3,386
views
1
recommends
+1 Recommend
1 collections
    4
    shares

      2023 Scopus CiteScore is 2.3, SNIP 0.757, ranking 15/35 in Category "Veterinary (Miscellaneous)" and 219/344 "Medicine (Infectious Diseases)".  

      Interested in becoming a Zoonoses published author? Check out the call for papers on our website https://zoonoses-journal.org/index.php/2023/04/26/zoonoses-call-for-papers-2/

      • Platinum Open Access with no APCs & Fast peer review/Fast publication online after article acceptance
      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      CRISPR-Cas- and Aptamer-based Systems for Diagnosing Pathogens: A Review

      Published
      review-article
      Bookmark

            Abstract

            Pathogenic infections cause severe clinical illnesses in humans and animals. Increased encounters between humans and animals and constant environmental changes exacerbate the transmission of zoonotic infectious diseases. Recently, the World Health Organization has declared some zoonotic epidemics as public health emergencies of international concern. Hence, rapid and accurate detection of the causative pathogen is particularly essential in combating emerging and re-emerging infectious diseases. Traditional pathogen detection tools are time-consuming, costly, and require skilled personnel, which greatly hinder the development of rapid diagnostic tests, particularly in resource-constrained regions. Clustered regularly interspaced short palindromic repeats (CRISPR-)-Cas- and aptamer-based platforms have replaced traditional pathogen detection methods. Herein we review two novel next-generation core pathogen detection platforms that are utilized for clinical and foodborne pathogenic microorganisms: CRISPR-Cas-based systems, including dCas9, Cas12a/b, Cas13, and Cas14; and aptamer-based biosensor detection tools. We highlight CRISPR-Cas- and aptamer-based techniques and compare the strengths and weaknesses. CRISPR-Cas-based tools require cumbersome procedures, such as nucleic acid amplification and extraction, while aptamer-based tools require improved sensitivity. We review the combination of CRISPR-Cas- and aptamer-based techniques as a promising approach to overcome these deficiencies. Finally, we discuss Cas14-based tools as functionally stronger platforms for the detection of non-nucleic acid targets.

            Main article text

            INTRODUCTION

            Zoonoses (infectious diseases transmitted between animals and humans) are widely prevalent worldwide, and viral infections are the most urgent emerging zoonoses [13]. Greater 200 zoonoses that have been identified [4] include most of the emerging and re-emerging infectious diseases [5]. Based on pathogenic characteristics, zoonoses can be classified as bacterial, viral, fungal, parasitic, rickettsial, and mycoplasma zoonoses [6]. For example, plague [7], colibacillosis, and brucellosis [8] are zoonoses caused by bacteria and coronavirus disease (COVID-19), which is caused by severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) [9], acquired immunodeficiency syndrome [10], Lassa fever [11], Ebola hemorrhagic fever [12], and Zika virus disease [13] are viral zoonoses, whereas cryptosporidiosis [14] and giardiasis [15] are zoonoses that arise from parasites. These diseases severely threaten human and animal health and can cause devastating global economic damage. Under the dual pressures of emerging and re-emerging zoonoses, many uncertainties remain in the prevention and control of these infectious diseases. Furthermore, infections due to these pathogens do not respond well to treatment and are associated with low survival rates; thus, early disease diagnosis and pathogen identification are particularly important in controlling the spread of zoonoses [3,16,17], with the current global SARS-CoV-2 pandemic a strong case in point.

            The last two decades have witnessed a fast expansion in the repertoire of tools developed for pathogen detection. The clinical diagnosis of pathogens is generally based on the detection of pathogen-specific genes and proteins using techniques, such as reverse transcription (RT-)-polymerase chain reaction (PCR), RT-quantitative PCR (qPCR), enzyme-linked immunosorbent assay (ELISA), next-generation sequencing (NGS), fluorescence in situ hybridization (FISH), and chemiluminescence. These tools are very effective and have facilitated the evolution of diagnostics; however, traditional platforms, including serologic methods, which are only workable after antibodies have been produced [18,19], are time-consuming and can identify non-contagious cases as contagious (false-positive results), thus limiting the application of these methods in point-of-care testing (POCT). Pathogen detection tools based on antigen-antibody binding include ELISA [20], which is high in cost due to the heavy use of antibodies. Nucleic acid-based diagnostic methods, such as RT-PCR and RT-qPCR [21], are not convenient and require specialized equipment and trained personnel, although RT-qPCR is the gold-standard detection method for some RNA viruses, such as SARS-CoV-2 [21,22]. Nevertheless, nucleic acid-based tools have several advantages, including lower cost and higher sensitivity, over serologic methods [23,24]. Overall, these approaches are time-consuming, require strict operating procedures [25], are difficult in the rapid identification of pathogens, and cannot facilitate the interruption of pathogen transmission in real time, which is a challenge for global public health. In the setting of an increasing number of emerging and re-emerging zoonotic pathogens circulating among human populations, such as SARS-CoV-2, which remains as a pandemic since its emergence in late 2019, there is an urgent need for the development of new diagnostic tools that fulfill the affordable, sensitive, specific, user-friendly, rapid and robust, equipment-free, and deliverable to end-users needs [21].

            Following the rapid evolution in our understanding of the clustered regularly interspaced short palindromic repeat (CRISPR-)-Cas systems, different Cas effector proteins have been used for the identification of zoonotic pathogens based on their unique characteristics. This novel platform is highly dependent on nucleic acids isolated from pathogens and detection based on nucleic acid amplification. Isothermal amplification methods, such as recombinase polymerase amplification (RPA), loop-mediated isothermal amplification (LAMP), and RT-LAMP, are effective for pathogen detection in low-resource settings [19,21,2628]. The CRISPR-Cas-based diagnostic platforms utilize isothermal amplification techniques to detect pathogens for high-speed POCT with improved sensitivity.

            Aptamers, which are short single-stranded artificial oligo (deoxy) nucleotides, have several advantages over antibodies, including smaller molecular weight and easier synthesis and screening [29]. Many aptamer-based biosensor detection platforms that are combined with physical, chemical, and electrochemical approaches are widely used for POCT. These pathogen detection platforms often involve multiple disciplines. Although CRISPR-Cas- and aptamer-based detection methods provide several advantages over conventional tools for pathogen detection, shortcomings remain. For example, CRISPR-Cas-based methods require nucleic acid extraction and amplification, whereas aptamer-based detection methods require aptamer screening, which is time-consuming (Table 1). Recent studies have introduced the combination of CRISPR-Cas- and aptamer-based platforms as a means to overcome these shortcomings (Table 1). These novel platforms exhibit robust detection capabilities and advantages over traditional methods and have potential utility for pathogen diagnosis and food health surveillance.

            TABLE 1 |

            Advantages and disadvantages of tools for pathogen detection.

            ToolsAdvantagesDisadvantages
            TraditionalAccurate, sensitive, gold standard for particular pathogensHigh cost, time-consuming, need for sophisticated equipment and skilled operators
            CRISPR-Cas-based systemsRapid, convenient, accurate, sensitive, robust, specific, time savings, reduce the false-positive rate, does not require sophisticated equipment, easy to operate, can be combined with other platforms, such as microfluidics and gene chip technologiesNeed for cumbersome procedures (nucleic acid extraction and amplification), need a target site near the PAM sequence (Cas12), only recognize the RNA target (Cas13)
            Aptamer-based systemsConvenient read out, accurate, low cost, high-binding specificity, and affinity, stable, wide target range, multimode readoutScreening process is difficult and requires significant time, not sensitive to clinical samples, leading to increased cost of screening
            Combined CRISPR-Cas- and aptamer-based systemsMerits of both systems, aptamer-mediated enhanced activation of the Cas proteinMainly focuses on the detection of pathogenic bacteria

            PAM, protospacer adjacent motif.

            RAPID DETECTION PLATFORMS BASED ON CRISPR-CAS SYSTEMS

            Discovery and evolution of the CRISPR-Cas systems

            CRISPRs are short palindromic DNA sequences that were discovered in the Escherichia coli genome in 1987 [30]; CRISPRs are absent in eukaryotes and viruses [31]. The two components of the CRISPR-Cas systems are as follows: Cas proteins, which are CRISPR-associated endonucleases that cleave specific sequences; and small guide RNAs (sgRNAs), which are composed of CRISPR RNA (crRNA) and trans-activating RNA, recognize substrate DNA or RNA, and direct the Cas protein to the target site for editing. In nature, the CRISPR-Cas systems are part of the adaptive immune system of bacteria and archaea. Through the formation of RNA-guided endonuclease, CRISPR-Cas cleaves nucleic acids (DNA or RNA) that have invaded the prokaryote, thereby acting as a defense against foreign pathogens and plasmids [3235]. The CRISPR-Cas systems involve three main phases of immunity: adaptation; expression/maturation; and interference [3639]. As a result, specific Cas proteins record nucleic acid information in the CRISPR array following the invasion of foreign mobile genetic elements. The effector complex formed by the endonuclease recognizes the exogenous nucleic acid information previously stored in the CRISPR array and cleaves the invading extraneous elements of secondary infection.

            The CRISPR-Cas systems can be categorized as class 1 and 2 based on genetic loci and the conserved sequences of the Cas protein. Class 1 CRISPR-Cas systems include types I, III, and IV, whereas class 2 CRISPR-Cas systems include types II, V, and VI (Table 2) [40,41]. Additionally, in contrast to class 1 CRISPR-Cas systems, which are composed of multiple proteins, class 2 CRISPR-Cas systems, which are composed of a single protein, hold promise for utility in a wide range of applications in genome editing, gene regulation, molecular diagnostics, in vitro pathogen detection, and DNA imaging. Class 2 CRISPR-Cas systems, especially those including Cas9 and Cas12a (former known as Cpf1), have been more extensively studied than class 1 CRISPR-Cas systems. The first Cas protein utilized in genetic manipulation was Cas9, which was purified from Streptococcus pyogenes (spCas9) [42]; however, activated Cas9 proteins, even those Cas9 proteins isolated from other bacteria, cannot be used for in vitro pathogen detection due to the lack of ability to non-specifically cleave nucleic acids. The subsequently identified Cas12a protein, however, has been recognized to perform this function. Interestingly, deactivated Cas9 (dCas9) is primarily used for pathogen detection based on its sequence-specific binding feature in the absence of cleavage activity [43,44]. Cas9 and Cas12a are the most extensively studied Cas proteins for gene editing and show promise for clinical application [45]. With the advent of Cas12, Cas13, and Cas14, several rapid detection techniques based on CRISPR-Cas systems have been explored. Notably, both Cas12 and Cas14 belong to the type V CRISPR-Cas system; the latter occupies only one-half of the amino acids of the former and does not require the identification of the protospacer adjacent motif (PAM) sequence, which is necessary for recognition by Cas9 and Cas12 [46]. Therefore, Cas14 may have more robust features and applications than Cas12.

            TABLE 2 |

            Characteristics of class 2 Cas proteins.

            Cas proteinDomainProtein lengthTarget molecularPAMType of RNATrans-Cleavage
            Cas9RuvC HNH1368aadsDNANGGgRNA (crRNA+tracrRNA)No
            Cas12RuvC1300aadsDNA ssDNATTTVcrRNAYes (ssDNA)
            Cas13Two HEPN1250aassRNA*crRNAYes (ssRNA)
            Cas14RuvC530aassDNAgRNA (crRNA+tracrRNA)Yes (ssDNA)

            *Non-G nucleotide at the 3’ protospacer flanking site (PFS).

            Gene editing and detection based on the type II CRISPR-Cas system

            CRISPR-Cas9 can cleave double-stranded DNA (dsDNA) [32], which can be inserted into exogenous gene fragments for homologous recombination or non-homologous end-joining repair for gene knock-in or -out purposes [37,44,47], and can be used to successfully edit diverse genes by changing the sequence of gRNA to arbitrarily switch genes of interest. In addition, silence mutations in Cas9 that lead to dCas9 can be applied to diagnostics.

            CRISPR-dCas9

            The primary function of CRISPR-Cas9 is to splice target genes. Additionally, CRISPR-Cas9 can be applied to pathogen detection by exploiting its sequence-specific binding property that does not depend on cleavage activity (Fig 1). This detection procedure requires the introduction of two mutations to Cas9 (D10A and H840A mutations in the RuvC-I and HNH nuclease domains, respectively) [44,47]. The dCas9 harboring these silence mutations results in the removal of dsDNA cleavage activity but exhibits strong affinity for the substrate dsDNA [48] and can therefore regulate the expression of endogenous genes without gene editing. This feature of dCas9 has been used for labeling specific sequences. The CRISPR-dCas9 system can be developed for genetic screening, overexpression of genes of interest, regulation of target gene expression using transcriptional activators and repressors, and gene detection [49]. The majority of CRISPR-dCas9-based diagnostic tools can detect different target pathogens by changing the sequence of sgRNA.

            FIGURE 1 |

            Schematic illustration of the diagnostic platforms. (A) Tools based on the CRISPR-Cas system for pathogen detection using collateral cleavage activity and sequence-specific binding of Cas12, Cas13, Cas14 and dCas9. The procedure involves sample collection, nucleic acid extraction and amplification, Cas protein activation, fluorescence probe degradation, and readout of the results. (B) Aptamer-based diagnostic tools for bacteria, viruses, and parasite-specific proteins. Aptamers capture the pathogen, causing a change in the current or resistance that indicates the presence or absence of the target pathogen. CRISPR, clustered regularly interspaced short palindromic repeat.

            Assays based on CRISPR-dCas9 systems can potentially complement and enrich existing platforms used for pathogen detection. For example, the CRISPR-Tag system can detect herpes simplex virus 1 DNA in host cells [50,51] and CRISPR-mediated DNA-FISH has been used as a convenient, high-speed detection approach for methicillin-resistant Staphylococcus aureus [52]; however, CRISPR-dCas9-based detection tools have not become mainstream diagnostic platforms.

            Establishing rapid detection platforms based on CRISPR-Cas trans-cleavage activity

            Phylogenetic tree analysis has revealed that Cas12 and Cas14 belong to class 2 (type V) [53]. Unlike Cas9, Cas12a and Cas14 contain only one nuclease domain (RuvC [RuvC-I, RuvC-II, and RuvC-III]), which cleaves DNA, whereas Cas13 contains two HNH domains, which only cleave RNA [5355]. Additionally, multiple Cas12 variants, such as Cas12b, Cas12e, Cas12f, and Cas12j, have been identified. Similar to Cas12a, Cas12b has been harnessed for the development of diagnostic tools [21]. Cas12e (formerly known as CasX) and Cas12j, have been reported to edit the human genome [5659]. A recent study demonstrated a novel method using gene disruption and therapy using CRISPR-Cas12f, which was effectively packaged into an adeno-associated virus [45]. Given that dCas9, which has evolved from Cas9, can be used in diagnostics, whether these Cas12 variants can be applied to pathogen detection requires further investigation.

            Notably, Cas12a, Cas12b, Cas13, and Cas14 possess collateral cleavage activity. The CRISPR-Cas systems can be activated to non-sepcifically cleave surrounding single-stranded DNA (ssDNA) or RNA following the recognition of predetermined specific sequences [53,55,60]. This collateral trans-cleavage activity is the basis for a highly sensitive and specific in vitro rapid detection platform that utilizes a single-stranded fluorescent reporter probe. The CasN-sgRNA complex, wherein CasN represents Cas12, Cas13, or Cas14, is combined with a target sequence that can activate the trans-cleavage activity of the Cas protein and cut a fluorescent probe, causing positive signals. Efficient degradation of the fluorescent probe can be used to confirm the presence of pathogens (Fig 1). CRISPR-Cas-based diagnostic tools convert the detection of target microorganism from a molecular level to a fluorescent signal that can be directly assessed by the naked eye or quickly identified by equipment, which is a more convenient, intuitive, and time-saving approach than conventional platforms.

            In summary, the CRISPR-Cas systems have already been employed for the detection of disease-causing viruses, such as SARS-CoV-2 (Table 3), human papillomavirus (HPV) types 16 and 18, human immunodeficiency virus (HIV), Zika virus (ZIKV), and Dengue virus (DENV), and for the testing of food for organisms, such as E. coli, S. aureus, and Salmonella enterica serotype Enteritidis (S. enteritidis). Furthermore, the CRISPR-Cas systems have been harnessed to detect single nucleotide polymorphisms (SNPs), genetic mutations, and cancer (Table 4).

            TABLE 3 |

            CRISPR-based diagnostic tools for rapid and accurate detection of SARS-CoV-2.

            NucleasePlatformAmplification methodYearRefs.
            Cas12aDETECTRRT-LAMP2020[28,61]
            CRISPR-Cas12a-NERRT-RAA2020[62]
            AIOD-CRISPRRPA2020[63]
            iSCANRT-LAMP2020[21]
            STOPRT-LAMP2020[64]
            OR-DETECRTRT-RPA2021[65]
            M-CDCRT-RPA2021[66]
            sPAMCRPA2022[67]
            Cas12bCASdetecRT-RAA2020[68]
            iSCAN (one-pot)RT-LAMP2020[21]
            Cas13aSHERLOCKRPA/RT-RPA2020[69]
            CRESTAmplification-free2021[70]
            Cas13bSHERLOCKv2RPA2020[64]
            TABLE 4 |

            Reported CRISPR-Cas-based diagnostic tools.

            NucleasePlatformModelAssessmentTesting timeRefs.
            Cas12DETECTRHPV16/18FluorescenceWithin 1 hour[60]
            Electrochemical DNA biosensorChange of current (ΔI)∼1 hour[71]
            ASFVFluorescenceWithin 2 hours[72]
            Cas-goldGold nanoparticle-antibody strips1- or 2- minutes[73]
            HOLMESJEVFluorescence∼1 hour[74]
            HOLMESv2JEV, SNPFluorescence∼1 hour[75]
            CRISPR-actuate hydrogelsEbolaμPAD∼1 hour[76]
            Hydrazone chemistry-mediated P. aeruginosa Fluorescence[77]
            BM-TOAEGFR mutationsFluorescence[78]
            Cas13SHERLOCK E. coli, KPC, P. aeruginosa, ZIKV, DENVFluorescenceWithin 2 hours[79,80]
            CRMENMERS, SARS, HIV, ZIKV, DENV, HCV, HPV4, influenzaFluorescence[81]
            SHERLOCK+HUDSONZIKV, DENV, YFV, WNVFluorescenceWithin 2 hours[82]
            Treponema pallidum Fluorescence1-4 hours[83]
            Cas14DETECTR-Cas14HBoV1[46]
            HUDSON+Cas14SNP (iris color variability)[46]
            Diagnostic tools based on the type V CRISPR-Cas system
            CRISPR-Cas12

            DNA endonuclease-targeted CRISPR trans reporter (DETECTR), which is based on Cas12a coupled with RPA, is a well-known diagnostic method that utilizes a fluorophore quencher (FQ)-labeled reporter gene to monitor the trans-cleavage activity of Cas12a [48,60]. In the presence of pathogens, the DNA substrate typically binds to the Cas12a-gRNA complex to change the protein conformation of Cas12a. The nearby FQ-labeled ssDNA reporter is non-specifically cleaved by Cas12a, resulting in the release of FQ and recovery of the reporter fluorescence. Several Cas12-based diagnostic tools have been employed to detect various viruses, particularly SARS-CoV-2, following the global COVID-19 pandemic that emerged in late 2019 (Table 3). Notably, this system can quickly respond to emerging infectious pathogens and has a critical role in controlling epidemics and disease spread. Here, we provide an overview of a highly representative Cas12a-based tool developed for the detection of SARS-CoV-2.

            SARS-CoV-2 can be detected using an in vitro specific CRISPR-based assay for nucleic acid detection (iSCAN) [21], which combines a CRISPR-Cas12a detection system with the RT-LAMP technology. This novel detection platform, called two-pot iSCAN, has several benefits, including substantial time savings, high accuracy, enforceability, and ease of operation [21,41]. Most CRISPR-Cas12a-based detection tools, including iSCAN platform, typically involve two steps, which complicate the testing process and increase the risk of contamination [84]. POCT may require minimal liquid handling and “one-pot” reactions. Normally, this barrier can be overcome by mixing all reaction components into a single tube for a one-pot reaction [21,85]. The one-pot iSCAN system can be used for robust testing and identification of other pathogens and provides a new basis for the development of early pathogen detection platforms. The application of one-pot detection systems to the domain of self-inspection by non-technical personnel faces substantial hurdles due to difficulties in finding suitable materials and carriers to control the incorporation of the Cas12a/b-sgRNA complex into the reaction system. A technique recently developed by Broughton et al. [61] might be able to address this barrier.

            The CRISPR-Cas12a diagnostic system relies on the indiscriminate cleavage of ssDNA by Cas12a activation; however, there are only two activation modes based on single- or double-stranded activators, which hinder specificity and universality [78]. Further, the Cas12a activation velocity affects the performance of the detection system. Therefore, new activation patterns have been developed to address these issues. A novel approach to detect Pseudomonas aeruginosa, developed by Sheng et al. [77], activates the CRISPR-Cas12a system by harnessing hydrazone chemistry, which has superior reactivity and selectivity [86,87], and improves the specificity and flexibility of the method.

            To break through the technical bottleneck created by the presence of only two activation methods, Khodakov et al. [88] developed an unprecedented approach to Cas12 activation by using a toehold-mediated strand displacement reaction that strictly follows base-pairing rules. The toehold-mediated strand displacement reaction can also be applied for the detection of DNA point mutations such as that used in cancer diagnosis where DNA point mutations are a major cause of carcinogenesis and important markers of prognosis and diagnosis [8991]. A combination of specific techniques, such as RPA, LAMP, rolling circle amplification [92], and primer exchange reaction (PER) [93] with the CRISPR-Cas technology, can promote the development of diagnostic platforms that use a one-step reaction as promising POCT tools [78,94].

            CRISPR-Cas14

            Cas14, which was first reported in 2018 by the Doudna group [53], has several unique traits compared to Cas12 [46]. Cas14a has 530 amino acids, and is the smallest RNA-guided nuclease identified to date. Cas14 binds and cleaves ssDNA, and RNA is a central component of the Cas14a-sgRNA complex [37,46]. Cas14 does not require a target sequence, such as protospacer adjacent motif (PAM), for cleavage activity [95,96]. DETECTR-Cas14 is a recently developed diagnostic tool that combines an RPA tool with Cas14 and is based on the ability of Cas14 to cleave ssDNA through non-specific trans-cleavage activity [53]. This molecular scissors-based platform harnesses the ability of Cas14 to detect DNA with high fidelity to elicit the appearance of a fluorescent signal following the collateral cleavage of ssDNA [53]. This approach provides new possibilities for the detection of viruses [46,97]; however, the limited number of studies that have reported CRISPR-Cas14-based diagnostic platforms only elucidate the prospects of the application. The DETECTR-Cas14 platform makes it possible to study the effects of ssDNA on the human virome, which is an important component in understanding virus-disease associations [46,98]. The CRISPR-Cas14-based platform still requires nucleic acid extraction and amplification, a procedure that is undoubtedly cumbersome. A new approach that involves heating unextracted diagnostic samples to obliterate nucleases (HUDSON) in combination with CRISPR-Cas14 without nucleic acids extraction process and shipping of the samples simplifies the workflow [46,80].

            Furthermore, this new molecular scissors-based diagnostic tool exhibits great potential in SNP identification [53]. The CRISPR-Cas14 system has been applied to molecular diagnostics and SNP genotyping. In addition, CRISPR-Cas14 coupled with metal elements has also been harnessed for the sensitive detection of non-nucleic acid targets [99,100]. There is no doubt that CRISPR-Cas14 detects ssDNA viruses and foodborne bacteria in clinical settings; however, the CRISPR-Cas14-based diagnostic platforms have been explored to a more limited extent than the CRISPR-Cas12-based platforms. Therefore, the applicability for non-diagnostic purposes remains to be investigated.

            Diagnostic tools built on type VI CRISPR-Cas systems
            CRISPR-Cas13

            CRISPR-Cas13-based platforms are promising diagnostic tools and potential next-generation diagnostic strategies for virus detection. Specific high-sensitivity enzymatic reporter unlocking (SHERLOCK) based on Cas13 was reported prior to the introduction of DETECTR. Nonetheless, both approaches are based on similar detection principles and require combination with isothermal amplification technologies [80]. The CRISPR-Cas13-based diagnostic platforms directly detect RNA viruses, such as ZIKV, DENV, HIV, SARS, and SARS-CoV-2, because Cas13 identifies RNA substrates rather than DNA substrates. Isothermal DNA amplification using RT-RPA also detects DNA viruses [80]. Furthermore, SHERLOCK has been shown to detect pathogenic bacteria, such as KPC and NDM-1. The next version of SHERLOCK (SHERLOCKv2) has been reported to provide a robust, 3.5-fold increase in sensitivity by coupling Cas13b to Csm6 [79].

            Although SHERLOCK can specifically and precisely detect ZIKV and DENV at the level of 1 copy/μL [82], complicated operating procedures, especially nucleic acid extraction, have hindered widespread utility. HUDSON was developed to compensate for this deficiency and to simplify SHERLOCK by reducing lysed viral particles and deactivating RNases in body fluids [80,82]. HUDSON permits the use of RPA after HUDSON to directly detect virus from body fluids without the need for nucleic acid extraction, dilution, and purification [101]. A viral diagnostic platform that pairs SHERLOCK and HUDSON has been constructed to effectively and rapidly detect ZIKV and DENV [82]. This new platform is highly sensitive and specific and can be used without instruments. More importantly, it is a potential platform for multiplexing detection of viruses [79].

            In 2020, Ackerman et al. [81] introduced combinatorial arrayed reactions for multiplexed evaluation of nucleic acids (CARMEN) as a flexible platform that enables highly multiplexed detection of pathogens. The combination of CARMEN with Cas13 (CARMEN-Cas13) stably detects >4500 crRNAs-substrate complexes in a single array. The CARMEN-Cas13 platform detects Middle East respiratory syndrome virus, SARS, SARS-CoV-2, HIV, ZIKV, DENV, hepatitis C virus, and HPV4, but not γHPV or Aroa virus. The flexibility and high throughput of CARMEN allow the introduction of crRNAs or amplification primers to detect emerging and re-emerging pathogens. Additionally, CARMEN complements the shortcomings of next-generation sequencing. In summary, the CARMEN-Cas13 platform expands the application of CRISPR-based platforms for pathogen detection by providing flexible testing.

            RAPID DETECTION PLATFORMS BASED ON APTAMERS

            CRISPR-Cas-based diagnostic platforms are cumbersome due to complex manipulations that are needed, such as extraction and amplification of nucleic acids. These tools have shown robust functionality in pathogen detection. Simple, convenient, and highly effective tools are urgently needed. Aptamer-based biosensor detection methods have subsequently attracted attention to address this issue.

            Aptamers are 20–80-base ssDNA or RNA sequences identified by screening of nucleic acid libraries with the systematic evolution of ligands using an exponential enrichment (SELEX) strategy [102,103]. Aptamers are excellent alternatives to antibodies for antigen-specific identification [104]. Furthermore, aptamers permit stable immobilization of target molecules, such as viruses, bacteria, amino acids, proteins, and cells [105]. Typically, the screened aptamer binds specifically to the targeted substrate and indirectly detects pathogens based on physical, chemical, or electrochemical properties, such as changes in current or electrical resistance or the naked-eye recognition of fluorescence. Therefore, platforms based on aptamer-based biosensors (aptasensors) have been broadly used to detect pathogenic viruses and foodborne bacteria in clinical settings or diagnosis of human diseases (Fig 1). Aptasensors are divided into optical and electronic biosensors depending on the type of transducer.

            Detection of bacteria via aptamer-based tools

            Bacteria colonize a wide range of nature environments, and infection with pathogenic bacteria can cause severe clinical illness. Therefore, the early and rapid detection of bacteria in food or water samples is essential. Currently, most of the described aptamer-based detection tools are used for the detection of E. coli and S. aureus.

            E. coli is a pathogenic microorganism widely found in nature and the human gut. E. coli serotypes can cause various clinical symptoms, such as diarrhea, abdominal pain, ulcers, and hemolysis [106,107]. S. aureus is a common pathogen that causes infectious diseases, such as pneumonia, arthritis, and sepsis [108]. In addition, following the abuse of antibiotics in clinical settings and the addition to poultry feed, S. aureus has gradually become insensitive to clinical treatment, a considerable challenge for human health [109]. Early and rapid diagnosis is very important in efforts to curb the spread of E. coli and S. aureus, guide medical treatment, and detect bacteria in food and water sources. Development of methods for the quantitative and rapid detection of E. coli and S. aureus has attracted the attention of food safety, medicine, and other fields, and fast and precise detection techniques based on aptamers have been exploited. The combination of optical or electrochemical sensors with aptamers has been widely used to test E. coli and S. aureus proteins, and even whole cells.

            Aptamers assembled with optical sensors for the detection of E. coli and S. aureus

            Visible light, ultraviolet light, and fluorescent sensors can be used to detect E. coli using aptamers; these approaches provide several advantages, including ease of operation and low cost compared to ELISA, which is a conventional technique that requires an abundant amount of antibodies and is expensive [110]. Several optical-sensors-based tools for detecting E. coli have already been developed. In 2016, Fu et al. [111] developed a method based on sandwich-like assay to detect E. coli K88. The authors introduced biotin-labeled and Au nanoparticle (NP)-labeled aptamers to create a sandwich-type complex (biotin-labeled aptamer I/E. coli K88/NP-labeled aptamer II). The presence of E. coli K88 was visualized based on a color reaction, and the assay had a linear range of 10–105 colony-forming units (CFU)/mL. A colorimetric method based on an aptamer-polydiacetylene optical sensor was reported to detect E. coli O157:H7 at ranges between 104 and 108 CFU/mL [112]. Pandit et al. [113] developed a new method to detect E. coli in water samples. Specifically, two E. coli-specific aptamers, which were identified by screening, were separately conjugated with superparamagnetic iron oxide NPs and CdTe-MPA quantum dots, respectively. The use of CdTe-MPA quantum dots allows the qualitative and quantitative estimation of E. coli by fluorescence spectrophotometry. The limit of detection (LOD) of this biosensor is as high as 102 CFU. Yuan et al. [114] developed a gold NP-based colorimetric aptasensor to detect S. aureus based on tyramine signal amplification technology. In this method, the biotinylated anti-S. aureus aptamer is immobilized on the surface of a microtiter plate before the addition of other necessary ingredients (biotin-tyramine, streptavidin-horseradish peroxidase, avidin-catalase, and S. aureus). The catalase added during the assay depletes endogenous hydrogen peroxide. Finally, gold (III) chloride trihydrate is used to change the color of the reaction product and the presence of S. aureus is determined by measuring absorbance at 550 nm. The assay could detect S. aureus ranging from 10-106 CFU/mL under ideal conditions, and the LOD was 9 CFU/mL. Aptamer-based techniques using fluorescence for the detection of E. coli and S. aureus exhibit higher sensitivity than techniques based on visual and colorimetric assessment. Fluorescence aptamer-based techniques often require sophisticated instruments, such as epifluorescence, confocal fluorescence, and confocal laser scanning microscopy.

            Aptamers assembled with electrochemical sensors for the detection of E. coli and S. aureus

            Electrochemical signals can be displayed by current, resistance, or potential, offering several possibilities for constructing electrochemical biosensors for E. coli and S. aureus testing [110]. Notably, aptamers coupled to electrochemical sensor detection platforms have the potential to achieve tractability and miniaturization and have been reported to directly detect pathogens in turbid media. A study recently described a method to detect the outer membrane proteins (OMPs) of E. coli through a change in resistance [115]. In that assay, potassium ferricyanide was used as a redox probe for electron exchange between the gold electrodes. E. coli OMPs combined with aptamers could hinder this process, leading to an electrochemical reaction. The relationship between the concentration of E. coli OMPs (1 × 10−7 – 2 × 10−6 mol/L) and electron transfer resistance was excellent. In addition to OMPs, E. coli lipopolysaccharide can be used as a detection site [116]. Furthermore, aptamer-based electrochemical sensors can be used to detect E. coli genes. For example, E. coli ribosomal RNA was detected by monitoring the oxidation state of guanine nucleotides [117], and E. coli genomic DNA was directly recognized by utilizing methylene blue as a DNA hybridization probe molecule [118]. Several tools have emerged for detecting whole cells of E. coli [119121].

            Nguyen et al. [122] reported a new method for the detection of S. aureus. Nguyen et al. [122] utilized a cognate pair of aptamers for S. aureus to fabricate a sandwich-type aptasensor (aptamer I/S. aureus/horseradish peroxidase-conjugated aptamer II) on a screen-printed gold electrode. S. aureus can be detected based on a significant decrease in faradaic peak current and peak-to-peak potential separation. The predicted LOD of this sandwich-type signal-on electrochemical biosensor was 39 CFU in buffer and 414 CFU in commercially available bottled water samples. Further, the new biosensor could easily detect S. aureus in a sample volume as low as 5 μL. As rapid, sample, sensitive, and specific approaches, sandwich-type electrochemical aptasensors have promising applicability in food safety management.

            Detecting viruses via aptamer-based tools

            Aptamer-based biosensors have been developed for bacterial and viral detection. Table 5 summarizes the aptasensor-based techniques for the detection of viruses. Optical aptasensors for virus detection can be categorized into six groups. Surface plasmon resonance (SPR) aptasensors are based on the detection of resonance of free electrons based on the change in refractive index. Aptamers are prefixed to the surface of a gold plate, and the thickness of the gold surface is altered after the capture of the virus; the presence of the virus is determined by measuring changes in the angle or the intensity of polarized light. Colorimetric aptasensors determine the presence of virus based on an apparent shift in color, which can be directly observed with the naked eye or using a spectrophotometer. Fluorescence aptasensors are based on the transition of fluorescence signals or the production of fluorescence polarization. Chemiluminescence (CL) aptasensors are commonly and extensively used to detect viruses, such as hepatitis B virus, HIV, SARS-CoV-2, and other emerging and re-emerging infectious viruses in clinical practice. These assays are based on colorimetric detection. The aptamer is co-incubated with virus particles, followed by the addition of catalytically-active complexes that bind to the virus, and the addition of a chromogenic or CL reagent for the quantitative detection of the virus. Surface-enhanced Raman scattering aptasensors utilize Raman scattering spectroscopy, which reflects the characteristics of the medium by the change in the energy of photons after passing a laser light through the medium. Interferometric aptasensors are the sixth category developed on the basis of interferometry. It is a label-free technique for measuring light intensity, which includes a refractive index or physical property.

            TABLE 5 |

            Aptasensors applied in virus detection.

            DetectionTechniqueVirusLimit of DetectionRefs.
            Optical aptasensorsSPR(AIV) H5N10.128 HAU[123]
            200 EID50/mL[124]
            HIV-10.12 ppm[125]
            ColorimetricMNV200 MNV/mL[126]
            H3N211.16 μg/mL[127]
            H5N10.1 μg/well[128]
            H5N21.27 × 105 EID50/mL[129]
            HCV11 nM[130]
            3.91 × 102 FFU/mL[131]
            HuNoV10 RNA copies[132]
            ZIKA virus0.1 ng/mL[133]
            Multiplex strain-specific influenza virus2 × 106 virus particles[134]
            SFTS virus9 pg/mL[135]
            SARS-CoV-210 ng/mL[136]
            1 ng/mL[137]
            FluorescenceHBV1.25 mIU/mL[138]
            SARS-CoV-2130 fg/mL[139]
            RSV80 nm[140]
            H1N13.45 nM[141]
            CLSARS-CoV2 pg/mL[142]
            HBV0.1 ng/mL[143]
            SERSH1N197 PFU/mL[144]
            InterferometryHCV700 pg/mL[145]
            Electronic aptasensorsElectrochemical aptasensorsH1N110 pM[146]
            HCV0.16 fg/mL[147]
            3.3 pg/mL[148]
            H5N18 × 10−4 HAU/200 μL[149]
            (AIV) H5N12−9 HAU[150]
            HIV-11 nM[151]
            SARS-CoV-21.6 × 10 PFU/mL[152]
            Noro viruses180 virus particles[153]
            Piezoelectric transducersHIV-10.25 mg/L[154]
            (AIV) H5N10.0128 HAU[155]
            HCV0.1 pM[156]

            Furthermore, viruses can be detected with electronic biosensors, which are a type of electrochemical aptasensor, or piezoelectric transducers based on different mechanisms. Electrochemical aptasensors follow the principle of a change in electrical signal caused by the binding of the aptamer to the target virus. Aptamers used to capture viral molecules are prefixed on electrode plates. Detection tools based on electrochemical aptasensors can be further categorized into three subclasses: enzymatically-labeled aptasensors, in which enzyme catalysis triggers a change in the electrical signals; enzyme-free aptasensors, in which the binding of viral particles to the aptasensors directly causes a change in the electrical signal; and field-effect transistor (FET)-based aptasensors. The aptamer is immobilized on the FET surface to capture target viral molecules and the change in the charge distribution on the FET surface is detected and translated into an easy-to-read signal in FET-based aptasensor detection tools. Several viral assays based on these technologies have been developed (Table 5). Piezoelectric transducers are based on the piezoelectric effect principle, in which an electrical charge is generated when certain materials are subjected to mechanical stress.

            Detecting parasites via aptamer-based tools

            Parasites are a large family of pathogens that cause zoonoses. Seven of the top 10 global tropical diseases are caused by parasites. As such, parasites are a serious threat to human health and a significant burden to the economy, both of which can be mitigated by the rapid detection of parasites. Recently developed aptamer-based tools for parasite detection provide rapid and accurate approaches with a low LOD and have been widely implemented to identify and monitor various parasites.

            Roca et al. [157] selected a DNA aptamer (D10) against Plasmodium falciparum 1-deoxy-D-xylulose-5-phosphate reductoisomerase (DXR) using the SELEX strategy, a vital enzyme in the methyl erythritol phosphate (MEP) pathway of isoprenoid biosynthesis. The D10 aptamer binds to recombinant DXR in vitro and explicitly targets the apicoplast, in which the MEP pathway is localized, an essential organelle in P. falciparum. Therefore, D10 is a highly specific marker for naive erythrocytes and red blood cells parasitized by P. falciparum and for the apicoplast. Therefore, the D10 aptamer is a promising lead for future diagnostic strategies for Plasmodium and a potential tool for other studies of apicomplexan parasites, such as Toxoplasma gondii. In addition, the D10 aptamer detects bacteria harboring the MEP pathway, such as E. coli and P. aeruginosa. To address the lack of aptamer-based methods for the detection of SAG-1, the major T. gondii surface protein, Cui et al. [158] developed a direct enzyme-linked aptamer assay to target native SAG-1 (nSAG-1) for the detection of T. gondii. This novel technique is a potential tool for the rapid and precise diagnosis of toxoplasmosis. Homann and Goringer [159] was the first to employ the SELEX strategy to screen aptamers for the detection of African trypanosomes. The authors identified an RNA aptamer that crosslinked to a 42-kDa protein in the flagellar capsule of trypanosomes through 13 rounds of SELEX selection. Interestingly, although aptamer-based assays have been proposed for the detection of parasites, with many tools developed over the years, only a handful of reports use whole cells as detection targets. Commonly, the screening substrates of aptamers for parasites are enzymes, proteins, and infected cells [160162] and these aptamers detect the secretions and excretions of the target parasite or specific virulence-related molecules.

            RAPID DETECTION PLATFORMS COUPLING CRISPR-CAS- AND APTAMER-BASED SYSTEMS

            CRISPR-Cas- and aptamer-based techniques utilize many tools for pathogen detection and exhibit robust efficacy, sensitivity, specificity, and convenience compared to conventional detection approaches; however, cumbersome procedures and the detection of the LOD are issues that remain to be resolved. For example, CRISPR-Cas-based tools require nucleic acid extraction and amplification. Conversely, multiple rounds of aptamer screening are essential for aptamer-based platforms and detection with aptamers only does not achieve good sensitivity and the LOD is usually in the picomolar and micromolar ranges [3,163]. A study recently reported that these two complementary techniques can be combined to develop effective detection tools for pathogen detection to address these obstacles (Fig 2). Notably, this cross-cutting approach has been used to detect pathogenic bacteria in samples, and should be studied more for the diagnosis of viruses or parasites, and therefore has promising applications in the POCT of foodborne bacteria. This CRISPR-Cas- and aptamer-based detection platform plays an essential role in preventing the spread of pathogens in resource-poor countries and regions. Some of the complementary tools can distinguish between dead and living bacteria, providing a feasible method for further control of pathogenic bacteria at the source.

            FIGURE 2 |

            Schematic illustration of CRISPR-Cas systems coupled with aptamers for pathogen detection. Aptamers are usually prestabilized in various ways. In the presence of the target pathogen, the aptamer dissociates from the system and the remaining components synthesize a short-chain nucleic acid to activate the Cas protein which then cleavages the indicated probe, leading to a change in fluorescence or current.

            Diagnostic tools built on the CRISPR-Cas12 system coupled with aptamers

            Bu et al. [164] constructed an electrochemical aptasensor to detect pathogenic bacteria without nucleic acid extraction by combining the CRISPR-Cas12a system with PER. In this approach, the DNA aptamer binds to the hairpin loop of the primer, which remains inactive in the absence of pathogens, thereby hindering primer amplification. In the presence of target bacteria, the DNA aptamer disentangles the hairpin loop structure and the primer is extended to form ssDNA, which activates the trans-cleavage activity of CRISPR-Cas12a to crush the MB probe pre-immobilized on the Au surface, resulting in a decrease in the monitored peak current. This novel technique has been utilized to detect bacteria in milk samples, such as the detection of E. coli O157:H7 with an LOD of 19 CFU/mL. To expand the repertoire of diagnostic methods for various disease biomarkers, Yuan et al. [165] recently developed a “signal on-off-super on” sandwich-type aptasensor of CRISPR-Cas12a coupled voltage enrichment for the detection of vascular endothelial growth factor with an LOD of 0.33 pM. Using this technique, AuNPs@Ti3C2Tx-Mxene was synthesized by immobilizing AuNPs on the surface of Ti3C2Tx-Mxene, which stabilized the trapped aptamer and therefore altered the electrochemical signal more effectively. This approach enriched ssDNA using an electrochemical signal via CRISPR-Cas12a-coupled voltage enrichment instead of amplification.

            Diagnostic tools built on the CRISPR-Cas13 system coupled with aptamers

            Many platforms combining CRISPR-Cas13 with aptamers have been developed for pathogen detection. The difference between these platforms and the CRISPR-Cas12a-based platforms lies in the Cas enzyme. Shen et al. [55] presented the new APC-Cas platform for the detection of S. enteritidis. The allosteric probe used in this platform contains three sections: the T7 promoter; an aptamer; and a primer-binding site. The probe is in a quiescent state due to the integration of the primer-binding site into the aptamer. In the presence of the target pathogen, the allosteric probe undergoes conformational changes and ssRNA is generated using the T7 RNA polymerase. The platform exhibits strong sensitivity and functionality for the quantitative detection of S. enteritidis and is a promising approach for pathogen detection. Zhang et al. [25] reported a system to distinguish between viable and dead pathogenic bacteria using a light-up RNA aptamer combined with CRISPR-Cas13; the LOD of this system was 10 CFU for the detection of Bacillus cereus.

            Diagnostic tools built on the CRISPR-Cas14 system coupled with aptamers

            Cas14 is the smallest identified Cas enzyme and does not require PAM sequence identification; therefore, CRISPR-Cas14-based aptasensors exhibit enhanced sensitivity and versatility compared to the biosensors based on Cas12 or Cas13. Although reports of Cas14-based aptasensors are rare, Cas14-based aptasensors may be more potent. AcasB, a novel platform developed for on-site detection of live S. aureus, contains an aptamer biosensor combined with Cas14a1 [3]. This platform includes 3 probes, a 45-nucleotide aptamer to target live S. aureus, a 21-nucleotide blocker for activation of Cas14a1, and an sgRNA to bind the blocker. Importantly, this platform does not require nucleic acid extraction and amplification. The blocker bound to the aptamer forms a stable hybrid, which dissociates to release the blocker in the presence of live S. aureus and specifically targets the aptamer. The subsequently released blocker activates Cas14a1 by binding to sgRNA, which then cleaves the surrounding fluorescence probe, resulting in fluorescence. This platform directly distinguishes whether the bacteria are alive or dead and has an LOD of 400 CFU/mL for live S. aureus in the tested samples.

            Cas14 combined with aptamers can also be used to detect non-nucleic-acids targets. Zhou et al. [166] developed a biosensor platform designated as a highly-sensitive aptamer-regulated Cas14 Rloop for bioanalysis (HARRY). A diblock ssDNA was designed to accommodate the aptamer and Cas14 activation sequences in this platform. The ssDNA activates Cas14a, then the Cas14a trans-cleavages the fluorescent reporter, causing fluorescence enhancement in the absence of the target. Non-nucleic acids substrates, such as ATP, Cd2+, histamine, aflatoxin B1, and thrombin, have been detected by the HARRY platform, which possesses LOD at the nanomolar level.

            CONCLUSIONS AND FUTURE PERSPECTIVES

            Pathogens pose a severe threat to the survival of humans and animals, and are a public health issue of concern. CRISPR-Cas- and aptamer-based systems have been harnessed for POCT research primarily based on the application of CRISPR-Cas systems for pathogen detection. Conventional diagnostic methods are complicated and cumbersome. Therefore, diagnostic platforms based on the CRISPR-Cas system and aptamers have been exploited to improve detection efficiency and simplify the procedures, while reducing cost and improving applicability. Nevertheless, most of these tools are theoretical and few have been introduced for commercial use. The reasons that prevent the marketization of these tools, i.e., nucleic acid extraction and amplification, are a necessary component of CRISPR-Cas-based approaches, which are cumbersome processes that require skill. Notably, technological advances, such as isothermal amplification, microfluidics, and gene chips, should overcome these barriers [78]. SELEX is a time-consuming, not-yet-mature, costly, and unstable strategy, and sensitive simultaneous detection of multiple pathogens in a mixed medium is challenging. For clinical samples, the selected aptamers cannot be completely and accurately matched, and the affinity and specificity change [29]. Recent studies achieved the detection of pathogens in turbid media and the optimization of the SELEX screening process. Consequently, combination of the complementary CRISPR-Cas- and aptamer-based systems is an effective optimization approach to fill the gaps in pathogen detection. Indeed, the combination of these two systems has gradually replaced the traditional detection methods over the past decade. However, combinations of various Cas proteins with different aptamer techniques, such as allosteric probe-initiated catalysis and PER, are typically limited to bacterial detection, therefore, future studies should be conducted to advance viruses and parasite detection in clinical settings.

            The recently described CRISPR FISHer [167] can potentially track particular cell-intrinsic or exogenous DNA sequences, broadening the application range of live cell imaging and laying the foundation for biomedical diagnostics. Several recent studies have identified new Cas proteins in nature. For instance, Cas12a2 [168,169] is activated upon binding to RNA targets and can efficiently degrade ssRNA, ssDNA, and dsDNA. In contrast to the previously described Cas proteins, Cas12a2 is programable and capable of pathogen detection. Cas14 is a newly discovered and promising protein for the development of more efficient and convenient detection tools, while its use in conjunction with aptamer-based systems for the detection of non-nucleic acids offers a new idea for broadening diagnostic targets and establishing detection methods. Many aspects of the mechanism underlying the Cas protein effect, both newly and previously discovered, are still not fully understood. In addition, combining several Cas proteins with different functions to enable simultaneous pathogen detection and treatment is an intriguing potential approach.

            In conclusion, pathogen detection tools should be practical and generalizable, ideally with test results linked to a smartphone or smart bracelet, enabling citizens to self-test for pathogens in food or water and alleviating stress. With the constant expansion of technology, there is great promise for improvement in diagnostics, and there is an urgent need to establish novel detection tools to respond rapidly to emerging zoonotic pathogens in the near future.

            CONFLICTS OF INTEREST

            The authors declare no conflicts of interest.

            REFERENCES

            1. Keesing F, Belden LK, Daszak P, Dobson A, Harvell CD, Holt RD, et al.. Impacts of biodiversity on the emergence and transmission of infectious diseases. Nature. 2010. Vol. 468(7324):647–652

            2. Mertz GJ. Zoonoses: infectious diseases transmissible from animals to humans, fourth edition. Clinical Infectious Diseases. 2016. Vol. 63(7):148–149

            3. Wei Y, Tao Z, Wan L, Zong C, Wu J, Tan X, et al.. Aptamer-based Cas14a1 biosensor for amplification-free live pathogenic detection. Biosens Bioelectron. 2022. Vol. 211:114282

            4. World Health Organization. https://www.who.int/news-room/fact-sheets/detail/zoonosesAccessed on date 11/2022

            5. Jones KE, Patel NG, Levy MA, Storeygard A, Balk D, Gittleman JL, et al.. Global trends in emerging infectious diseases. Nature. 2008. Vol. 451(7181):990–993

            6. Chinese Centers for Disease Control and Prevention. https://www.cdc.gov/onehealth/basics/zoonotic-diseases.htmlAccessed on date 11/2022

            7. Jones SD, Atshabar B, Schmid BV, Zuk M, Amramina A, Stenseth NC. Living with plague: lessons from the Soviet Union’s antiplague system. Proc Natl Acad Sci U S A. 2019. Vol. 116(19):9155–9163

            8. Suarez-Esquivel M, Chaves-Olarte E, Moreno E, Guzman-Verri C. Brucella genomics: macro and micro evolution. Int J Mol Sci. 2020. Vol. 21(20):7749

            9. Van Kerkhove MD, Ryan MJ, Ghebreyesus TA. Preparing for “Disease X”. Science. 2021. Vol. 374(6566):377

            10. Sauter D, Kirchhoff F. Key viral adaptations preceding the AIDS pandemic. Cell Host Microbe. 2019. Vol. 25(1):27–38

            11. Ogbu O, Ajuluchukwu E, Uneke CJ. Lassa fever in West African sub-region: an overview. J Vector Borne Dis. 2007. Vol. 44(1):1–11

            12. Rewar S, Mirdha D. Transmission of Ebola virus disease: an overview. Ann Glob Health. 2014. Vol. 80(6):444–451

            13. Hills SL, Morrison A, Stuck S, Sandhu K, Mason KL, Stanek D, et al.. Case series of laboratory-associated Zika virus disease, United States, 2016-2019. Emerg Infect Dis. 2021. Vol. 27(5):1296–1300

            14. Hotchkiss EJ. Zoonotic cryptosporidiosis - challenges for control and source attribution. Vet Rec. 2021. Vol. 189(4):150–152

            15. Cai W, Ryan U, Xiao L, Feng Y. Zoonotic giardiasis: an update. Parasitol Res. 2021. Vol. 120(12):4199–4218

            16. Bishara J, Leibovici L, Gartman-Israel D, Sagie A, Kazakov A, Miroshnik E, et al.. Long-term outcome of infective endocarditis: the impact of early surgical intervention. Clin Infect Dis. 2001. Vol. 33(10):1636–1643

            17. van de Beek D, Brouwer M, Hasbun R, Koedel U, Whitney CG, Wijdicks E. Community-acquired bacterial meningitis. Nat Rev Dis Primers. 2016. Vol. 2(1):16074

            18. Dohla M, Boesecke C, Schulte B, Diegmann C, Sib E, Richter E, et al.. Rapid point-of-care testing for SARS-CoV-2 in a community screeing setting shows low sensitivity. Public Health. 2020. Vol. 182(182):170–172

            19. Niemz A, Ferguson TM, Boyle DS. Point-of-care nucleic acid testing for infectious diseases. Trends Biotechnol. 2011. Vol. 29(5):240–250

            20. Ong D, Fragkou PC, Schweitzer VA, Chemaly RF, Moschopoulos CD, Skevaki C. How to interpret and use COVID-19 serology and immunology tests. Clin Microbiol Infect. 2021. Vol. 27(7):981–986

            21. Ali Z, Aman R, Mahas A, Rao GS, Tehseen M, Marsic T, et al.. iSCAN: an RT-LAMP-coupled CRISPR-Cas12 module for rapid, sensitive detection of SARS-CoV-2. Virus Res. 2020. Vol. 288:198129

            22. Morshed MG, Lee MK, Jorgensen D, Isaac-Renton JL. Molecular methods used in clinical laboratory: prospects and pitfalls. FEMS Immunol Med Microbiol. 2007. Vol. 49(2):184–191

            23. Qi X, Wang Z, Lu R, Liu J, Li Y, Chen Y. One-step and DNA amplification-free detection of Listeria monocytogenes in ham samples: combining magnetic relaxation switching and DNA hybridization reaction. Food Chem. 2021. Vol. 338:127837

            24. Sheng L, Lu Y, Deng S, Liao X, Zhang K, Ding T, et al.. A transcription aptasensor: amplified, label-free and culture-independent detection of foodborne pathogens via light-up RNA aptamers. Chem Commun (Camb). 2019. Vol. 55(68):10096–10099

            25. Zhang T, Zhou W, Lin X, Khan MR, Deng S, Zhou M, et al.. Light-up RNA aptamer signaling-CRISPR-Cas13a-based mix-and-read assays for profiling viable pathogenic bacteria. Biosens Bioelectron. 2021. Vol. 176:112906

            26. Mori Y, Notomi T. Loop-mediated isothermal amplification (LAMP): a rapid, accurate, and cost-effective diagnostic method for infectious diseases. J Infect Chemother. 2009. Vol. 15(2):62–69

            27. Notomi T, Okayama H, Masubuchi H, Yonekawa T, Watanabe K, Amino N, et al.. Loop-mediated isothermal amplification of DNA. Nucleic Acids Res. 2000. Vol. 28(12):E63

            28. Pang B, Xu J, Liu Y, Peng H, Feng W, Cao Y, et al.. Isothermal amplification and ambient visualization in a single tube for the detection of SARS-CoV-2 using loop-mediated amplification and CRISPR technology. Anal Chem. 2020. Vol. 92(24):16204–16212

            29. Zou X, Wu J, Gu J, Shen L, Mao L. Application of aptamers in virus detection and antiviral therapy. Front Microbiol. 2019. Vol. 10:1462

            30. Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol. 1987. Vol. 169(12):5429–5433

            31. Ding W, Zhang Y, Shi S. Development and application of CRISPR/Cas in microbial biotechnology. Front Bioeng Biotechnol. 2020. Vol. 8:711

            32. Chen JS, Ma E, Harrington LB, Da Costa M, Tian X, Palefsky JM, et al.. Erratum for the report “CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity”. Science. 2021. Vol. 371(6531):eabh0317

            33. East-Seletsky A, O’Connell MR, Knight SC, Burstein D, Cate JH, Tjian R, et al.. Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature. 2016. Vol. 538(7624):270–273

            34. Makarova KS, Wolf YI, Alkhnbashi OS, Costa F, Shah SA, Saunders SJ, et al.. An updated evolutionary classification of CRISPR-Cas systems. Nat Rev Microbiol. 2015. Vol. 13(11):722–736

            35. Sorek R, Lawrence CM, Wiedenheft B. CRISPR-mediated adaptive immune systems in bacteria and archaea. Annu Rev Biochem. 2013. Vol. 82:237–266

            36. Amitai G, Sorek R. CRISPR-Cas adaptation: insights into the mechanism of action. Nat Rev Microbiol. 2016. Vol. 14(2):67–76

            37. Paul B, Montoya G. CRISPR-Cas12a: functional overview and applications. Biomed J. 2020. Vol. 43(1):8–17

            38. van der Oost J, Westra ER, Jackson RN, Wiedenheft B. Unravelling the structural and mechanistic basis of CRISPR-Cas systems. Nat Rev Microbiol. 2014. Vol. 12(7):479–492

            39. Wright AV, Nuñez JK, Doudna JA. Biology and applications of CRISPR systems: harnessing nature’s toolbox for genome engineering. Cell. 2016. Vol. 164(1-2):29–44

            40. Nishimasu H, Nureki O. Structures and mechanisms of CRISPR RNA-guided effector nucleases. Curr Opin Struct Biol. 2017. Vol. 43:68–78

            41. Rahimi H, Salehiabar M, Barsbay M, Ghaffarlou M, Kavetskyy T, Sharafi A, et al.. CRISPR systems for COVID-19 diagnosis. ACS Sens. 2021. Vol. 6(4):1430–1445

            42. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al.. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013. Vol. 339(6121):819–823

            43. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014. Vol. 346(6213):1258096

            44. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012. Vol. 337(6096):816–821

            45. Xin C, Yin J, Yuan S, Ou L, Liu M, Zhang W, et al.. Comprehensive assessment of miniature CRISPR-Cas12f nucleases for gene disruption. Nat Commun. 2022. Vol. 13(1):5623

            46. Aquino-Jarquin G. CRISPR-Cas14 is now part of the artillery for gene editing and molecular diagnostic. Nanomedicine. 2019. Vol. 18:428–431

            47. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al.. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013. Vol. 152(5):1173–1183

            48. Kim S, Ji S, Koh HR. CRISPR as a diagnostic tool. Biomolecules. 2021. Vol. 11(8):1162

            49. Moradpour M, Abdulah S. CRISPR/dCas9 platforms in plants: strategies and applications beyond genome editing. Plant Biotechnol J. 2020. Vol. 18(1):32–44

            50. Xu H, Wang J, Deng Y, Hou F, Fu Y, Chen S, et al.. Two-color CRISPR imaging reveals dynamics of herpes simplex virus 1 replication compartments and virus-host interactions. J Virol. 2022. Vol. 96(24):e0092022

            51. Knipe DM, Cliffe A. Chromatin control of herpes simplex virus lytic and latent infection. Nat Rev Microbiol. 2008. Vol. 6(3):211–221

            52. Guk K, Keem JO, Hwang SG, Kim H, Kang T, Lim E, et al.. A facile, rapid and sensitive detection of MRSA using a CRISPR-mediated DNA FISH method, antibody-like dCas9/sgRNA complex. Biosens Bioelectron. 2017. Vol. 95:67–71

            53. Harrington LB, Burstein D, Chen JS, Paez-Espino D, Ma E, Witte IP, et al.. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science. 2018. Vol. 362(6416):839–842

            54. Qiu M, Zhou XM, Liu L. Improved strategies for CRISPR-Cas12-based nucleic acids detection. J Anal Test. 2022. Vol. 6(1):44–52

            55. Shen J, Zhou X, Shan Y, Yue H, Huang R, Hu J, et al.. Sensitive detection of a bacterial pathogen using allosteric probe-initiated catalysis and CRISPR-Cas13a amplification reaction. Nat Commun. 2020. Vol. 11(1):267

            56. Burstein D, Harrington LB, Strutt SC, Probst AJ, Anantharaman K, Thomas BC, et al.. New CRISPR-Cas systems from uncultivated microbes. Nature. 2017. Vol. 542(7640):237–241

            57. Liu JJ, Orlova N, Oakes BL, Ma E, Spinner HB, Baney K, et al.. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature. 2019. Vol. 566(7743):218–223

            58. Tsuchida CA, Zhang S, Doost MS, Zhao Y, Wang J, O’Brien E, et al.. Chimeric CRISPR-CasX enzymes and guide RNAs for improved genome editing activity. Mol Cell. 2022. Vol. 82(6):1199–1209.e6

            59. Wang Y, Qi T, Liu J, Yang Y, Wang Z, Wang Y, et al.. A highly specific CRISPR-Cas12j nuclease enables allele-specific genome editing. Sci Adv. 2023. Vol. 9(6):eabo6405

            60. Chen JS, Ma E, Harrington LB, Da Costa M, Tian X, Palefsky JM, et al.. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science. 2018. Vol. 360(6387):436–439

            61. Broughton JP, Deng X, Yu G, Fasching CL, Servellita V, Singh J, et al.. CRISPR-Cas12-based detection of SARS-CoV-2. Nat Biotechnol. 2020. Vol. 38(7):870–874

            62. Wang X, Zhong M, Liu Y, Ma P, Dang L, Meng Q, et al.. Rapid and sensitive detection of COVID-19 using CRISPR/Cas12a-based detection with naked eye readout, CRISPR/Cas12a-NER. Sci Bull (Beijing). 2020. Vol. 65(17):1436–1439

            63. Ding X, Yin K, Li Z, Liu C. All-in-one dual CRISPR-Cas12a (AIOD-CRISPR) assay: a case for rapid, ultrasensitive and visual detection of novel coronavirus SARS-CoV-2 and HIV virus. bioRxiv.

            64. Joung J, Ladha A, Saito M, Kim NG, Woolley AE, Segel M, et al.. Detection of SARS-CoV-2 with SHERLOCK one-pot testing. N Engl J Med. 2020. Vol. 383(15):1492–1494

            65. Sun Y, Yu L, Liu C, Ye S, Chen W, Li D, et al.. One-tube SARS-CoV-2 detection platform based on RT-RPA and CRISPR/Cas12a. J Transl Med. 2021. Vol. 19(1):74

            66. Jiang Y, Hu M, Liu AA, Lin Y, Liu L, Yu B, et al.. Detection of SARS-CoV-2 by CRISPR/Cas12a-enhanced colorimetry. ACS Sens. 2021. Vol. 6(3):1086–1093

            67. Lu S, Tong X, Han Y, Zhang K, Zhang Y, Chen Q, et al.. Fast and sensitive detection of SARS-CoV-2 RNA using suboptimal protospacer adjacent motifs for Cas12a. Nat Biomed Eng. 2022. Vol. 6(3):286–297

            68. Guo L, Sun X, Wang X, Liang C, Jiang H, Gao Q, et al.. SARS-CoV-2 detection with CRISPR diagnostics. Cell Discov. 2020. Vol. 6:34

            69. Hou T, Zeng W, Yang M, Chen W, Ren L, Ai J, et al.. Development and evaluation of a rapid CRISPR-based diagnostic for COVID-19. PLoS Pathog. 2020. Vol. 16(8):e1008705

            70. Rauch JN, Valois E, Solley SC, Braig F, Lach RS, Audouard M, et al.. A scalable, easy-to-deploy protocol for Cas13-based detection of SARS-CoV-2 genetic material. J Clin Microbiol. 2021. Vol. 59(4):e02402-20

            71. Zhang D, Yan Y, Que H, Yang T, Cheng X, Ding S, et al.. CRISPR/Cas12a-mediated interfacial cleaving of hairpin DNA reporter for electrochemical nucleic acid sensing. ACS Sens. 2020. Vol. 5(2):557–562

            72. He Q, Yu D, Bao M, Korensky G, Chen J, Shin M, et al.. High-throughput and all-solution phase African swine fever virus (ASFV) detection using CRISPR-Cas12a and fluorescence based point-of-care system. Biosens Bioelectron. 2020. Vol. 154:112068

            73. Lu S, Li F, Chen Q, Wu J, Duan J, Lei X, et al.. Rapid detection of African swine fever virus using Cas12a-based portable paper diagnostics. Cell Discov. 2020. Vol. 6:18

            74. Li SY, Cheng QX, Wang JM, Li XY, Zhang ZL, Gao S, et al.. CRISPR-Cas12a-assisted nucleic acid detection. Cell Discov. 2018. Vol. 4:20

            75. Li L, Li S, Wu N, Wu J, Wang G, Zhao G, et al.. HOLMESv2: a CRISPR-Cas12b-assisted platform for nucleic acid detection and DNA methylation quantitation. ACS Synth Biol. 2019. Vol. 8(10):2228–2237

            76. English MA, Soenksen LR, Gayet RV, de Puig H, Angenent-Mari NM, Mao AS, et al.. Programmable CRISPR-responsive smart materials. Science. 2019. Vol. 365(6455):780–785

            77. Sheng AZ, Yang JY, Tang LF, Niu LL, Cheng LF, Zeng YJ, et al.. Hydrazone chemistry-mediated CRISPR/Cas12a system for bacterial analysis. Nucleic Acids Res. 2022. Vol. 50(18):10562–10570

            78. Wu Y, Luo W, Weng Z, Guo Y, Yu H, Zhao R, et al.. A PAM-free CRISPR/Cas12a ultra-specific activation mode based on toehold-mediated strand displacement and branch migration. Nucleic Acids Res. 2022. Vol. 50(20):11727–11737

            79. Gootenberg JS, Abudayyeh OO, Kellner MJ, Joung J, Collins JJ, Zhang F. Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science. 2018. Vol. 360(6387):439–444

            80. Gootenberg JS, Abudayyeh OO, Lee JW, Essletzbichler P, Dy AJ, Joung J, et al.. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science. 2017. Vol. 356(6336):438–442

            81. Ackerman CM, Myhrvold C, Thakku SG, Freije CA, Metsky HC, Yang DK, et al.. Massively multiplexed nucleic acid detection with Cas13. Nature. 2020. Vol. 582(7811):277–282

            82. Myhrvold C, Freije CA, Gootenberg JS, Abudayyeh OO, Metsky HC, Durbin AF, et al.. Field-deployable viral diagnostics using CRISPR-Cas13. Science. 2018. Vol. 360(6387):444–448

            83. Chen W, Luo H, Zeng L, Pan Y, Parr JB, Jiang Y, et al.. A suite of PCR-LwCas13a assays for detection and genotyping of Treponema pallidum in clinical samples. Nat Commun. 2022. Vol. 13(1):4671

            84. Chen Y, Xu X, Wang J, Zhang Y, Zeng W, Liu Y, et al.. Photoactivatable CRISPR/Cas12a strategy for one-pot DETECTR molecular diagnosis. Anal Chem. 2022. Vol. 94(27):9724–9731

            85. Aman R, Marsic T, Sivakrishna Rao G, Mahas A, Ali Z, Alsanea M, et al.. iSCAN-V2: a one-pot RT-RPA-CRISPR/Cas12b assay for point-of-care SARS-CoV-2 detection. Front Bioeng Biotechnol. 2021. Vol. 9:800104

            86. Chen T, Sheng A, Hu Y, Mao D, Ning L, Zhang J. Modularization of three-dimensional gold nanoparticles/ferrocene/liposome cluster for electrochemical biosensor. Biosens Bioelectron. 2019. Vol. 124-125:115–121

            87. Hang HC, Bertozzi CR. Chemoselective approaches to glycoprotein assembly. Acc Chem Res. 2001. Vol. 34(9):727–736

            88. Khodakov DA, Khodakova AS, Linacre A, Ellis AV. Toehold-mediated nonenzymatic DNA strand displacement as a platform for DNA genotyping. J Am Chem Soc. 2013. Vol. 135(15):5612–5619

            89. Bronner CE, Baker SM, Morrison PT, Warren G, Smith LG, Lescoe MK, et al.. Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature. 1994. Vol. 368(6468):258–261

            90. Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, et al.. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006. Vol. 66(8):3992–3995

            91. Shah SP, Morin RD, Khattra J, Prentice L, Pugh T, Burleigh A, et al.. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature. 2009. Vol. 461(7265):809–813

            92. Tian B, Minero G, Fock J, Dufva M, Hansen MF. CRISPR-Cas12a based internal negative control for nonspecific products of exponential rolling circle amplification. Nucleic Acids Res. 2020. Vol. 48(5):e30

            93. Dai Y, Xu W, Somoza RA, Welter JF, Caplan AI, Liu CC. An integrated multi-function heterogeneous biochemical circuit for high-resolution electrochemistry-based genetic analysis. Angew Chem Int Ed Engl. 2020. Vol. 59(46):20545–20551

            94. Ding X, Yin K, Li Z, Lalla RV, Ballesteros E, Sfeir MM, et al.. Ultrasensitive and visual detection of SARS-CoV-2 using all-in-one dual CRISPR-Cas12a assay. Nat Commun. 2020. Vol. 11(1):4711

            95. Hillary VE, Ceasar SA. A review on the mechanism and applications of CRISPR/Cas9/Cas12/Cas13/Cas14 proteins utilized for genome engineering. Mol Biotechnol. 2023. Vol. 65(3):311–325

            96. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, et al.. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. 2015. Vol. 163(3):759–771

            97. Guido M, Tumolo MR, Verri T, Romano A, Serio F, De Giorgi M, et al.. Human bocavirus: current knowledge and future challenges. World J Gastroenterol. 2016. Vol. 22(39):8684–8697

            98. Krupovic M, Forterre P. Single-stranded DNA viruses employ a variety of mechanisms for integration into host genomes. Ann N Y Acad Sci. 2015. Vol. 1341:41–53

            99. Hu J, Zhou J, Liu R, Lv Y. Element probe based CRISPR/Cas14 bioassay for non-nucleic-acid targets. Chem Commun (Camb). 2021. Vol. 57(80):10423–10426

            100. Wu P, Ye X, Wang D, Gong F, Wei X, Xiang S, et al.. A novel CRISPR/Cas14a system integrated with 2D porphyrin metal-organic framework for microcystin-LR determination through a homogeneous competitive reaction. J Hazard Mater. 2022. Vol. 424(Pt D):127690

            101. Arizti-Sanz J, Freije CA, Stanton AC, Petros BA, Boehm CK, Siddiqui S, et al.. Streamlined inactivation, amplification, and Cas13-based detection of SARS-CoV-2. Nat Commun. 2020. Vol. 11(1):5921

            102. Kanwar JR, Mohan RR, Kanwar RK, Roy K, Bawa R. Applications of aptamers in nanodelivery systems in cancer, eye and inflammatory diseases. Nanomedicine (Lond). 2010. Vol. 5(9):1435–1445

            103. Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990. Vol. 249(4968):505–510

            104. Thiviyanathan V, Gorenstein DG. Aptamers and the next generation of diagnostic reagents. Proteomics Clin Appl. 2012. Vol. 6(11-12):563–573

            105. Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990. Vol. 346(6287):818–822

            106. Manning SD, Motiwala AS, Springman AC, Qi W, Lacher DW, Ouellette LM, et al.. Variation in virulence among clades of Escherichia coli O157:H7 associated with disease outbreaks. Proc Natl Acad Sci U S A. 2008. Vol. 105(12):4868–4873

            107. Saxena T, Kaushik P, Krishna MM. Prevalence of E. coli O157:H7 in water sources: an overview on associated diseases, outbreaks and detection methods. Diagn Microbiol Infect Dis. 2015. Vol. 82(3):249–264

            108. Chakraborty SP, Mahapatra SK, Sahu SK, Chattopadhyay S, Pramanik P, Roy S. Nitric oxide mediated staphylococcus aureus pathogenesis and protective role of nanoconjugated vancomycin. Asian Pac J Trop Biomed. 2011. Vol. 1(2):102–109

            109. Hiramatsu K, Katayama Y, Matsuo M, Sasaki T, Morimoto Y, Sekiguchi A, et al.. Multi-drug-resistant staphylococcus aureus and future chemotherapy. J Infect Chemother. 2014. Vol. 20(10):593–601

            110. Zhao YW, Wang HX, Jia GC, Li Z. Application of aptamer-based biosensor for rapid detection of pathogenic Escherichia coli . Sensors (Basel). 2018. Vol. 18(8):2518

            111. Meng-Yu FU, Li-Ping L, Yun WU, Jin W, Guo-Ping WU. Establishment of a visual and rapid detection method for enterotoxigenic Escherichia coli K88. Mod Food Sci Technol. 2016. Vol. 32(10):283–289

            112. Wu W, Zhang J, Zheng M, Zhong Y, Yang J, Zhao Y, et al.. An aptamer-based biosensor for colorimetric detection of Escherichia coli O157:H7. PLoS One. 2012. Vol. 7(11):e48999

            113. Pandit C, Alajangi HK, Singh J, Khajuria A, Sharma A, Hassan MS, et al.. Development of magnetic nanoparticle assisted aptamer-quantum dot based biosensor for the detection of Escherichia coli in water samples. Sci Total Environ. 2022. Vol. 831:154857

            114. Yuan J, Wu S, Duan N, Ma X, Xia Y, Chen J, et al.. A sensitive gold nanoparticle-based colorimetric aptasensor for Staphylococcus aureus . Talanta. 2014. Vol. 127:163–168

            115. Queirós RB, De-Los-Santos-álvarez N, Noronha JP, Sales MGF. A label-free DNA aptamer-based impedance biosensor for the detection of E. coli outer membrane proteins. Sens Actuators B Chem. 2013. Vol. 181:766–772

            116. Jiang Y, Zou S, Cao X. A simple dendrimer-aptamer based microfluidic platform for E. coli O157:H7 detection and signal intensification by rolling circle amplification. Sens Actuators B Chem. 2017. Vol. 251:976–984

            117. Lagier MJ, Scholin CA, Fell JW, Wang J, Goodwin KD. An electrochemical RNA hybridization assay for detection of the fecal indicator bacterium Escherichia coli . Mar Pollut Bull. 2005. Vol. 50(11):1251–1261

            118. Arora K, Prabhakar N, Chand S, Malhotra BD. Escherichia coli genosensor based on polyaniline. Anal Chem. 2007. Vol. 79(16):6152–6158

            119. Brosel-Oliu S, Ferreira R, Uria N, Abramova N, Gargallo R, Muñoz-Pascual F, et al.. Novel impedimetric aptasensor for label-free detection of Escherichia coli O157:H7. Sens Actuators B Chem. 2018. Vol. 255:2988–2995

            120. Ding S, Mosher C, Lee XY, Das SR, Cargill AA, Tang X, et al.. Rapid and label-free detection of interferon Gamma via an electrochemical aptasensor comprising a ternary surface monolayer on a gold interdigitated electrode array. ACS Sens. 2017. Vol. 2(2):210–217

            121. Hao N, Zhang X, Zhou Z, Hua R, Zhang Y, Liu Q, et al.. AgBr nanoparticles/3D nitrogen-doped graphene hydrogel for fabricating all-solid-state luminol-electrochemiluminescence Escherichia coli aptasensors. Biosens Bioelectron. 2017. Vol. 97:377–383

            122. Nguyen TT, Kim ER, Gu MB. A new cognate aptamer pair-based sandwich-type electrochemical biosensor for sensitive detection of Staphylococcus aureus . Biosens Bioelectron. 2022. Vol. 198:113835

            123. Bai H, Wang R, Hargis B, Lu H, Li Y. A SPR aptasensor for detection of avian influenza virus H5N1. Sensors (Basel). 2012. Vol. 12(9):12506–12518

            124. Nguyen VT, Seo HB, Kim BC, Kim SK, Song CS, Gu MB. Highly sensitive sandwich-type SPR based detection of whole H5Nx viruses using a pair of aptamers. Biosens Bioelectron. 2016. Vol. 86:293–300

            125. Tombelli S, Minunni M, Luzi E, Mascini M. Aptamer-based biosensors for the detection of HIV-1 tat protein. Bioelectrochemistry. 2005. Vol. 67(2):135–141

            126. Weerathunge P, Ramanathan R, Torok VA, Hodgson K, Xu Y, Goodacre R, et al.. Ultrasensitive colorimetric detection of murine norovirus using nanozyme aptasensor. Anal Chem. 2019. Vol. 91(5):3270–3276

            127. Chen C, Zou Z, Chen L, Ji X, He Z. Functionalized magnetic microparticle-based colorimetric platform for influenza A virus detection. Nanotechnology. 2016. Vol. 27(43):435102

            128. Shiratori I, Akitomi J, Boltz DA, Horii K, Furuichi M, Waga I. Selection of DNA aptamers that bind to influenza A viruses with high affinity and broad subtype specificity. Biochem Biophys Res Commun. 2014. Vol. 443(1):37–41

            129. Kim SH, Lee J, Lee BH, Song CS, Gu MB. Specific detection of avian influenza H5N2 whole virus particles on lateral flow strips using a pair of sandwich-type aptamers. Biosens Bioelectron. 2019. Vol. 134:123–129

            130. Liu M, Zhao H, Chen S, Yu H, Quan X. Interface engineering catalytic graphene for smart colorimetric biosensing. ACS Nano. 2012. Vol. 6(4):3142–3151

            131. Park JH, Jee MH, Kwon OS, Keum SJ, Jang SK. Infectivity of hepatitis C virus correlates with the amount of envelope protein E2: development of a new aptamer-based assay system suitable for measuring the infectious titer of HCV. Virology. 2013. Vol. 439(1):13–22

            132. Escudero-Abarca BI, Suh SH, Moore MD, Dwivedi HP, Jaykus LA. Selection, characterization and application of nucleic acid aptamers for the capture and detection of human norovirus strains. PLoS One. 2014. Vol. 9(9):e106805

            133. Lee KH, Zeng H. Aptamer-based ELISA assay for highly specific and sensitive detection of Zika NS1 protein. Anal Chem. 2017. Vol. 89(23):12743–12748

            134. Le TT, Chang P, Benton DJ, Mccauley JW, Iqbal M, Cass A. Dual recognition element lateral flow assay toward multiplex strain specific influenza virus detection. Anal Chem. 2017. Vol. 89(12):6781–6786

            135. Yeom G, Kang J, Jang H, Nam HY, Kim MG, Park CJ. Development of DNA aptamers against the nucleocapsid protein of severe fever with thrombocytopenia syndrome virus for diagnostic application: catalytic signal amplification using replication protein A-conjugated liposomes. Anal Chem. 2019. Vol. 91(21):13772–13779

            136. Chen Z, Wu Q, Chen J, Ni X, Dai J. A DNA aptamer based method for detection of SARS-CoV-2 nucleocapsid protein. Virol Sin. 2020. Vol. 35(3):351–354

            137. Zhang L, Fang X, Liu X, Ou H, Zhang H, Wang J, et al.. Discovery of sandwich type COVID-19 nucleocapsid protein DNA aptamers. Chem Commun (Camb). 2020. Vol. 56(70):10235–10238

            138. Suh SK, Song S, Oh HB, Hwang SH, Hah SS. Aptamer-based competitive binding assay for one-step quantitation of hepatitis B surface antigen. Analyst. 2014. Vol. 139(17):4310–4314

            139. Pramanik A, Gao Y, Patibandla S, Mitra D, Mccandless MG, Fassero LA, et al.. Aptamer conjugated gold nanostar-based distance-dependent nanoparticle surface energy transfer spectroscopy for ultrasensitive detection and inactivation of corona virus. J Phys Chem Lett. 2021. Vol. 12(8):2166–2171

            140. Szakacs Z, Meszaros T, de Jonge MI, Gyurcsanyi RE. Selective counting and sizing of single virus particles using fluorescent aptamer-based nanoparticle tracking analysis. Nanoscale. 2018. Vol. 10(29):13942–13948

            141. Zhang J, Tian J, He Y, Chen S, Jiang Y, Zhao Y, et al.. Protein-binding aptamer assisted signal amplification for the detection of influenza A (H1N1) DNA sequences based on quantum dot fluorescence polarization analysis. Analyst. 2013. Vol. 138(17):4722–4727

            142. Ahn DG, Jeon IJ, Kim JD, Song MS, Han SR, Lee SW, et al.. RNA aptamer-based sensitive detection of SARS coronavirus nucleocapsid protein. Analyst. 2009. Vol. 134(9):1896–1901

            143. Xi Z, Huang R, Li Z, He N, Wang T, Su E, et al.. Selection of HBsAg-specific DNA aptamers based on carboxylated magnetic nanoparticles and their application in the rapid and simple detection of hepatitis B virus infection. ACS Appl Mater Interfaces. 2015. Vol. 7(21):11215–11223

            144. Liu J, Qin Q, Zhang X, Li C, Yu Y, Huang X, et al.. Development of a novel lateral flow biosensor combined with aptamer-based isolation: application for rapid detection of grouper nervous necrosis virus. Front Microbiol. 2020. Vol. 11:886

            145. Roh C, Kim SE, Jo SK. Label free inhibitor screening of hepatitis C virus (HCV) NS5B viral protein using RNA oligonucleotide. Sensors (Basel). 2011. Vol. 11(7):6685–6696

            146. Park JA, Kim J, Kim SM, Sohn H, Park C, Kim TH, et al.. Fabrication of electrochemical influenza virus (H1N1) biosensor composed of multifunctional DNA four-way junction and molybdenum disulfide hybrid material. Materials (Basel). 2021. Vol. 14(2):343

            147. Rahmati Z, Roushani M, Hosseini H. Three-dimensional NiCo2O4 nanowires encapsulated in nitrogen-doped carbon networks as a high-performance aptamer stabilizer for impedimetric ultrasensitive detection of hepatitis C virus core antigen. Surf Interfaces. 2021. Vol. 22:100813

            148. Ghanbari K, Roushani M, Azadbakht A. Ultra-sensitive aptasensor based on a GQD nanocomposite for detection of hepatitis C virus core antigen. Anal Biochem. 2017. Vol. 534:64–69

            149. Fu Y, Callaway Z, Lum J, Wang R, Lin J, Li Y. Exploiting enzyme catalysis in ultra-low ion strength media for impedance biosensing of avian influenza virus using a bare interdigitated electrode. Anal Chem. 2014. Vol. 86(4):1965–1971

            150. Wang R, Xu L, Li Y. Bio-nanogate controlled enzymatic reaction for virus sensing. Biosens Bioelectron. 2015. Vol. 67:400–407

            151. Rahim RA, Tanabe K, Ibori S, Wang X, Kawarada H. Effects of diamond-FET-based RNA aptamer sensing for detection of real sample of HIV-1 tat protein. Biosens Bioelectron. 2013. Vol. 40(1):277–282

            152. Seo G, Lee G, Kim MJ, Baek SH, Choi M, Ku KB, et al.. Rapid detection of COVID-19 causative virus (SARS-CoV-2) in human nasopharyngeal swab specimens using field-effect transistor-based biosensor. ACS Nano. 2020. Vol. 14(4):5135–5142

            153. Giamberardino A, Labib M, Hassan EM, Tetro JA, Springthorpe S, Sattar SA, et al.. Ultrasensitive norovirus detection using DNA aptasensor technology. PLoS One. 2013. Vol. 8(11):e79087

            154. Minunni M, Tombelli S, Gullotto A, Luzi E, Mascini M. Development of biosensors with aptamers as bio-recognition element: the case of HIV-1 tat protein. Biosens Bioelectron. 2004. Vol. 20(6):1149–1156

            155. Wang R, Li Y. Hydrogel based QCM aptasensor for detection of avian influenza virus. Biosens Bioelectron. 2013. Vol. 42:148–155

            156. Pleshakova TO, Kaysheva AL, Bayzyanova JM, Anashkina AS, Uchaikin VF, Ziborov VS, et al.. The detection of hepatitis c virus core antigen using afm chips with immobolized aptamers. J Virol Methods. 2018. Vol. 251:99–105

            157. Roca C, Avalos-Padilla Y, Prieto-Simon B, Iglesias V, Ramirez M, Imperial S, et al.. Selection of an aptamer against the enzyme 1-deoxy-D-xylulose-5-phosphate reductoisomerase from Plasmodium falciparum . Pharmaceutics. 2022. Vol. 14(11):2515

            158. Cui W, Shen X, Wang C, Bibi A, Cudjoe O, Zhao L, et al.. Direct enzyme-linked aptamer assay (DELAA) for diagnosis of Toxoplasmosis by detection of SAG1 protein in mice and humans. Acta Trop. 2022. Vol. 226:106255

            159. Homann M, Goringer HU. Combinatorial selection of high affinity RNA ligands to live African trypanosomes. Nucleic Acids Res. 1999. Vol. 27(9):2006–2014

            160. Cheung YW, Kwok J, Law AW, Watt RM, Kotaka M, Tanner JA. Structural basis for discriminatory recognition of Plasmodium lactate dehydrogenase by a DNA aptamer. Proc Natl Acad Sci U S A. 2013. Vol. 110(40):15967–15972

            161. Joseph DF, Nakamoto JA, Garcia RO, Penaranda K, Sanchez-Castro AE, Castillo PS, et al.. DNA aptamers for the recognition of HMGB1 from Plasmodium falciparum . PLoS One. 2019. Vol. 14(4):e0211756

            162. Oteng EK, Gu W, Mckeague M. High-efficiency enrichment enables identification of aptamers to circulating Plasmodium falciparum-infected erythrocytes. Sci Rep. 2020. Vol. 10(1):9706

            163. Acquah C, Agyei D, Obeng EM, Pan S, Tan KX, Danquah MK. Aptamers: an emerging class of bioaffinity ligands in bioactive peptide applications. Crit Rev Food Sci Nutr. 2020. Vol. 60(7):1195–1206

            164. Bu S, Liu X, Wang Z, Wei H, Yu S, Li Z, et al.. Ultrasensitive detection of pathogenic bacteria by CRISPR/Cas12a coupling with a primer exchange reaction. Sens Actuators B Chem. 2021. Vol. 347:130630

            165. Yuan G, Xia X, Zhang J, Huang J, Xie F, Li X, et al.. A novel “signal on-off-super on” sandwich-type aptamer sensor of CRISPR-Cas12a coupled voltage enrichment assay for VEGF detection. Biosens Bioelectron. 2023. Vol. 221:114424

            166. Zhou B, Yang R, Sohail M, Kong X, Zhang X, Fu N, et al.. CRISPR/Cas14 provides a promising platform in facile and versatile aptasensing with improved sensitivity. Talanta. 2023. Vol. 254:124120

            167. Lyu XY, Deng Y, Huang XY, Li ZZ, Fang GQ, Yang D, et al.. CRISPR FISHer enables high-sensitivity imaging of nonrepetitive DNA in living cells through phase separation-mediated signal amplification. Cell Res. 2022. Vol. 32(11):969–981

            168. Bravo J, Hallmark T, Naegle B, Beisel CL, Jackson RN, Taylor DW. RNA targeting unleashes indiscriminate nuclease activity of CRISPR-Cas12a2. Nature. 2023. Vol. 613(7944):582–587

            169. Dmytrenko O, Neumann GC, Hallmark T, Keiser DJ, Crowley VM, Vialetto E, et al.. Cas12a2 elicits abortive infection through RNA-triggered destruction of dsDNA. Nature. 2023. Vol. 613(7944):588–594

            Author and article information

            Contributors
            Journal
            Zoonoses
            Zoonoses
            Zoonoses
            Compuscript (Shannon, Ireland )
            2737-7466
            2737-7474
            09 May 2023
            : 3
            : 1
            : e978
            Affiliations
            [1 ]Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
            [2 ]Department of Medical Microbiology and Parasitology, School of Basic Medical Science, Fudan University, Shanghai, China
            Author notes
            *Corresponding author E-mail: xjcheng@ 123456shmu.edu.cn (XC)
            Article
            10.15212/ZOONOSES-2023-0008
            64f47072-0ac2-4f46-9959-645288cec37f
            Copyright © 2023 The Authors.

            This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY) 4.0, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

            History
            : 02 February 2023
            : 04 April 2023
            : 19 April 2023
            Page count
            Figures: 2, Tables: 5, References: 169, Pages: 17
            Funding
            Funded by: Shanghai Municipal Science and Technology Major Project
            Award ID: ZD2021CY001
            This work was funded by Shanghai Municipal Science and Technology Major Project (ZD2021CY001). Figs 1 and 2 were created with BioRender.com.
            Categories
            Review Article

            Parasitology,Animal science & Zoology,Molecular biology,Public health,Microbiology & Virology,Infectious disease & Microbiology
            aptamer,Clustered regularly interspaced short palindromic repeats-Cas,pathogen detection,diagnostic tool

            Comments

            Comment on this article