key: cord-0814495-bkmt53rx authors: Yoshimi, Kazuto; Takeshita, Kohei; Yamayoshi, Seiya; Shibumura, Satomi; Yamauchi, Yuko; Yamamoto, Masaki; Yotsuyanagi, Hiroshi; Kawaoka, Yoshihiro; Mashimo, Tomoji title: CRISPR-Cas3-based diagnostics for SARS-CoV-2 and influenza virus date: 2022-01-30 journal: iScience DOI: 10.1016/j.isci.2022.103830 sha: 7368766c4d29be72953a3f4db719c0b90b96d371 doc_id: 814495 cord_uid: bkmt53rx CRISPR-based diagnostics (CRISPR-dx), including the Cas12-based DETECTR and Cas13-based SHERLOCK Class 2 CRISPRs, have been used to detect the presence of DNA or RNA from pathogens, such as the 2009 pandemic influenza virus A (IAV) and the 2019 novel coronavirus SARS-CoV-2. Here, we describe the collateral single-stranded DNA cleavage with Class 1 type I CRISPR-Cas3 and highlight its potential for development as a Cas3-mediated rapid (within 40 min), low-cost, instrument-free detection method for SARS-CoV-2. This assay, which we call Cas3-Operated Nucleic Acid detectioN (CONAN), not only detects SARS-CoV-2 in clinical samples, but also offers specific detection of single-base-pair mutations in IAV variants. This tool allows rapid and accurate point-of-care testing for patients with suspected SARS-CoV-2 or drug-resistant IAV infections in hospitals. infections are generally not ready until the day after sample collection because the samples need to be shipped to reference laboratories for accurate diagnostic testing (Bustin and Mueller, 2005; Corman et al., 2020; R et al., 2020) . RT-qPCR assays also require expensive equipment and well-trained personnel for their operation. Alternatively, rapid antigen tests can directly detect viral components without the amplification steps needed for RT-PCR; however, the evidence for their sensitivity and diagnostic accuracy require evaluation (Green and StGeorge, 2018) . In contrast, serology antibody tests with lateral flow immunoassays can rapidly and sensitively determine the infection rate in a population, although IgG antibodies to viruses are generally only detectable 10-14 days post-infection (Kumar and Henrickson, 2012; Theel et al., 2020) . To overcome such limitations in the current diagnostic technologies, CRISPR-based diagnostics (CRISPR-dx) have been used to rapidly, robustly, and sensitively detect emerging viruses. These systems rely on type V CRISPR-Cas12, otherwise called DETECTR (DNA Endonuclease-TargEted CRISPR Trans Reporter) (Chen et al., 2018) or type VI CRISPR-Cas13, otherwise called SHERLOCK (Specific High-sensitive Enzymatic Reporter UnLOCKing) (Gootenberg et al., 2017) . Both of these Cas enzymes, but not Cas9, exhibit nonspecific endonuclease activity in trans after binding to a specific cis target via programmable CRISPR RNAs (crRNAs). By combining isothermal amplification methods (e.g., Recombinase Polymerase Amplification, RPA (Piepenburg et al., 2006) , or Loop-mediated isothermal AMPlification (LAMP) (Notomi et al., 2000) with reporting formats such as lateral flow detection with antigen-labeled reporters (Kellner et al., 2019) , DETECTR (Broughton et al., 2020; Ding et al., 2020; Lucia et al., 2020) and SHERLOCK J o u r n a l P r e -p r o o f (Ackerman et al., 2020; Metsky et al., 2020) have recently been used for rapid and highly sensitive SARS-CoV-2 detection. Furthermore, during the peer reviewing period of this manuscript, type III CRISPR-Cas systems have also been reported for SARS-CoV-2 diagnosis (Santiago-Frangos et al., 2021) . The Cas13-based strategy, PAC-MAN (Prophylactic Antiviral CRISPR in huMAN cells), has also been shown to inhibit and degrade SARS-CoV-2 viral RNA in respiratory epithelial cells (Abbott et al., 2020) . Class 2 CRISPRs (type V Cas12, type VI Cas13, and type II Cas9) only use a single Cas protein, whereas the three Class 1 CRISPRs (I, III and IV) use multiple different Cas proteins. Several studies have reported on the mechanisms involved in type I CRISPR interference (Guo et al., 2017; Hochstrasser et al., 2014; Loeff et al., 2018; Mulepati and Bailey, 2013; Redding et al., 2015; Rutkauskas et al., 2015; van Erp et al., 2018; Westra et al., 2012; Xue et al., 2017) . Type I CRISPR-Cas complexes have been reported to be seahorse-like structures containing Cas5, Cas6, multiple Cas7, Cas8 (Cse1), and two Cas11 (Cse2), and are named Cascade (Chowdhury et al., 2017; Hayes et al., 2016; Jackson et al., 2014; Mulepati et al., 2014; Sashital et al., 2012; Wiedenheft et al., 2011; Xiao et al., 2017) . Cas3, a protein containing a serine-phenylalanine (SF) 2-helicase domain and a histidine-aspartic (HD) acid-nuclease domain, degrades its target DNA in a unidirectional ATP-dependent manner (Sinkunas et al., 2011; van Erp et 37°C (Fig. S1 and Methods) . To investigate whether the type I CRISPR-Cas3 system carries its trans-cleavage activity on nearby non-specific ssDNAs in a similar fashion as type V Cas12 and type VI Cas13, we used the collateral cleavage assay previously reported for DETECTR (Chen et al., 2018) and SHERLOCK (Gootenberg et al., 2017) . Briefly, we assembled the EcoCas3/EcoCascade-crRNA complex, a 60-bp double-stranded DNA (dsDNA) actftor for the specific target gene, human EMX1 (hEMX1) or mouse Tyr (mTyr) (Tables S1 and S2), and a fluorophore quencher (FQ)-labeled ssDNA probe in the Mg 2+ reaction buffer (Chen et al., 2018; Gootenberg et al., 2017; Mulepati and Bailey, 2013) . After 10 min incubation at 37°C, we observed non-specific trans-ssDNA cleavage from the EcoCas3/EcoCascade after target-specific dsDNA cleavage, which recognized the protospacer adjacent motif (PAM) (AAG), but not the non-PAM (CCA) (Figure 1b) . We next performed several additional experiments to understand the biochemical activities necessary for collateral cleavage activity by the Cascade-Cas3 system. First, the longer 3-Kb dsDNA activator (hEMX1 plasmid), instead of the 60-bp short fragments, similarly mediated the trans-ssDNA cleavage by EcoCas3/EcoCascade ( Fig. S3) . Second, changes in the concentration of the FQ-ssDNA probe clearly correlated with the signal intensity (Fig. S4) , suggesting that Cas3 can cleave several copies of ssDNA per bound Cascade. Third, in ATP-free reaction buffer (-), the collateral activity of the EcoCas3 protein was similar to that of wild-type EcoCas3 and the SF2 motif III S483A/T485A (dead helicase mutant, dhCas3) mutant (Morisaka et al., 2019) in ATP (+) buffer, meaning that the helicase activity of EcoCas3 is not essential for collateral cleavage (Fig. S5) . Finally, omitting Mg 2+ and Co 2+ from the reaction buffer weakened this trans-ssDNA cleavage activity, whereas adding a divalent ion J o u r n a l P r e -p r o o f chelating agent (EDTA) abolished the activity (Fig. S6) . Therefore, we named this protocol Cas3-Operated Nucleic Acid detectioN (CONAN) (Figure 1a ). We then investigated the detection sensitivity of CONAN by diluting the hEMX1 or mTyr dsDNA activator with Cas3, Cascade and FQ-ssDNA in the reaction buffer. CONAN's limit of detection (LoD) was >1.0 × 10 10 copies for the activator (Fig. S7 ). To improve the LoD, we performed isothermal RPA (TwistAmp Basic kit, Maidenhead, UK) at 37°C for 30 min, followed by a 10-min incubation with Cascade and Cas3, thereby determining the activator's single-copy sensitivity level (~1.7 aM) ( Figure 1c ). We also achieved robust detection of the attomolar (aM) level activator by CONAN after mixing it with mouse genomic DNA (Figure 1d ). Detecting FQ-ssDNA cleavage needs a microplate reader for fluorescence intensity measurement. Instead of this laboratory instrument, a lateral flow strip can be used for instrument-free and portable diagnosis by the virus POCT (Kellner et al., 2019; Lucia et al., 2020) . In principle, abundant reporter accumulates anti-FITC antibody-gold nanoparticle conjugates at the first line (negative) on the strip, while cleavage of the reporter would reduce accumulation on the first line and result in signal on the second line (positive) with <2 min of flow (Fig. S8 ). Using this lateral flow strip we performed a one-pot assay with CONAN-RPA, thereby detecting a single copy level of the target dsDNA within 1 h ( Figure 1d ). We next examined whether the CONAN lateral flow assay would be effective for J o u r n a l P r e -p r o o f SARS-CoV-2 diagnosis (Figure 2a ), as has been recently reported for DETECTR (Broughton et al., 2020; Ding et al., 2020; Lucia et al., 2020) and for SHERLOCK (Ackerman et al., 2020; Metsky et al., 2020) . We designed primers to amplify the N (nucleoprotein) gene regions (N1 and N2) from SARS-CoV-2 (Table S3) , which overlap with the region used in the DETECTR-based assay (Broughton et al., 2020) , along with primers for the RT-qPCR assay from the United States Centers for Disease Control (US CDC) (Kimball et al., 2020) . The RT-qPCR assay successfully amplified both the N1 and N2 regions of SARS-CoV-2, with LoDs of <10 2 copies ( Figure 2b ). However, RT-RPA followed by Cas3-based CONAN or Cas12a-based DETECTR in the one-step 37°C 30 min incubation (Table S1 ) did not effectively detect SARS-CoV-2, probably because the N1 and N2 primers we designed for RT-RPA (Table S3 ) did not match the sensitivity of the CRISPR-based assay (Fig. S9 ). Designing primers for LAMP assays can be complicated, but the upside is that these assays seem to be less sensitive to inhibitors or off-target nucleic acid contamination in the samples (Francois et al., 2011; Notomi et al., 2000) . RT and LAMP at 62°C for 30 min, followed by CONAN or DETECTR for 10 min at 37°C, both specifically detected SARS-CoV-2 with crRNA-N1 and -N2 ( Figure 2c ). The LoDs for CONAN-LAMP and DETECTR-LAMP (<10 2 copies) compared favorably with the CDC's RT-qPCR assay for SARS-CoV-2 detection (Broughton et al., 2020; Corman et al., 2020; Udugama et al., 2020) . This rapid detection by CONAN-LAMP was achieved at 62°C for 30 min, with the lateral flow strip achieving a LoD of <10 2 copies for SARS-CoV-2 ( Figure 2d and Video S1). We then tested the extracted RNA samples from 10 PCR-positive COVID-19 patients and 21 PCR-negative samples from nasopharyngeal swabs, using the CONAN J o u r n a l P r e -p r o o f RT-LAMP and DETCTR RT-LAMP assays with lateral flow strip readouts ( Figure 2e and Fig. S10 ). SARS-CoV-2 was detected by the CONAN RT-LAMP assay in 9 of 10 patient swabs and detected in 1 of 21 negative swab samples (positive predictive agreement, 90%; negative predictive agreement, 95%). One negative swab from a COVID-19 patient was confirmed to be below the established LoD of <10 2 copies ( Fig. S10 ). This 94% detection rate (29/31) is comparable with that of the DETECTR RT-LAMP assay in this study ( Figure 2e ) and that previously reported for a COVID-19 POCT (Broughton et al., 2020) . Finally, we investigated whether CONAN was able to discriminate a single-base-pair mutation in the target sequences, as previously shown by Cas13 SHERLOCK (Gootenberg et al., 2018; Gootenberg et al., 2017) and by Cas12b CDetection (Teng et al., 2019) . The I38T drug-resistance mutation in influenza virus A confers reduced susceptibility to baloxavir treatment for IAV infections (Imai et al., 2020; Sato et al., 2020) . We designed a set of crRNAs for CONAN specifically targeting the I38T variant in IAV H1N1pdm09 and H3N2 strains (Table S1 ). After incubation, the CONAN assay clearly only detected wild-type (WT) H1N1pdm09 and H3N2 strains, but not I38T variants ( Figure 2f ). To further evaluate the specificity of single-base-pair discrimination by CONAN, several crRNAs were also designed to detect the neuraminidase mutations that reduce viral susceptibility to oseltamivir in the IAV H1N1pdm09 strain (I222R and H274Y) and in the IAV H3N2 strain (N294S) (Hatakeyama et al., 2007; McKimm-Breschkin, 2013) (Table S1 ). Again, the CONAN assay specifically identified all three mutations (encoding I222R, H274Y, and N294S), In this study, we reported on the discovery of Class I Cas3-mediated collateral trans cleavage activity. Our newly developed CONAN lateral flow assay, which uses this type of collateral cleavage, facilitated the rapid, robust, and sensitive detection of the novel coronavirus, SARS-CoV-2. Unlike the Class II single Cas12 and Cas13 platforms, CONAN employs multiple Cas proteins (Cas3, 5, 6, 7, 8, and 11) , which can be pre-mixed in the reaction buffer. By screening mismatches for each nucleotide in the 32-nt spacer, we found that a single mismatch in the spacer region, even within the seed region (positions 1-8), resulted in little or no effect on collateral ssDNA cleavage activity ( Fig. S11 and Table S4 ). This is supported by recently reported results showing that Cas12a has multiple nicking activities with tolerance of 4-8 mismatches within the spacer sequences (Fu et al., 2019; Murugan et al., 2020) . In contrast, CONAN shows high specificity for single-base-pair discrimination within the PAM site, supporting the applicability of CONAN-based detection assays for POCTs, even on novel emerging coronavirus mutants such as the A,B,C types (Forster et al., 2020) as well as spike protein mutations (Korber et al., 2020) . In summary, we have shown that CONAN, a Cas3-based novel in vitro nucleic acid-detection platform, is a rapid (within 30-40 min), low-cost, and instrument-free detection method for SARS-CoV-2. We also showed that this CONAN-based assay enables single-base-pair discrimination (Figure 2f , g) (Yoshimi et al., 2021) , thereby facilitating the deployment of CRISPR-dx for quick and mobile POCTs for drug-resistant IAV variants in hospitals or clinics. Although we have discovered the collateral ssDNA cleavage activity in the EcoCascade-Cas3 system, this study does not describe the mechanisms underlying how Cas3 mediates the collateral ssDNA cleavages as well as targeted double-stranded DNA cleavages. Our preprint manuscript describes several insights for these molecular mechanisms (Yoshimi et al., 2021) . Positive (red arrow) and negative (black arrow) bands for CONAN (see Fig. S8 ). e. Comparison of SARS-CoV-2 CONAN and DETECTR assays on 31 clinical samples (10 positive and 21 negative for SARS-CoV-2 by the CDC RT-qPCR assay) (see also J o u r n a l P r e -p r o o f Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Tomoji Mashimo (mashimo@ims.u-tokyo.sc.jp). All reagents and materials used in this manuscript are available upon request or prepared to be available from commercial sources. This study did not generate any unique datasets or code. Any additional information required to reanalyze the data reported in this work paper is available from the Lead Contact upon request. Vero E6 cells expressing human serine protease TMPRSS2 (VeroE6-TMPRSS2), derived from the kidney of an African green monkey, were kindly provided from and maintained at 28 °C with 130 rpm in PSFM-J1 medium (Fujifilm-wako, Osaka, Japan). Baculoviruses for EcoCas3 protein were produced using the Bac-to-Bac™ vector kit (Thermo Fisher Scientific) following to the manufacturer's protocol. Briefly, EcoCas3 cDNA were cloned into the pFastbac-1 plasmids, which were transformed to the MAX Efficiency™ DH10Bac Competent cells (Thermo Fisher Scientific) in LB medium. After the collection of the recombinant bacmid DNA by single colony screening, the bacmid DNA were transfected to the Sf9 cells by FuGENE HD transfection reagent in PSFM-J1 medium. We incubated the cells at 27°C for 72 hours and collected the supernatant as a first virus stock after the centrifuge of 3,500 rpm for 5 minutes. To increase the virus titer, 15 ml of the virus stock were added to the Sf9 cells at 0.6 × 10 6 cells/ml in 100 ml of PSFM-J1 medium. We again collected the supernatant as a baculovirus stock and used it for further experiments. Clinical nasopharyngeal and oropharyngeal swab samples from patients infected with SARS-CoV-2 were collected by IMSUT hospital (The University of Tokyo). Negative nasopharyngeal swabs were collected from healthy donors at IMSUT. RNA from the samples of patients and healthy donors was extracted as described in the NIID-approved protocol (input 140 µl; elution, 60 µl) using the Viral RNA mini kit (QIAGEN). These clinical specimens were discriminated by RT-PCR for positive or negative for SARS-CoV-2 infection prior to CRISPR-based assays. We used small sample size (10 positive and 21 negative) to validate the CONAN-based assays in this study, but more trials with a larger sample size will be needed before considering for clinical application. The information about the patients and healthy individuals such as the age and gender are not available due to ethical and privacy restriction. However, we don't anticipate the sex or age of the participants to have any influence on the results. Human samples were collected by following protocols approved by the Research Ethics Review Committee of the Institute of Medical Science, the University of Tokyo (approval number 2019-71-0201). Cascade/Cas3 and Cas12a target sites were based on the human EMX1 gene, the mouse Tyr gene, and the N gene from SARS-CoV-2 (Table S1) . A baculovirus expression system was used to purify the EcoCas3 protein, as previously shown (Yoshimi et al., 2021) . Briefly, EcoCas3 cDNA was cloned using an octa-histidine tag and a six asparagine-histidine repeat tag into pFastbac-1 plasmids. Sf9 cells were infected with baculovirus at a multiplicity of infection (MOI) of two with 2% FBS in PSFM-J1 medium at 28°C with 130 rpm for 24 h. We changed the culture temperature to 20°C with 110 rpm for 4 d. The expressed EcoCas3 protein was purified using nickel affinity resin (Ni-NTA, QIAGEN, Venlo, the Netherlands). To remove the tags, purified EcoCas3 was digested with TEV protease, and further purified by size-exclusion J o u r n a l P r e -p r o o f chromatography (Superdex 200 Increase 10/300 GL; Thermo Fisher Scientific) in 0.2 M NaCl, 10% glycerol, 1 mM DTT, and 20 mM HEPES-Na (pH 7.0). Purified EcoCas3 protein was evaluated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) (Fig. S1a) . The EcoCascade and crRNA complex were purified in accordance with previously reported methods (Hochstrasser et al., 2014; Jore et al., 2011; Yoshimi et al., 2021) . Briefly, EcoCascade/crRNA ribonucleoproteins (RNPs) were expressed in JM109 (DE3) by co-transformation with three plasmids: one plasmid encoding a hexahistidine tag and HRV3C protease recognition site in the N-terminus of Cas11 (plasmid pCDFDuet-1); one plasmid containing the EcoCascade operon and the genes encoding Cas5, Cas6, Cas7, Cas8, and Cas11 (plasmid pRSFDuet-1); and the final plasmid encoding crRNA (pACYCDuet-1) (Yoshimi et al., 2021) . The transformed bacteria were cultured in 2xYT medium at 37°C with 130 rpm. After the OD600 became 0.6 to 0.8, we added IPTG (final concentration 0.4 mM) and cultured at 26°C with 110 rpm for 16 hours. The expressed Cascade-crRNA RNPs were purified by Ni-NTA resin. After removing the hexahistidine tag using HRV3C protease, the EcoCascade/crRNA RNPs were further purified by size-exclusion chromatography in 350 mM NaCl, 1 mM DTT, and 20 mM HEPES-Na (pH 7.0) and size-evaluated by SDS-PAGE (Fig. S1b) . RNP sizes were found to be consistent with those of previous reports (Hochstrasser et al., 2014; Jore et al., 2011) . LbCas12a were purchased from Integrated DNA Technologies (IDT, Coralville, IA). Target-specific crRNAs were also purchased from IDT. For the Cascade/Cas3 activator templates, DNA fragments of hEMX1, mTyr and IAV variants (which include a target site) were designed and purchased from IDT (Table S2) . Total mouse genomic DNA from a C57BL/6 strain was used after purification (Maxwell Table S3 . Viral RNAs from SARS-CoV-2 were prepared according to the established protocol from the National Institute of Infectious Diseases in Japan (Matsuyama et al., 2020) . Viral RNAs were purified from an infected TMPRSS2-expressing VeroE6 cell line using the QIAamp Viral RNA Mini Kit (QIAGEN) according to the manufacturer's protocol. RT-qPCR was used to determine SARS-CoV-2 RNA copy numbers using Reliance One-Step Multiplex RT-qPCR Supermix and CFX Connect (Bio-Rad Laboratories, Hercules, CA) according to the manufacturer's protocols. N gene-specific primer and probe sets for RT-qPCR assays and the SARS-CoV-2 plasmid (positive control) were purchased from IDT (Table S3 ). Copy numbers of SARS-CoV-2 were determined based on the regression line (y = -3.3125x + 40.527), which was generated using the serial-diluted plasmids and the N2-primer set. To characterize the Cas3 collateral cleavage assays, DNA templates were added to 100 J o u r n a l P r e -p r o o f nM Cascade-crRNA complex, 250 nM Cas3 and 2.5 mM ATP in CRISPR-Cas3 system working buffer (60 mM KCl, 10 mM MgCl2, 50 µM CoCl2, 5 mM HEPES-KOH pH 7.5), as previously described (Yoshimi et al., 2021) . The ssDNA reporter probe (5ʹ-/5HEX/AAGGTCGGA/ZEN/GTCAACGGATTTGGTC/3IBFQ/-3ʹ) (250 nM) was added, and the probe's cleavage-related change in the fluorescence signal was measured every 30 s for 10 min under 37°C incubation. To detect DNAs, isothermal amplification and RPA were performed using the Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza Massively multiplexed nucleic acid detection using Cas13 Structure and activity of the Cas3 HD nuclease MJ0384, an effector enzyme of the CRISPR interference CRISPR-Cas12-based detection of SARS-CoV-2 Real-time reverse transcription PCR (qRT-PCR) and its potential use in clinical diagnosis Structure Reveals Mechanisms of Viral Suppressors that Intercept a CRISPR RNA-Guided Surveillance Complex Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR 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 Introducing a Spectrum of Long-Range Genomic Deletions in Human Embryonic Stem Cells Using Type I CRISPR-Cas Phylogenetic network analysis of SARS-CoV-2 genomes Target-dependent nickase activities of the CRISPR-Cas nucleases Cpf1 and Cas9 Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6 Nucleic acid detection with CRISPR-Cas13a/C2c2 Rapid Antigen Tests for Influenza: Rationale and Significance of the FDA Reclassification Cryo-EM Structures Reveal Mechanism and Inhibition of DNA Targeting by a CRISPR-Cas Surveillance Complex Structural basis for promiscuous PAM recognition in type I-E Cascade from E. coli Characterization and applications of Type I CRISPR-Cas systems Baculovirus expression systems for recombinant protein production in insect cells CasA mediates Cas3-catalyzed target degradation during CRISPR RNA-guided interference Influenza A variants with reduced susceptibility to baloxavir isolated from Japanese patients are fit and transmit through respiratory droplets Structural biology. Crystal structure of the CRISPR RNA-guided surveillance complex from Escherichia coli Structural basis for CRISPR RNA-guided DNA recognition by Cascade SHERLOCK: nucleic acid detection with CRISPR nucleases Infections in Residents of a Long-Term Care Skilled Nursing Facility Spike mutation pipeline reveals the emergence of a more transmissible form of SARS-CoV-2. bioRxiv An ultrasensitive, rapid, and portable coronavirus SARS-CoV-2 sequence detection method based on CRISPR-Cas12. bioRxiv Enhanced isolation of SARS-CoV-2 by TMPRSS2-expressing cells Influenza neuraminidase inhibitors: antiviral action and mechanisms of resistance CRISPR-based surveillance for COVID-19 using genomically-comprehensive machine learning design. bioRxiv CRISPR-Cas3 induces broad and unidirectional genome editing in human cells Structural and biochemical analysis of nuclease domain of clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 3 (Cas3) In vitro reconstitution of an Escherichia coli RNA-guided immune system reveals unidirectional, ATP-dependent degradation of DNA target Crystal structure of a CRISPR RNA-guided surveillance complex bound to a ssDNA target CRISPR-Cas12a has widespread off-target and dsDNA-nicking effects Loop-mediated isothermal amplification of DNA DNA detection using recombination proteins Detection methods for influenza A H1N1 virus with special reference to biosensors: a review Surveillance and Processing of Foreign DNA by the Escherichia coli CRISPR-Cas System Directional R-Loop Formation by the CRISPR-Cas Surveillance Complex Cascade Provides Efficient Off-Target Site Rejection Intrinsic signal amplification by type III CRISPR-Cas systems provides a sequence-specific SARS-CoV-2 diagnostic Mechanism of foreign DNA selection in a bacterial adaptive immune system Detection of variants with reduced baloxavir marboxil susceptibility after treatment of children with influenza A during the 2018/2019 influenza season NIH Image to ImageJ: 25 years of image analysis Cas3 is a single-stranded DNA nuclease and ATP-dependent helicase in the CRISPR/Cas immune system CDetection: CRISPR-Cas12b-based DNA detection with sub-attomolar sensitivity and single-base specificity The Role of Antibody Testing for SARS-CoV-2: Is There One? Diagnosing COVID-19: The Disease and Tools for Detection Conformational Dynamics of DNA Binding and Cas3 Recruitment by the CRISPR RNA-Guided Cascade Complex CRISPR immunity relies on the consecutive binding and degradation of negatively supercoiled invader DNA by Cascade and Cas3 Structures of the RNA-guided surveillance complex from a bacterial immune system. for RNA-guided DNA degradation by Cascade and Cas3 Structure Basis for Directional R-loop Formation and Substrate Handover Mechanisms in Type I CRISPR-Cas System Real-Time Observation of Target Search by the CRISPR Surveillance Complex Cascade Dynamic mechanisms of CRISPR interference by Escherichia coli CRISPR-Cas3. bioRxiv, 2021 Endogenous Type I CRISPR-Cas: From Foreign DNA Defense to Prokaryotic Engineering Cas3-Operated Nucleic Acid detecƟoN (CONAN)