the in vitro transcription process. In the presence of tetracycline, the tetracycline binds to the aTF, causing the aTF to be released from the operator sequence. As a result, the in vitro transcription process can proceed normally, resulting in the acquisition of crRNA to activate the CRISPR/Cas12a system and generate fluorescent signals (Figure 3C). This assay platform is a valuable addition to the development of CRISPR/Cas-based, cell-free biosensors with great potential for detecting non-nucleic acid small molecules in situ.
In 2023, Yee et al. developed aptamer sensors (Figure 3D) for the susceptible and specific detection of the antibiotic agent ampicillin by the CRISPR/Cas system using three different ampicillin-specific aptamers [41]. The ssDNA activators bind to aptamers by complementary base pairing. The aptamer attracts the ampicillin target and releases bound ssDNA, activating the CRISPR/Cas system. It activates the trans-cleavage activity of Cas12a, cleaves the probe, and outputs a fluorescent signal. The method can be completed in 30 min with a detection limit of 0.01 nM. The sensor is also sensitive to ampicillin under complex substrates.
Chen et al. developed a portable biosensor detecting kanamycin based on glucometer and CRISPR/Cas12a [130]. With a detection limit of 1 pM, this technique for the on-site and low-cost monitoring of antibiotic residues in water samples using equipment readily available to the public is a significant discovery.
Researchers have recently combined hybridization chain reaction (HCR) with the CRISPR/Cas12a system to construct more flexible detection platforms. For example, Zhu et al. combined NH2-Co-MOF as electrocatalytic active material, HCR, catalytic hairpin assembly (CHA), and CRISPR/Cas 12, a technology to construct a sensitive and label-free electrochemical sensor for the detection of ampicillin [131], with a detection limit of 1.60 pM.
The detection platform constructed by Zhang et al. using the same method (Figure 3E) can reach a detection limit of 60 fM and 10 fM when used to detect the antibiotics kanamycin and ampicillin, respectively [132]. This electrochemical sensor, which uses HCR-triggered CRISPR/Cas12 with NH2-Cu-MOF as the signaling molecule, has considerable advantages. Initially, NH2-Cu-MOF, characterized by high electroactivity and accessible coupling properties, is used as an electrochemical signaling marker without the incorporation of additional redox mediators, thus increasing the output of the electrochemical signal and streamlining the structural complexity of the sensing system; subsequently, HCR was integrated with CRISPR/Cas12a to establish a cascade amplification circuit, which enhances the efficiency and sensitivity of the signal amplification of the sensing system. We summarized the reports published in recent years on the use of CRISPR/Cas for molecular detection of antibiotics (Table 3) and compared the sensitivity of the different detection methods.
Figure 3. CRISPR/Cas-based detection platform for antibiotics. (A) The AMP aptamer releases an activator upon target binding, activating Cas14 to cleave a probe, and AMP concentration is quantified by ICPMS detection of terbium isotope intensity. Used with permission of © The Royal Society of Chemistry 2021, from [128]. Permission conveyed through Copyright Clearance Center. (B) Tobramycin-bound aptamer triggers SDA to generate ssDNA activators that activate CRISPR/Cas12a to cleave reporter probes and output signals. Reproduced with permission from Ref. [42]. Copyright 2021 Elsevier. (C) In the presence of the ligand, dissociation of the aTF allows transcription of the CRISPR array, activating CRISPR/Cas12a and cleaving the probe, outputting a signal. Copyright©2022 Ahmed Mahas. Published by American Chemical Society. This publication is licensed under CC-BY 4.0. (D) The aptamer recognizes ampicillin and releases the ssDNA activator, which activates the trans-cleavage of Cas12a and outputs a fluorescent signal. Reproduced with permission from Ref. [41]. Copyright 2023 Elsevier. (E) Without kanamycin, S1 triggers HCR1, generating a strong electrical signal. In the presence of kanamycin, activation of CRISPR/Cas12a blocks HCR1 and reduces the electrical signal. Reproduced with permission from Ref. [132]. Copyright 2024 Elsevier.
Figure 3. CRISPR/Cas-based detection platform for antibiotics. (A) The AMP aptamer releases an activator upon target binding, activating Cas14 to cleave a probe, and AMP concentration is quantified by ICPMS detection of terbium isotope intensity. Used with permission of © The Royal Society of Chemistry 2021, from [128]. Permission conveyed through Copyright Clearance Center. (B) Tobramycin-bound aptamer triggers SDA to generate ssDNA activators that activate CRISPR/Cas12a to cleave reporter probes and output signals. Reproduced with permission from Ref. [42]. Copyright 2021 Elsevier. (C) In the presence of the ligand, dissociation of the aTF allows transcription of the CRISPR array, activating CRISPR/Cas12a and cleaving the probe, outputting a signal. Copyright©2022 Ahmed Mahas. Published by American Chemical Society. This publication is licensed under CC-BY 4.0. (D) The aptamer recognizes ampicillin and releases the ssDNA activator, which activates the trans-cleavage of Cas12a and outputs a fluorescent signal. Reproduced with permission from Ref. [41]. Copyright 2023 Elsevier. (E) Without kanamycin, S1 triggers HCR1, generating a strong electrical signal. In the presence of kanamycin, activation of CRISPR/Cas12a blocks HCR1 and reduces the electrical signal. Reproduced with permission from Ref. [132]. Copyright 2024 Elsevier.
Biosensors 14 00633 g003

4. Summary and Prospects

The emergence of the CRISPR/Cas system has revolutionized gene-editing technology and reshaped the landscape in the field of analysis and assays. The CRISPR/Cas system has rapidly evolved and is used to design powerful molecular diagnostic tools. CRISPR/Cas-based biosensors outperform traditional techniques in terms of accuracy, versatility, portability, timeliness, and efficiency due to specific target recognition and cis/trans cleavage activity. Therefore, by combining novel materials, novel methods, and novel sensing technologies, CRISPR/Cas biosensing systems are expected to satisfy a variety of needs and contribute to a number of domains, including environmental analysis, food safety testing, and disease detection.
However, the current technology platform based on the CRISPR/Cas system for antibiotic detection still faces several problems to be solved: (1) Most methods need to be combined with nucleic acid-amplification techniques (such as RPA, LAMP, etc.) to improve detection sensitivity. However, this process still needs to be performed step by step, which introduces the risk of contamination and dramatically increases detection time and workload. Although studies have been conducted to develop amplification-free nucleic acid-detection platforms, there are still problems, such as low detection limits. Integrated, fully enclosed microfluidic chips and devices that integrate isothermal amplification and signal output should be further explored to improve their clinical applications. (2) PAM sequence limitation. Most of the target sequences recognized by Cas12 are highly dependent on PAM sequences, limiting its detection’s versatility. In addition to introducing PAM sequences using amplification primers, more Cas proteins that are not strictly dependent on PAM sites should be identified to increase the flexibility of the assay. (3) False positive results. False positive results occur due to off-target cleavage by binding crRNA to nontarget sequences. The accuracy of the detection results can be ensured by the careful design of the crRNA to improve the target-cutting effect. (4) High-throughput multi-target detection. Most detection platforms based on the CRISPR/Cas system can only achieve qualitative detection of a single marker for a single sample, and it is challenging to realize multi-target quantitative detection for multiple samples. Based on the close correlation between the concentration of antibiotics and drug-resistant genes and the sensing strength of the CRISPR/Cas system-detection signals, the combination of microfluidics, microdroplets, microarrays, and other technologies with optical sensors and other ultrasensitive detection technologies to build a new type of detection platform will promote the solution of this problem. (5) The detection system should be standardized. The current CRISPR/Cas-based detection technology has not yet been formally standardized, and no approved products have been approved However, this situation will change with the growing maturity of this detection technology.
In summary, this paper points out the current development, challenges, and future directions of the CRISPR/Cas toolbox and its use in antibiotic detection for nucleic acid and non-nucleic acid target detection. The functionality and detection utility of the CRISPR/Cas system, especially its significant applications in antibiotic drug detection, are mainly introduced. A thorough overview of the CRISPR/Cas-based antibiotic-detection technique has not yet been published. This review provides a comprehensive overview of the development of this new technology in detecting nucleic acid and non-nucleic acid substances, focusing on its application in detecting antibiotic-resistance genes and antibiotic molecules. It provides a reference for controlling and reducing the expansion of antibiotic resistance.

Author Contributions

Conceptualization, X.Z. and Q.T.; methodology, X.Z., Z.H. and W.W.; software, X.Z., Z.H. and Y.Z.; validation, X.Z., Z.H. and Z.Y.; formal analysis, X.Z., Z.H. and Q.T.; investigation, X.Z., Q.T. and K.S.; data curation, X.Z., Z.H. and X.Y.; writing—original draft preparation, X.Z., Z.H. and Q.T.; writing—review and editing, X.Z., X.Y., W.K. and P.L.; supervision, X.Y.; project administration, X.Y. and Q.T.; funding acquisition, X.Y., Q.T. and K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the “Pioneer” and “Leading Goose” R&D Program of Zhejiang (2022C02049); Zhejiang Provincial Department of Agriculture and Rural Affairs Project (2024SNJF044); Zhejiang Provincial Department of Agriculture and Rural Affairs Project (2023SNJF066).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wright, G.D. The antibiotic resistome. Expert Opin. Drug Discov. 2010, 5, 779–788. [Google Scholar] [CrossRef] [PubMed]
  2. Yu, W.; Xu, Y.; Wang, Y.; Sui, Q.; Xin, Y.; Wang, H.; Zhang, J.; Zhong, H.; Wei, Y. An extensive assessment of seasonal rainfall on intracellular and extracellular antibiotic resistance genes in Urban River systems. J. Hazard. Mater. 2023, 455, 131561. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, Y.R.; Duan, Y.P.; Zhang, Z.B.; Gao, Y.F.; Dai, C.M.; Tu, Y.J.; Gao, J. Comprehensive evaluation of antibiotics pollution the Yangtze River basin, China: Emission, multimedia fate and risk assessment. J. Hazard. Mater. 2024, 465, 133247. [Google Scholar] [CrossRef]
  4. Antimicrobial Additives Market to Reach $5.63 Billion By 2030. Available online: https://www.grandviewresearch.com/press-release/global-antimicrobial-additives-market (accessed on 31 January 2024).
  5. Su, H.-C.; Liu, Y.-S.; Pan, C.-G.; Chen, J.; He, L.-Y.; Ying, G.-G. Persistence of antibiotic resistance genes and bacterial community changes in drinking water treatment system: From drinking water source to tap water. Sci. Total Environ. 2018, 616–617, 453–461. [Google Scholar] [CrossRef] [PubMed]
  6. Martinez, J.L. Environmental pollution by antibiotics and by antibiotic resistance determinants. Environ. Pollut. 2009, 157, 2893–2902. [Google Scholar] [CrossRef]
  7. Mestrovic, T.; Aguilar, G.R.; Swetschinski, L.R.; Ikuta, K.S.; Gray, A.P.; Weaver, N.D.; Han, C.; Wool, E.E.; Hayoon, A.G.; Hay, S.I.; et al. The burden of bacterial antimicrobial resistance in the WHO European region in 2019: A cross-country systematic analysis. Lancet Public Health 2022, 7, e897–e913. [Google Scholar] [CrossRef] [PubMed]
  8. Tan, Q.; Li, W.; Zhang, J.; Zhou, W.; Chen, J.; Li, Y.; Ma, J. Presence, dissemination and removal of antibiotic resistant bacteria and antibiotic resistance genes in urban drinking water system: A review. Front. Environ. Sci. Eng. 2019, 13, 1–15. [Google Scholar] [CrossRef]
  9. Rice, L.B. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: No ESKAPE. J. Infect. Dis. 2008, 197, 1079–1081. [Google Scholar] [CrossRef]
  10. WHO Updates List of Drug-Resistant Bacteria Most Threatening to Human Health. Available online: https://www.who.int/publications/i/item/9789240093461 (accessed on 17 May 2024).
  11. Hoffman, P.S. Antibacterial Discovery: 21st Century Challenges. Antibiotics 2020, 9, 213. [Google Scholar] [CrossRef]
  12. Wong, F.; Zheng, E.J.; Valeri, J.A.; Donghia, N.M.; Anahtar, M.N.; Omori, S.; Li, A.; Cubillos-Ruiz, A.; Krishnan, A.; Jin, W.; et al. Discovery of a structural class of antibiotics with explainable deep learning. Nature 2023, 626, 177–185. [Google Scholar] [CrossRef] [PubMed]
  13. Ebrahim, A.; Chandrima, B.; Ana, B.G.; Eduardo, C.N.; Youping, D.; Christelle, D.; Emmanuel, D.N.; Eran, E.; Gregorio, I.; Soojin, J.; et al. COVID-19 drug practices risk antimicrobial resistance evolution. Lancet Microbe 2021, 2, e135–e136. [Google Scholar]
  14. Baljit, S.; Abhijnan, B.; Lesa, D.; Riya, P.K.; Yaroslav, K.; Isha, D. Electrochemical Biosensors for the Detection of Antibiotics in Milk: Recent Trends and Future Perspectives. Biosensors 2023, 13, 867. [Google Scholar] [CrossRef]
  15. Liu, K.; Gan, C.; Peng, Y.; Gan, Y.; He, J.; Du, Y.; Tong, L.; Shi, J.; Wang, Y. Occurrence and source identification of antibiotics and antibiotic resistance genes in groundwater surrounding urban hospitals. J. Hazard. Mater. 2023, 465, 133368. [Google Scholar] [CrossRef]
  16. Magdalena, M.; Martyna, B.-H.; Łukasz, P.; Mateusz, M.; Izabela, W.; Monika, H.; Ewa, K. Poultry manure-derived microorganisms as a reservoir and source of antibiotic resistance genes transferred to soil autochthonous microorganisms. J. Environ. Manag. 2023, 348, 119303. [Google Scholar]
  17. Wanyan, R.; Pan, M.; Mai, Z.; Xiong, X.; Wang, S.; Han, Q.; Yu, Q.; Wang, G.; Wu, S.; Li, H. Fate of high-risk antibiotic resistance genes in large-scale aquaculture sediments: Geographical differentiation and corresponding drivers. Sci. Total Environ. 2023, 905, 167068. [Google Scholar] [CrossRef]
  18. Bueno, I.; Williams-Nguyen, J.; Hwang, H.; Sargeant, J.M.; Nault, A.J.; Singer, R.S. Impact of point sources on antibiotic resistance genes in the natural environment: A systematic review of the evidence. Anim. Health Res. Rev. 2017, 18, 112–127. [Google Scholar] [CrossRef] [PubMed]
  19. Maximilian, S.; Anton, S.; Markus, F.; Christian, L.; Stefan, H.; Oliver, S. A liquid chromatography-tandem mass spectrometry method for the quantification of ampicillin/sulbactam and clindamycin in jawbone, plasma, and platelet-rich fibrin: Application to patients with osteonecrosis of the jaw. J. Pharm. Biomed. Anal. 2022, 224, 115167. [Google Scholar]
  20. Jank, L.; Martins, M.T.; Arsand, J.B.; Motta, T.M.C.; Hoff, R.B.; Barreto, F.; Pizzolato, T.M. High-throughput method for macrolides and lincosamides antibiotics residues analysis in milk and muscle using a simple liquid–liquid extraction technique and liquid chromatography–electrospray–tandem mass spectrometry analysis (LC–MS/MS). Talanta 2015, 144, 686–695. [Google Scholar] [CrossRef] [PubMed]
  21. Moudgil, P.; Bedi, J.S.; Aulakh, R.S.; Gill, J.P.S.; Kumar, A. Validation of HPLC Multi-residue Method for Determination of Fluoroquinolones, Tetracycline, Sulphonamides and Chloramphenicol Residues in Bovine Milk. Food Anal. Methods 2019, 12, 338–346. [Google Scholar] [CrossRef]
  22. Saleh, H.; Elhenawee, M.; Hussien, E.M.; Ahmed, N.; Ibrahim, A.E. Validation of HPLC-UV Multi-Residue Method for the Simultaneous Determination of Tetracycline, Oxytetracycline, Spiramycin and Neospiramycin in Raw Milk. Food Anal. Methods 2020, 14, 36–43. [Google Scholar] [CrossRef]
  23. Dou, X.; Wu, Q.; Luo, S.; Yang, J.; Dong, B.; Wang, L.; Qu, H.; Zheng, L. A miniaturized biosensor for rapid detection of tetracycline based on a graphene field-effect transistor with an aptamer modified gate. Talanta 2024, 271, 125702. [Google Scholar] [CrossRef] [PubMed]
  24. Wei, X.; Liu, C.; Qin, H.; Ye, Z.; Liu, X.; Zong, B.; Li, Z.; Mao, S. Fast, specific, and ultrasensitive antibiotic residue detection by monolayer WS2-based field-effect transistor sensor. J. Hazard. Mater. 2022, 443, 130299. [Google Scholar] [CrossRef]
  25. Huang, Y.H.; Wei, H.; Santiago, P.J.; Thrift, W.J.; Ragan, R.; Jiang, S. Sensing Antibiotics in Wastewater Using Surface-Enhanced Raman Scattering. Environ. Sci. Technol. 2023, 57, 4880–4891. [Google Scholar] [CrossRef] [PubMed]
  26. Ahmed, S.; Ning, J.; Peng, D.; Chen, T.; Ahmad, I.; Ali, A.; Lei, Z.; Abu bakr Shabbir, M.; Cheng, G.; Yuan, Z. Current advances in immunoassays for the detection of antibiotics residues: A review. Food Agric. Immunol. 2020, 31, 268–290. [Google Scholar] [CrossRef]
  27. Kumaran, A.; Jude Serpes, N.; Gupta, T.; James, A.; Sharma, A.; Kumar, D.; Nagraik, R.; Kumar, V.; Pandey, S. Advancements in CRISPR-Based Biosensing for Next-Gen Point of Care Diagnostic Application. Biosensors 2023, 13, 202. [Google Scholar] [CrossRef] [PubMed]
  28. Lu, N.; Chen, J.; Rao, Z.; Guo, B.; Xu, Y. Recent Advances of Biosensors for Detection of Multiple Antibiotics. Biosensors 2023, 13, 850. [Google Scholar] [CrossRef] [PubMed]
  29. Manickam, P.; Mariappan, S.A.; Murugesan, S.M.; Hansda, S.; Kaushik, A.; Shinde, R.; Thipperudraswamy, S.P. Artificial Intelligence (AI) and Internet of Medical Things (IoMT) Assisted Biomedical Systems for Intelligent Healthcare. Biosensors 2022, 12, 562. [Google Scholar] [CrossRef] [PubMed]
  30. Garg, P.; Gupta, R.; Priyadarshi, N.; Sagar, P.; Bisht, V.; Navani, N.K.; Singhal, N.K. Emerging trends: Smartphone-assisted aptasensors enabling detection of pathogen and chemical contamination. Microchem. J. 2024, 207, 111736. [Google Scholar] [CrossRef]
  31. Zhu, C.; Zhang, F.; Li, H.; Chen, Z.; Yan, M.; Li, L.; Qu, F. CRISPR/Cas systems accelerating the development of aptasensors. Trends Anal. Chem. 2023, 158, 116775. [Google Scholar] [CrossRef]
  32. Zein, M.I.; Hardianto, A.; Irkham, I.; Zakiyyah, S.N.; Devi, M.J.; Manan, N.S.; Ibrahim, A.U.; Hartati, Y.W. Recent development of electrochemical and optical aptasensors for detection of antibiotics in food monitoring applications. J. Food Compos. Anal. 2023, 124, 105644. [Google Scholar] [CrossRef]
  33. Hu, M.; Yue, F.; Dong, J.; Tao, C.; Bai, M.; Liu, M.; Zhai, S.; Chen, S.; Liu, W.; Qi, G.; et al. Screening of broad-spectrum aptamer and development of electrochemical aptasensor for simultaneous detection of penicillin antibiotics in milk. Talanta 2024, 269, 125508. [Google Scholar] [CrossRef]
  34. Wang, X.; Li, J.; Jian, D.; Zhang, Y.; Shan, Y.; Wang, S.; Liu, F. Paper-based antibiotic sensor (PAS) relying on colorimetric indirect competitive enzyme-linked immunosorbent assay for quantitative tetracycline and chloramphenicol detection. Sens. Actuators B Chem. 2020, 329, 129173. [Google Scholar] [CrossRef]
  35. Hu, W.; Xia, L.; Hu, Y.; Li, G. Fe3O4-carboxyl modified AuNPs-chitosan@AgNPs as a robust surface-enhanced Raman scattering substrate for rapid analysis of tryptamine and ofloxacin in aquatic products. Talanta 2024, 266, 125057. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, Y.; Zou, M.; Chen, Y.; Tang, F.; Dai, J.; Jin, Y.; Wang, C.; Xue, F. Ultrasensitive and selective detection of sulfamethazine in milk via a Janus-labeled Au nanoparticle-based surface-enhanced Raman scattering-immunochromatographic assay. Talanta 2023, 267, 125208. [Google Scholar] [CrossRef]
  37. Wang, X.; Li, Q.; Zong, B.; Fang, X.; Liu, M.; Li, Z.; Mao, S.; Ostrikov, K.K. Discriminative and quantitative color-coding analysis of fluoroquinolones with dual-emitting lanthanide metal-organic frameworks. Sens. Actuators B Chem. 2022, 373, 132701. [Google Scholar] [CrossRef]
  38. Varsha, M.V.; Nageswaran, G. Ruthenium doped Cu-MOF as an efficient sensing platform for the voltammetric detection of ciprofloxacin. Microchem. J. 2023, 188, 108481. [Google Scholar] [CrossRef]
  39. Wang, B.B.; Zhao, X.; Chen, L.J.; Yang, C.; Yan, X.P. Functionalized Persistent Luminescence Nanoparticle-Based Aptasensor for Autofluorescence-free Determination of Kanamycin in Food Samples. Anal. Chem. 2021, 93, 2589–2595. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, X.; Xu, N.-Y.; Ruan, Q.; Lu, D.-Q.; Yang, Y.-H.; Hu, R. A label-free and sensitive photoluminescence sensing platform based on long persistent luminescence nanoparticles for the determination of antibiotics and 2,4,6-trinitrophenol. RSC Adv. 2018, 8, 5714–5720. [Google Scholar] [CrossRef]
  41. Yee, B.J.; Shafiqah, N.F.; Mohd-Naim, N.F.; Ahmed, M.U. A CRISPR/Cas12a-based fluorescence aptasensor for the rapid and sensitive detection of ampicillin. Int. J. Biol. Macromol. 2023, 242, 125211. [Google Scholar] [CrossRef] [PubMed]
  42. Li, D.; Ling, S.; Wu, H.; Yang, Z.; Lv, B. CRISPR/Cas12a-based biosensors for ultrasensitive tobramycin detection with single- and double-stranded DNA activators. Sens. Actuators B Chem. 2022, 355, 131329. [Google Scholar] [CrossRef]
  43. Hu, J.; Song, H.; Zhou, J.; Liu, R.; Lv, Y. Metal-Tagged CRISPR/Cas12a Bioassay Enables Ultrasensitive and Highly Selective Evaluation of Kanamycin Bioaccumulation in Fish Samples. Anal. Chem. 2021, 93, 14214–14222. [Google Scholar] [CrossRef] [PubMed]
  44. He, L.; Shen, Z.; Cao, Y.; Li, T.; Wu, D.; Dong, Y.; Gan, N. A microfluidic chip based ratiometric aptasensor for antibiotic detection in foods using stir bar assisted sorptive extraction and rolling circle amplification. Analyst 2019, 144, 2755–2764. [Google Scholar] [CrossRef] [PubMed]
  45. Makarova, K.S.; Wolf, Y.I.; Iranzo, J.; Shmakov, S.A.; Alkhnbashi, O.S.; Brouns, S.J.; Charpentier, E.; Cheng, D.; Haft, D.H.; Horvath, P.; et al. Evolutionary classification of CRISPR-Cas systems: A burst of class 2 and derived variants. Nat. Rev. Microbiol. 2020, 18, 67–83. [Google Scholar] [CrossRef]
  46. Bhatia, S.; Yadav, S.K. CRISPR-Cas for genome editing: Classification, mechanism, designing and applications. Int. J. Biol. Macromol. 2023, 238, 124054. [Google Scholar] [CrossRef]
  47. Chylinski, K.; Le Rhun, A.; Charpentier, E. The tracrRNA and Cas9 families of type II CRISPR-Cas immunity systems. RNA Biol. 2013, 10, 726–737. [Google Scholar] [CrossRef]
  48. Zetsche, B.; Gootenberg, J.S.; Abudayyeh, O.O.; Slaymaker, I.M.; Makarova, K.S.; Essletzbichler, P.; Volz, S.E.; Joung, J.; Oost, J.v.d.; Regev, A.; et al. Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System. Cell 2015, 163, 759–771. [Google Scholar] [CrossRef] [PubMed]
  49. Abudayyeh, O.O.; Gootenberg, J.S.; Konermann, S.; Joung, J.; Slaymaker, I.M.; Cox, D.B.T.; Shmakov, S.; Makarova, K.S.; Semenova, E.; Minakhin, L.; et al. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science 2016, 353, aaf5573. [Google Scholar] [CrossRef] [PubMed]
  50. Wu, H.; Chen, X.; Zhang, M.; Wang, X.; Chen, Y.; Qian, C.; Wu, J.; Xu, J. Versatile detection with CRISPR/Cas system from applications to challenges. Trends Anal. Chem. 2021, 135, 116150. [Google Scholar] [CrossRef]
  51. Chen, J.S.; Ma, E.; Harrington, L.B.; Costa, M.D.; Tian, X.; Palefsky, J.M.; Doudna, J.A. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science 2018, 360, 436–439. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, T.; Wei, J.J.; Sabatini, D.M.; Lander, E.S. Genetic Screens in Human Cells Using the CRISPR-Cas9 System. Science 2014, 343, 80–84. [Google Scholar] [CrossRef]
  53. Sun, W.; Huang, X.; Wang, X. CRISPR-based molecular diagnostics: A review. Chin. J. Biotechnol. 2023, 39, 60–73. [Google Scholar]
  54. Pardee, K.; Green, A.A.; Takahashi, M.K.; Braff, D.; Lambert, G.; Lee, J.W.; Ferrante, T.; Ma, D.; Donghia, N.; Fan, M.; et al. Rapid, Low-Cost Detection of Zika Virus Using Programmable Biomolecular Components. Cell 2016, 165, 1255–1266. [Google Scholar] [CrossRef] [PubMed]
  55. Yan, W.X.; Hunnewell, P.; Alfonse, L.E.; Carte, J.M.; Keston-Smith, E.; Sothiselvam, S.; Garrity, A.J.; Chong, S.; Makarova, K.S.; Koonin, E.V.; et al. Functionally diverse type V CRISPR-Cas systems. Science 2018, 363, 88–91. [Google Scholar] [CrossRef]
  56. Li, L.; Li, S.; Wu, N.; Wu, J.; Wang, G.; Zhao, G.; Wang, J. HOLMESv2: A CRISPR-Cas12b-Assisted Platform for Nucleic Acid Detection and DNA Methylation Quantitation. ACS Synth. Biol. 2019, 8, 2228–2237. [Google Scholar] [CrossRef]
  57. Teng, F.; Guo, L.; Cui, T.; Wang, X.-G.; Xu, K.; Gao, Q.; Zhou, Q.; Li, W. CDetection: CRISPR-Cas12b-based DNA detection with sub-attomolar sensitivity and single-base specificity. Genome Biol. 2019, 20, 1–7. [Google Scholar] [CrossRef]
  58. Harrington, L.B.; Burstein, D.; Chen, J.S.; Paez-Espino, D.; Ma, E.; Witte, I.P.; Cofsky, J.C.; Kyrpides, N.C.; Banfield, J.F.; Doudna, J.A. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science 2018, 362, 839–842. [Google Scholar] [CrossRef]
  59. Heng, Z.; Zhuang, L.; Renjian, X.; Leifu, C. Mechanisms for target recognition and cleavage by the Cas12i RNA-guided endonuclease. Nat. Struct. Mol. Biol. 2020, 27, 1069–1076. [Google Scholar]
  60. Marraffini, L.A. CRISPR-Cas immunity in prokaryotes. Nature 2015, 526, 55–61. [Google Scholar] [CrossRef]
  61. Li, S.Y.; Cheng, Q.X.; Liu, J.K.; Nie, X.Q.; Zhao, G.P.; Wang, J. CRISPR-Cas12a has both cis- and trans-cleavage activities on single-stranded DNA. Cell Res. 2018, 28, 491–493. [Google Scholar] [CrossRef]
  62. Abavisani, M.; Khayami, R.; Hoseinzadeh, M.; Kodori, M.; Kesharwani, P.; Sahebkar, A. CRISPR-Cas system as a promising player against bacterial infection and antibiotic resistance. Drug Resist. Updates 2023, 68, 100948. [Google Scholar] [CrossRef]
  63. Shmakov, S.; Abudayyeh, O.O.; Makarova, K.S.; Wolf, Y.I.; Gootenberg, J.S.; Semenova, E.; Minakhin, L.; Joung, J.; Konermann, S.; Severinov, K.; et al. Discovery and Functional Characterization of Diverse Class 2 CRISPR-Cas Systems. Mol. Cell 2015, 60, 385–397. [Google Scholar] [CrossRef] [PubMed]
  64. East-Seletsky, A.; O’Connell, M.R.; Knight, S.C.; Burstein, D.; Cate, J.H.D.; Tjian, R.; Doudna, J.A. Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature 2016, 538, 270–273. [Google Scholar] [CrossRef] [PubMed]
  65. Konermann, S.; Lotfy, P.; Brideau, N.J.; Oki, J.; Shokhirev, M.N.; Hsu, P.D. Transcriptome Engineering with RNA-Targeting Type VI-D CRISPR Effectors. Cell 2018, 173, 665–676.e14. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, B.; Ye, Y.; Ye, W.; Perčulija, V.; Jiang, H.; Chen, Y.; Li, Y.; Chen, J.; Lin, J.; Wang, S.; et al. Two Hepn Domains Dictate Crispr Rna Maturation and Target Cleavage in Cas13d. Sci. Lett. 2019, 10, 2544. [Google Scholar] [CrossRef]
  67. Han, Y.; Li, F.; Yang, L.; Guo, X.; Dong, X.; Niu, M.; Jiang, Y.; Li, L.; Li, H.; Sun, Y. Imunocapture Magnetic Beads Enhanced and Ultrasensitive CRISPR-Cas13a-Assisted Electrochemical Biosensor for Rapid Detection of SARS-CoV-2. Biosensors 2023, 13, 597. [Google Scholar] [CrossRef] [PubMed]
  68. Burstein, D.; Harrington, L.B.; Strutt, S.C.; Probst, A.J.; Anantharaman, K.; Thomas, B.C.; Doudna, J.A.; Banfield, J.F. New CRISPR-Cas systems from uncultivated microbes. Nature 2017, 542, 237–241. [Google Scholar] [CrossRef] [PubMed]
  69. Malinin, N.L.; Lee, G.; Lazzarotto, C.R.; Li, Y.; Zheng, Z.; Nguyen, N.T.; Liebers, M.; Topkar, V.V.; Iafrate, A.J.; Le, L.P.; et al. Defining genome-wide CRISPR-Cas genome-editing nuclease activity with GUIDE-seq. Nat. Protoc. 2021, 16, 5592–5615. [Google Scholar] [CrossRef] [PubMed]
  70. Son, H. Harnessing CRISPR/Cas Systems for DNA and RNA Detection: Principles, Techniques, and Challenges. Biosensors 2024, 14, 460. [Google Scholar] [CrossRef]
  71. Feng, W.; Newbigging, A.M.; Tao, J.; Cao, Y.; Peng, H.; Le, C.; Wu, J.; Pang, B.; Li, J.; Tyrrell, D.L.; et al. CRISPR technology incorporating amplification strategies: Molecular assays for nucleic acids, proteins, and small molecules. Chem. Sci. 2021, 12, 4683–4698. [Google Scholar] [CrossRef]
  72. Kellner, M.J.; Koob, J.G.; Gootenberg, J.S.; Abudayyeh, O.O.; Zhang, F. SHERLOCK: Nucleic acid detection with CRISPR nucleases. Nat. Protoc. 2019, 14, 2986–3012. [Google Scholar] [CrossRef]
  73. Li, S.Y.; Cheng, Q.X.; Wang, J.M.; Li, X.Y.; Zhang, Z.L.; Gao, S.; Cao, R.B.; Zhao, G.P.; Wang, J. CRISPR-Cas12a-assisted nucleic acid detection. Cell Discov. 2018, 4, 20. [Google Scholar] [CrossRef] [PubMed]
  74. Shi, K.; Xie, S.; Tian, R.; Wang, S.; Lu, Q.; Gao, D.; Lei, C.; Zhu, H.; Nie, Z. A CRISPR-Cas autocatalysis-driven feedback amplification network for supersensitive DNA diagnostics. Sci. Adv. 2021, 7, eabc7802. [Google Scholar] [CrossRef]
  75. Xu, J.; Bai, X.; Zhang, X.; Yuan, B.; Guo, Y.; Cui, Y.; Liu, J.; Cui, H.; Ren, X.; Wang, J.; et al. Development and application of DETECTR-based rapid detection for pathogenic Bacillusanthracis. Anal. Chim. Acta 2023, 1247, 340891. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, T.; Chen, Y.; He, J.; Wu, J.; Wang, M.; Zhong, X. A Designed Vessel Using Dissolvable Polyvinyl Alcohol Membrane as Automatic Valve to Couple LAMP with CRISPR/Cas12a System for Visual Detection. Biosensors 2023, 13, 111. [Google Scholar] [CrossRef]
  77. Atçeken, N.; Yigci, D.; Ozdalgic, B.; Tasoglu, S. CRISPR-Cas-Integrated LAMP. Biosensors 2022, 12, 1035. [Google Scholar] [CrossRef] [PubMed]
  78. Myhrvold, C.; Freije, C.A.; Gootenberg, J.S.; Abudayyeh, O.O.; Metsky, H.C.; Durbin, A.F.; Kellner, M.J.; Tan, A.L.; Paul, L.M.; Parham, L.A.; et al. Field-deployable viral diagnostics using CRISPR-Cas13. Science 2018, 360, 444–448. [Google Scholar] [CrossRef] [PubMed]
  79. Gootenberg, J.S.; Abudayyeh, O.O.; Kellner, M.J.; Joung, J.; Collins, J.J.; Zhang, F. Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science 2018, 360, 439–444. [Google Scholar] [CrossRef]
  80. Li, H.; Kielich, D.M.; Liu, G.; Smith, G.; Bello, A.; Strong, J.E.; Pickering, B.S. Strategies to Improve Multi-enzyme Compatibility and Coordination in One-Pot SHERLOCK. Anal. Chem. 2023, 95, 10522–10531. [Google Scholar] [CrossRef]
  81. Pena, J.M.; Manning, B.J.; Li, X.; Fiore, E.S.; Carlson, L.; Shytle, K.; Nguyen, P.P.; Azmi, I.; Larsen, A.; Wilson, M.K.; et al. Real-time, multiplexed SHERLOCK for in vitro diagnostics. J. Mol. Diagn. JMD 2023, 25, 428–437. [Google Scholar] [CrossRef] [PubMed]
  82. Quan, J.; Langelier, C.; Kuchta, A.; Batson, J.; Teyssier, N.; Lyden, A.; Caldera, S.; McGeever, A.; Dimitrov, B.; King, R.; et al. FLASH: A next-generation CRISPR diagnostic for multiplexed detection of antimicrobial resistance sequences. Nucleic Acids Res. 2019, 47, e83. [Google Scholar] [CrossRef]
  83. Munawar, N.; Ahsan, K.; Ahmad, A. CRISPR-edited plants’ social, ethical, policy, and governance issues. In Global Regulatory Outlook for CRISPRized Plants; Elsevier: Amsterdam, The Netherlands, 2024; pp. 367–396. [Google Scholar]
  84. Kolkur, S.U.; Sharma, A.; Gouda, M.R.; Praveen, K.; Singh, A. CRISPR in Agriculture and it’s Ethical Implications: A Bibliometric analysis. Food Humanit. 2024, 3, 100322. [Google Scholar] [CrossRef]
  85. Sobral, A.F.; Oliveira, R.J.D.; Barbosa, D.J. CRISPR-Cas technology in forensic investigations: Principles, applications, and ethical considerations. Forensic Sci. Int. Genet. 2024, 74, 103163. [Google Scholar] [CrossRef] [PubMed]
  86. Cheng, X.; Li, Y.; Kou, J.; Liao, D.; Zhang, W.; Yin, L.; Man, S.; Ma, L. Novel non-nucleic acid targets detection strategies based on CRISPR/Cas toolboxes: A review. Biosens. Bioelectron. 2022, 215, 114559. [Google Scholar] [CrossRef] [PubMed]
  87. Huang, Z.; Liu, S.; Pei, X.; Li, S.; He, Y.; Tong, Y.; Liu, G. Fluorescence Signal-Readout of CRISPR/Cas Biosensors for Nucleic Acid Detection. Biosensors 2022, 12, 779. [Google Scholar] [CrossRef] [PubMed]
  88. Liang, M.; Li, Z.; Wang, W.; Liu, J.; Liu, L.; Zhu, G.; Karthik, L.; Wang, M.; Wang, K.F.; Wang, Z.; et al. A CRISPR-Cas12a-derived biosensing platform for the highly sensitive detection of diverse small molecules. Nat. Commun. 2019, 10, 3672. [Google Scholar] [CrossRef]
  89. Wu, Y.; Chang, D.; Chang, Y.; Zhang, Q.; Liu, Y.; Brennan, J.D.; Li, Y.; Liu, M. Nucleic Acid Enzyme-Activated CRISPR-Cas12a with Circular CRISPR RNA for Biosensing. Small 2023, 19, e2303007. [Google Scholar] [CrossRef] [PubMed]
  90. Chandrasekhar, D.; Joseph, C.M.; Parambil, J.C.; Murali, S.; Yahiya, M.; K, S. Superbugs: An invicible threat in post antibiotic era. Clin. Epidemiol. Glob. Health 2024, 28, 101499. [Google Scholar] [CrossRef]
  91. Rayan, R.A. Pharmaceutical effluent evokes superbugs in the environment: A call to action. Biosaf. Health 2023, 5, 363–371. [Google Scholar] [CrossRef]
  92. Brolund, A.; Sandegren, L. Characterization of ESBL disseminating plasmids. Infect. Dis. 2016, 48, 18–25. [Google Scholar] [CrossRef] [PubMed]
  93. Von Wintersdorff, C.J.; Penders, J.; Van Niekerk, J.M.; Mills, N.D.; Majumder, S.; Van Alphen, L.B.; Savelkoul, P.H.; Wolffs, P.F. Dissemination of Antimicrobial Resistance in Microbial Ecosystems through Horizontal Gene Transfer. Front. Microbiol. 2016, 7, 173. [Google Scholar] [CrossRef]
  94. Evans, D.R.; Griffith, M.P.; Sundermann, A.J.; Shutt, K.A.; Saul, M.I.; Mustapha, M.M.; Marsh, J.W.; Cooper, V.S.; Harrison, L.H.; Van Tyne, D. Systematic detection of horizontal gene transfer across genera among multidrug-resistant bacteria in a single hospital. eLife 2020, 9, e53886. [Google Scholar] [CrossRef]
  95. Haaber, J.; Penadés, J.R.; Ingmer, H. Transfer of Antibiotic Resistance in Staphylococcus aureus. Trends Microbiol. 2017, 25, 893–905. [Google Scholar] [CrossRef] [PubMed]
  96. Lagier, J.C.; Edouard, S.; Pagnier, I.; Mediannikov, O.; Drancourt, M.; Raoult, D. Current and past strategies for bacterial culture in clinical microbiology. Clin. Microbiol. Rev. 2015, 28, 208–236. [Google Scholar] [CrossRef]
  97. Ouyang, B.; Yang, C.; Lv, Z.; Chen, B.; Tong, L.; Shi, J. Recent advances in environmental antibiotic resistance genes detection and research focus: From genes to ecosystems. Environ. Int. 2024, 191, 108989. [Google Scholar] [CrossRef]
  98. Shrestha, S.; Malla, B.; Haramoto, E. High-throughput microfluidic quantitative PCR system for the simultaneous detection of antibiotic resistance genes and bacterial and viral pathogens in wastewater. Environ. Res. 2024, 255, 119156. [Google Scholar] [CrossRef] [PubMed]
  99. Gorecki, A.; Decewicz, P.; Dziurzynski, M.; Janeczko, A.; Drewniak, L.; Dziewit, L. Literature-based, manually-curated database of PCR primers for the detection of antibiotic resistance genes in various environments. Water Res. 2019, 161, 211–221. [Google Scholar] [CrossRef] [PubMed]
  100. Manghwar, H.; Lindsey, K.; Zhang, X.; Jin, S. CRISPR/Cas System: Recent Advances and Future Prospects for Genome Editing. Trends Plant Sci. 2019, 24, 1102–1125. [Google Scholar] [CrossRef]
  101. Okaiyeto, S.A.; Sutar, P.P.; Chen, C.; Ni, J.B.; Wang, J.; Mujumdar, A.S.; Zhang, J.S.; Xu, M.Q.; Fang, X.M.; Zhang, C.; et al. Antibiotic resistant bacteria in food systems: Current status, resistance mechanisms, and mitigation strategies. Agric. Commun. 2024, 2, 100027. [Google Scholar] [CrossRef]
  102. Raro, O.H.F.; Bouvier, M.; Kerbol, A.; Poirel, L.; Nordmann, P. MultiRapid ATB NP test for detecting concomitant susceptibility and resistance of last-resort novel antibiotics available to treat multidrug-resistant Enterobacterales infections. Int. J. Antimicrob. Agents 2024, 64, 107206. [Google Scholar] [CrossRef]
  103. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  104. Müller, V.; Rajer, F.; Frykholm, K.; Nyberg, L.K.; Quaderi, S.; Fritzsche, J.; Kristiansson, E.; Ambjörnsson, T.; Sandegren, L.; Westerlund, F. Direct identification of antibiotic resistance genes on single plasmid molecules using CRISPR/Cas9 in combination with optical DNA mapping. Sci. Rep. 2016, 6, 37938. [Google Scholar] [CrossRef]
  105. Nyblom, M.; Johnning, A.; Frykholm, K.; Wrande, M.; Müller, V.; Goyal, G.; Robertsson, M.; Dvirnas, A.; Sewunet, T.; Kk, S.; et al. Strain-level bacterial typing directly from patient samples using optical DNA mapping. Commun. Med. 2023, 3, 31. [Google Scholar] [CrossRef] [PubMed]
  106. Qin, K.; Zhang, P.; Li, Z. Specific detection of antibiotic-resistant bacteria using CRISPR/Cas9 induced isothermal exponential amplification reaction (IEXPAR). Talanta 2023, 253, 124045. [Google Scholar] [CrossRef]
  107. Curti, L.A.; Pereyra-Bonnet, F.; Repizo, G.D.; Fay, J.V.; Salvatierra, K.; Blariza, M.J.; Ibañez-Alegre, D.; Rinflerch, A.R.; Miretti, M.; Gimenez, C.A. CRISPR-based platform for carbapenemases and emerging viruses detection using Cas12a (Cpf1) effector nuclease. Emerg. Microbes Infect. 2020, 9, 1140–1148. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, H.; Li, B.; Shi, S.; Zhou, T.; Wang, X.; Wang, Z.; Zhou, X.; Wang, M.; Shi, W.; Ren, L. Au–Fe3O4 nanozyme coupled with CRISPR-Cas12a for sensitive and visual antibiotic resistance diagnosing. Anal. Chim. Acta 2023, 1251, 341014. [Google Scholar] [CrossRef] [PubMed]
  109. Park, D.H.; Haizan, I.; Ahn, M.J.; Choi, M.Y.; Kim, M.J.; Choi, J.H. One-Pot CRISPR-Cas12a-Based Viral DNA Detection via HRP-Enriched Extended ssDNA-Modified Au@Fe3O4 Nanoparticles. Biosensors 2024, 14, 26. [Google Scholar] [CrossRef] [PubMed]
  110. Zhou, R.; Li, Y.; Dong, T.; Tang, Y.; Li, F. A sequence-specific plasmonic loop-mediated isothermal amplification assay with orthogonal color readouts enabled by CRISPR Cas12a. Chem. Commun. 2020, 56, 3536. [Google Scholar] [CrossRef] [PubMed]
  111. Mao, K.; Zhang, H.; Ran, F.; Cao, H.; Feng, R.; Du, W.; Li, X.; Yang, Z. Portable biosensor combining CRISPR/Cas12a and loop-mediated isothermal amplification for antibiotic resistance gene ermB in wastewater. J. Hazard. Mater. 2023, 462, 132793. [Google Scholar] [CrossRef]
  112. Shin, J.; Kim, S.R.; Xie, Z.; Jin, Y.S.; Wang, Y.C. A CRISPR/Cas12a-Based System for Sensitive Detection of Antimicrobial-Resistant Genes in Carbapenem-Resistant Enterobacterales. Biosensors 2024, 14, 194. [Google Scholar] [CrossRef]
  113. Gootenberg, J.S.; Abudayyeh, O.O.; Lee, J.W.; Essletzbichler, P.; Dy, A.J.; Joung, J.; Verdine, V.; Donghia, N.; Daringer, N.M.; Freije, C.A.; et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science 2017, 356, 438–442. [Google Scholar] [CrossRef]
  114. Hu, Q.; Li, H.; Hu, X.; Han, X.; Sun, Y.; Liu, Y. A CRISPR/Cas13a-based detection method for the drug resistance gene mecA of Staphylococcus aureus. Acta Microbiol. Sin. 2023, 63, 3628–3640. [Google Scholar] [CrossRef]
  115. Yan, K.C.; Wang, X.H.; Han, Y.; Tian, Y.; Niu, M.W.; Dong, X.; Li, X.W.; Li, H.; Sun, Y.S. Simultaneous Detection of Helicobacter pylori and Clarithromycin Resistance Mutations Using RAA-CRISPR/Cas13a Assay. Infect. Drug Resist. 2024, 17, 3001–3010. [Google Scholar] [CrossRef] [PubMed]
  116. Ortiz-Cartagena, C.; Pablo-Marcos, D.; Fernández-García, L.; Blasco, L.; Pacios, O.; Bleriot, I.; Siller, M.; López, M.; Fernández, J.; Aracil, B.; et al. CRISPR-Cas13a-Based Assay for Accurate Detection of OXA-48 and GES Carbapenemases. Microbiol. Spectr. 2023, 11, e0132923. [Google Scholar] [CrossRef]
  117. Price, V.J.; Huo, W.; Sharifi, A.; Palmer, K.L.; Fey, P.D. CRISPR-Cas and Restriction-Modification Act Additively against Conjugative Antibiotic Resistance Plasmid Transfer in Enterococcus faecalis. mSphere 2016, 1, e00064. [Google Scholar] [CrossRef] [PubMed]
  118. Hille, F.; Charpentier, E. CRISPR-Cas: Biology, mechanisms and relevance. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2016, 371, 20150496. [Google Scholar] [CrossRef] [PubMed]
  119. Mackow, N.A.; Shen, J.; Adnan, M.; Khan, A.S.; Fries, B.C.; Diago-Navarro, E. CRISPR-Cas influences the acquisition of antibiotic resistance in Klebsiella pneumoniae. PLoS ONE 2019, 14, e0225131. [Google Scholar] [CrossRef]
  120. Pursey, E.; Dimitriu, T.; Paganelli, F.L.; Westra, E.R.; van Houte, S. CRISPR-Cas is associated with fewer antibiotic resistance genes in bacterial pathogens. Philos. Trans. R. Soc. B 2022, 377, 20200464. [Google Scholar] [CrossRef]
  121. Du, Y.; Han, D.; An, Z.; Wang, J.; Gao, Z. CRISPR/dCas9—Surface-enhanced Raman scattering for the detection of drug resistance gene macB. Microchim. Acta 2022, 189, 394. [Google Scholar] [CrossRef]
  122. Gong, L.; Jin, Z.; Liu, E.; Tang, F.; Yuan, F.; Liang, J.; Wang, Y.; Liu, X.; Wang, Y. Highly Sensitive and Specific Detection of Mobilized Colistin Resistance Gene mcr-1 by CRISPR-Based Platform. Microbiol. Spectr. 2022, 10, e0188422. [Google Scholar] [CrossRef] [PubMed]
  123. Gao, H.; Li, Y.; Li, Y.; Qu, K.; Zhang, K.; Li, J. Detection of antibiotic-resistance genes in bacterial pathogens using a Cas12a/3D DNAzyme colorimetric paper sensor. Fundam. Res. 2023. [Google Scholar] [CrossRef]
  124. Fu, X.; Sun, J.; Ye, Y.; Zhang, Y.; Sun, X. A rapid and ultrasensitive dual detection platform based on Cas12a for simultaneous detection of virulence and resistance genes of drug-resistant Salmonella. Biosens. Bioelectron. 2022, 195, 113682. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, X.X.; Wang, Y.S.; Li, S.J.; Peng, R.Q.; Wen, X.; Peng, H.; Shi, Q.S.; Zhou, G.; Xie, X.B.; Wang, J. Rapid detection of mexX in Pseudomonas aeruginosa based on CRISPR-Cas13a coupled with recombinase polymerase amplification. Front. Microbiol. 2024, 15, 1341179. [Google Scholar] [CrossRef]
  126. Mingjun, L.; Bin, X.; Lidan, C.; Xiaoyan, H.; Jinchao, L.; Zhenzhan, K.; Xinping, C.; Xiuna, H.; Zhaohui, S.; Linhai, L. Rapid Detection of blaKPC in Carbapenem-Resistant Enterobacterales Based on CRISPR/Cas13a. Curr. Microbiol. 2023, 80, 352. [Google Scholar]
  127. Michael, E. Seven technologies to watch in 2022. Nature 2022, 601, 658–661. [Google Scholar]
  128. Jianyu, H.; Jing, Z.; Rui, L.; Yi, L. Element probe based CRISPR/Cas14 bioassay for non-nucleic-acid targets. Chem. Commun. 2021, 57, 10423–10426. [Google Scholar]
  129. Mahas, A.; Wang, Q.; Marsic, T.; Mahfouz, M.M. Development of Cas12a-Based Cell-Free Small-Molecule Biosensors via Allosteric Regulation of CRISPR Array Expression. Anal. Chem. 2022, 94, 4617–4626. [Google Scholar] [CrossRef] [PubMed]
  130. Chen, J.; Shi, G.; Yan, C. Portable biosensor for on-site detection of kanamycin in water samples based on CRISPR-Cas12a and an off-the-shelf glucometer. Sci. Total Environ. 2023, 872, 162279. [Google Scholar] [CrossRef] [PubMed]
  131. Zhu, L.; Zhang, X.; Yang, L.; Qiu, S.; Liu, G.; Xiong, X.; Xiao, T.; Huang, K.; Zhu, L. Label-free electrochemical sensing platform for sensitive detection of ampicillin by combining nucleic acid isothermal enzyme-free amplification circuits with CRISPR/Cas12a. Talanta 2024, 273, 125950. [Google Scholar] [CrossRef]
  132. Yee, B.J.; Zakaria, S.N.A.; Chandrawati, R.; Ahmed, M.U. Detection of Tetracycline with a CRISPR/Cas12a Aptasensor Using a Highly Efficient Fluorescent Polystyrene Microsphere Reporter System. ACS Synth. Biol. 2024, 13, 2166–2176. [Google Scholar] [CrossRef]
  133. Zhang, X.; Zhu, L.; Yang, L.; Liu, G.; Qiu, S.; Xiong, X.; Huang, K.; Xiao, T.; Zhu, L. A sensitive and versatile electrochemical sensor based on hybridization chain reaction and CRISPR/Cas12a system for antibiotic detection. Anal. Chim. Acta 2024, 1304, 342562. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Non-nucleic acid target-detection strategy-based on CRISPR-Dx. The innermost circle describes the principle of action of the four CRISPR effector proteins; the middle circle represents the non-nucleic acid targets detectable by the CRISPR effector proteins; and the outermost circle describes the non-nucleic acid targets in the environment that are converted by the bio-transduction element into recognizable nucleic acid signals, which are then outputted on the constructed CRISPR detection platform. Created in https://BioRender.com.
Figure 1. Non-nucleic acid target-detection strategy-based on CRISPR-Dx. The innermost circle describes the principle of action of the four CRISPR effector proteins; the middle circle represents the non-nucleic acid targets detectable by the CRISPR effector proteins; and the outermost circle describes the non-nucleic acid targets in the environment that are converted by the bio-transduction element into recognizable nucleic acid signals, which are then outputted on the constructed CRISPR detection platform. Created in https://BioRender.com.
Biosensors 14 00633 g001
Table 1. Commonly methods used for antibiotics detection.
Table 1. Commonly methods used for antibiotics detection.
No.Detection MethodsReadout DetectorTargetDetection RangeLODReal SampleTime
(min)
Ref.
1National Standard Method -LC-MS/MSMSMethicillinNR0.1 μg/kgFood of animal originNRGB/T 21315-2007
2LC-MS/MSMSAmpicillin(AMPI)/clarithromycin4 × 105~
2.5 × 107 nM
0.14~59.8
μM
JawboneNR[19]
3HPLCChromatographSulfadiazine50~500 ng/mL22.4 ng/mLMilkNR[21]
4ELISASmartphoneTetracycline
chloromycetin
1~103 ng/mL;
0.1~100 ng/ml
0.5 ng/mL 0.05 ng/mLMilk/fishNR[34]
5SERSRaman spectrometerloxacinNR15.8 μg/LAquatic products (fish)30[35]
6SERSSERS-ICA test stripSulfadimethoxine0.1~
103 pg/mL
0.1 ng/LMilk15[36]
7FETFETAmpicillin10−12~
10−6 M
0.556 pMCreek3[24]
8Aptamer-modified graphene field-effect transistors (Apt-SGGT)Digital seismographTetracyclineNR2.073 pMSkim milk8[23]
9MOF Fluorescence SensorSmartphoneFluoroquinolone0~90 μM16 nMNR20[37]
10MOF Electrochemical SensorsElectrochemical workstationCiprofloxacin2.5~
100 µM
3.29 nMTap water/seawaterNR[38]
11Long Afterglow Optical SensorsSpectrophotometerKanamycin1 pg/mL~
5 ng/m L
0.32 pg/mLMilk/honey/powdered milk90[39]
12Long Afterglow Markless SensorsFluorescence spectrophotometerFuracilinum0.1~50 mM5 nMMilk/Dianchi water samplesNR[40]
13CRISPR/Cas12a Light SensorsMicroplate ReaderAmpicillin0.01 nM~ 500 nM10 pMMilk/eggs/honey30[41]
14CRISPR/Cas12a BiosensorSpectrophotometerTobramycin10~300 pM3.719 pMMilk/lake water40[42]
15Metal-labeled CRISPR/Cas12a biosensorsICPMSKanamycin8~120 pm4.06 pMWild fish30[43]
16Microfluidic SensorsUV-visible spectrophotometerKanamycin0.8 pg/mL~10 ng/mL0.3 pg/mLMilk/fishNR[44]
Table 3. CRISPR/Cas for the detection of antibiotic micromolecules.
Table 3. CRISPR/Cas for the detection of antibiotic micromolecules.
CAS TypeDetection ObjectSensing MethodReadout DetectorDetecting Linear RangeLODTime
(min)
Real SampleRef.
CRISPR/Cas14AmpicillinMetal isotope labelingICPMSNR2.06 nM45NR[43]
CRISPR/Cas12aKanamycinMetal isotope labelingICPMS8–120 pM4.06 pM30Wild fish[128]
TobramycinAptasensorSpectrophotometer10–300 pM3.719 pM40Milk/lake Water[42]
TetracyclineaTFSmartphoneNR2 μMNRNR[129]
AmpicillinAptasensorMicroplate reader0.01–500 nM10 pM30Milk/eggs/honey[41]
KanamycinAptasensorglucometer1pM–100 nM1 pMNRWater sample[130]
AmpicillinHCR/
Electrochemical sensor
Electrochemical workstation5 pM–100 nM1.60 pM160Milk/
livestock wastewater
[131]
KanamycinAmpicillinHCR/
Electrochemical sensor
Electrochemical workstation0.10 pM–10 nM;
0.05 pM–10 nM
60 fM; 10 fMNRMilk/
livestock wastewater
[132]
TetracyclineAptasensorSpectrophotometer15–500 μM0.1 μM120Milk/raw Beef[133]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, X.; Huang, Z.; Zhang, Y.; Wang, W.; Ye, Z.; Liang, P.; Sun, K.; Kang, W.; Tang, Q.; Yu, X. Mitigating Antibiotic Resistance: The Utilization of CRISPR Technology in Detection. Biosensors 2024, 14, 633. https://doi.org/10.3390/bios14120633

AMA Style

Zhang X, Huang Z, Zhang Y, Wang W, Ye Z, Liang P, Sun K, Kang W, Tang Q, Yu X. Mitigating Antibiotic Resistance: The Utilization of CRISPR Technology in Detection. Biosensors. 2024; 14(12):633. https://doi.org/10.3390/bios14120633

Chicago/Turabian Style

Zhang, Xuejiao, Zhaojie Huang, Yanxia Zhang, Wen Wang, Zihong Ye, Pei Liang, Kai Sun, Wencheng Kang, Qiao Tang, and Xiaoping Yu. 2024. "Mitigating Antibiotic Resistance: The Utilization of CRISPR Technology in Detection" Biosensors 14, no. 12: 633. https://doi.org/10.3390/bios14120633

APA Style

Zhang, X., Huang, Z., Zhang, Y., Wang, W., Ye, Z., Liang, P., Sun, K., Kang, W., Tang, Q., & Yu, X. (2024). Mitigating Antibiotic Resistance: The Utilization of CRISPR Technology in Detection. Biosensors, 14(12), 633. https://doi.org/10.3390/bios14120633

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop