Advancing CRISPR-Based Solutions for COVID-19 Diagnosis and Therapeutics
Abstract
:1. Introduction
Strains of Concern | Notable Mutations | First Detected | References |
---|---|---|---|
Omicron (BA.2.86) | I332V, D339H, R403K, V445H, G446S, N450D, L452W, N481K, 483del, E484K, F486P | Botswana and South Africa (Nov 2021) | [10,11,12,13,14] |
Omicron (KP.3) | Q493E, F456L | Botswana and South Africa (Nov 2021) | [15] |
Omicron (JN.1) | L455S | Botswana and South Africa (Nov 2021) | [16] |
2. Importance of Rapid and Accurate Detection Methods
3. Introduction to CRISPR Technology
4. Molecular Basis of CRISPR-Based Diagnostics (CRISPR-Dx)
- (a)
- Target Binding: The specificity of CRISPR-Dx stems from the targeted binding of Cas proteins to specific nucleic acid sequences facilitated by a guide RNA (gRNA). The gRNA is engineered to complement the target DNA or RNA sequence, directing the Cas protein to bind exclusively to the intended sequence (Figure 4). Various Cas proteins, such as Cas9 for DNA and Cas12 and Cas13 for both DNA and RNA, are used depending on the nucleic acid targeted [42,43,44]. This precise interaction between the Cas–gRNA complex and the target sequence minimizes off-target effects, enhancing the accuracy and reliability of the diagnostics. Such targeted mechanisms are crucial for effective detection and analysis in various diagnostic applications [45,46,47].
- (b)
- Signal Amplification: Signal amplification in CRISPR-Dx is crucial for enhancing sensitivity and ensuring reliable detection. Techniques such as PCR and loop-mediated isothermal amplification (LAMP) are employed to increase the quantity of target nucleic acids, making more sequences available for detection. Additionally, certain Cas proteins like Cas12 and Cas13 exhibit collateral cleavage activity [48,49]. This activity involves cleaving adjacent reporter molecules upon binding to their specific target sequence, thus generating a detectable signal. For example, when Cas12 binds to its target DNA, it also cleaves nearby single-stranded DNA (ssDNA) reporters, producing a fluorescent signal [50]. These amplification strategies significantly enhance the sensitivity and reliability of CRISPR-Dx, making it an effective tool for various diagnostic applications [48,49,51,52,53].
- (c)
- Readout and Detection: The final step in CRISPR-Dx involves the detection of the amplified signal with various methods tailored to specific applications. Each method offers unique advantages in terms of sensitivity, speed, and practicality, making them suitable for different diagnostic settings (Figure 4).
5. Applications for Molecular Detection via CRISPR-Dx
- (a)
- Rapid Detection of Pathogens: CRISPR-Dx platforms are highly effective in the rapid detection of viral pathogens, employing innovative technologies such as SHERLOCK (Specific High-sensitivity Enzymatic Reporter unLOCKing) and DETECTR (DNA Endonuclease Targeted CRISPR Trans Reporter) [61,62]. In these systems, CRISPR-associated proteins (like Cas13 for SHERLOCK and Cas12 for DETECTR) are designed to recognize specific nucleic acid sequences of pathogens (Table 2). SHERLOCK was developed by the Broad Institute and Feng Zhang’s team in collaboration with Sherlock Biosciences and DETECTR was developed by Mammoth Biosciences [50,63]. Upon binding to their target RNA or DNA, these Cas proteins activate a collateral cleavage activity that can be harnessed to cleave reporter molecules, leading to a detectable signal [64]. This mechanism was crucial during the COVID-19 pandemic for the rapid detection of SARS-CoV-2, providing accurate results essential for controlling the virus’s spread [65]. Additionally, CRISPR-Dx is adept at identifying bacterial and fungal pathogens, leveraging the same Cas protein mechanisms to diagnose a broad spectrum of infections and improve patient outcome [66,67,68].
- (b)
- Identification of Genetic Mutations: CRISPR-Dx serves as a powerful tool for detecting genetic mutations, including single nucleotide polymorphisms (SNPs) and other variations linked to genetic diseases like cancer [72]. This application typically involves engineering gRNAs that are complementary to the target mutation sequences. The Cas9 protein, guided by these gRNAs, creates double-strand breaks at specific genomic locations, allowing for the detection of these mutations either by amplifying the surrounding DNA and sequencing it or by utilizing methods such as CRISPR-based amplification [72,73]. This enables early diagnosis and supports the development of personalized treatment strategies, which are particularly impactful in hereditary diseases and oncology, where early intervention can significantly alter patient outcomes [74].
- (c)
- Environmental and Food Safety Monitoring: CRISPR-Dx is also utilized in monitoring environmental samples and food products for pathogen contamination, which is vital for public health. This involves customizing CRISPR systems to target specific sequences associated with harmful microorganisms [75]. For instance, by employing Cas proteins in a detection format similar to that used in pathogen detection, researchers can identify contamination in food products and environmental samples rapidly [76]. The specificity of CRISPR, combined with its ability to work with various sample types, enables the reliable identification of pathogens, thus helping to prevent foodborne illnesses and mitigate environmental hazards. This enhances overall safety and health standards [77,78].
6. Advantages over Traditional Methods
- (a)
- Speed: CRISPR-Dx is notably faster than traditional methods, streamlining the diagnostic process. Unlike qRT-PCR, which requires multiple steps including sample preparation, nucleic acid extraction, amplification, and analysis, CRISPR-Dx often demands less extensive sample preparation [41]. The integration of Cas proteins with gRNAs enables the direct detection of target sequences from crude samples, reducing the need for time-consuming nucleic acid extraction and purification. This approach simplifies sample preparation, making it more efficient for rapid diagnostics and point-of-care testing [79]. Moreover, many CRISPR-Dx platforms use isothermal amplification techniques like loop-mediated isothermal amplification (LAMP), which rapidly amplify nucleic acids at a constant temperature, eliminating the need for the thermal cycling required in PCR. The readout phase, using techniques such as fluorescence or lateral flow assays, typically completes within minutes, reducing the overall diagnostic time and facilitating quicker clinical and field decision making [76,80].
- (b)
- Sensitivity and specificity: CRISPR-Dx is highly sensitive and specific, which are crucial attributes for accurate molecular detection. The collateral cleavage activity of Cas12 and Cas13 proteins amplifies the signal from low-abundance targets, enhancing sensitivity to levels often exceeding those of PCR [45]. Specificity is driven by the gRNA, which directs the Cas proteins to their target sequences with high precision. This specific targeting reduces the likelihood of false positives unlike PCR, where non-specific amplification can occur [45,81,82].
- (c)
- Accessibility: CRISPR-Dx platforms are user-friendly and accessible, making them suitable for a broad range of users, including those in resource-limited settings. Simple protocols, pre-designed kits, and the development of portable devices such as lateral flow assays and microfluidic platforms facilitate on-site testing without specialized laboratory equipment [83]. This is invaluable for point-of-care (POC) and field testing, where rapid and accurate diagnostics are essential. Additionally, the streamlined workflow and reduced reliance on complex equipment make CRISPR-Dx more cost-effective compared to traditional PCR methods, promoting wider adoption, especially in areas with limited access to advanced diagnostic tools [84].
7. Challenges and Limitations of CRISPR-Dx
- (a)
- Technical Challenges: Although the CRISPR systems are highly specific, there is still a risk of off-target binding, which can lead to false-positive results. Improving the design of gRNAs and optimizing the conditions for CRISPR activity is essential to minimize these off-target effects [41,85]. While CRISPR-Dx can work with crude samples, the presence of inhibitors and complex sample matrices can affect the accuracy and efficiency of detection. Developing robust sample preparation methods that can efficiently isolate and purify nucleic acids without extensive processing is critical [41]. However, a critical limitation is the high level of technical expertise required to operate CRISPR-based diagnostics compared to other methods. CRISPR technology demands advanced technical skills, which restricts its accessibility and usability in many diagnostic and therapeutic settings. As a result, its application may be limited not only by the complexity of the technology itself but also by the need for highly trained personnel, which could hinder widespread use in clinical and field settings.
- (b)
- Operational and Practical Challenges: Integrating CRISPR-Dx with automated high-throughput systems remains a challenge. Developing user-friendly devices that can seamlessly integrate sample processing, CRISPR-based detection, and result readout in a single platform is essential for widespread adoption [41,86]. Additionally, its real-world applicability is constrained by operational complexities. Establishing standardized protocols and guidelines for CRISPR diagnostics is necessary to ensure consistent performance across different laboratories and applications. This includes standardizing gRNA design, reaction conditions, and readout methods. Beyond the technical and operational hurdles, the potential for widespread use in real diagnostics and therapeutics is limited by regulatory, cost, and infrastructure challenges. Gaining regulatory approval requires rigorous clinical validation, which can be time consuming and complex, further delaying its entry into the market [41].
8. Key Targets Against COVID-19
- (a)
- Targets within the SARS-CoV-2 Genome
- (b)
- Targeting host factors
9. CRISPR-Based Diagnostics Against COVID-19
- (a)
- Antigen–antibody serum reactions are employed in COVID-19 diagnosis, where they detect either viral proteins (antigens) or the body’s immune response (antibodies). These tests are simple, rapid, and suitable for point-of-care use, though they tend to have lower sensitivity, particularly in the early stages of infection. On the other hand, PCR (Polymerase Chain Reaction) is the gold standard for COVID-19 detection [95], amplifying viral RNA through reverse transcription followed by DNA amplification. PCR offers excellent sensitivity and specificity, although it requires specialized equipment and trained personnel, making it less accessible in resource-limited settings.
- (b)
- The DNA Endonuclease Targeted CRISPR Trans Reporter (DETECTR) is a diagnostic technology that uses the CRISPR-Cas12 system to detect specific DNA sequences, particularly those of pathogens such as viruses (Table 3, Figure 5). DETECTR works by combining the CRISPR-Cas12a enzyme with a guide RNA to identify and bind to a target DNA sequence [96]. Upon binding, Cas12a activates its collateral cleavage activity, cutting nearby reporter molecules and producing a detectable signal. This allows for rapid, highly sensitive, and specific detection of pathogens, making DETECTR a valuable tool for diagnosing infectious diseases like COVID-19 [97]. It is portable, cost-effective, and can be used in point-of-care settings, offering a practical solution for widespread diagnostic needs [96,97,98,99].
- (c)
- Specific High-sensitivity Enzymatic Reporter unLOCKing (SHERLOCK) is a powerful diagnostic platform developed for the rapid, sensitive detection of nucleic acids, including viral RNA and DNA (Table 3, Figure 5) [63,82]. Leveraging CRISPR-Cas13, SHERLOCK can identify specific genetic sequences with high precision, making it ideal for detecting pathogens like SARS-CoV-2. The system uses a unique enzymatic amplification process, enhancing its sensitivity to detect even low levels of target nucleic acids [98]. SHERLOCK is designed to be portable, cost-effective, and suitable for point-of-care testing, allowing for quick and accurate diagnostics in a variety of settings, including resource-limited environments [82,96,98,99].
- (d)
- One-Hour Low-cost Multipurpose Highly Efficient System (HOLMES) against COVID-19 is designed to quickly and affordably detect the virus (Table 3, Figure 5) [98]. It uses CRISPR-based diagnostics or LAMP technology to identify COVID-19 within one hour, ensuring high sensitivity and specificity at a low cost. Additionally, this system would be accessible in resource-limited settings, offering a scalable solution for both diagnostics and treatment. HOLMES aims to reduce transmission rates and improve patient outcomes through its rapid, multipurpose approach [98,100].
- (e)
- The All-In-One Dual CRISPR-Cas12a (AIOD-CRISPR) is an innovative diagnostic tool designed for the rapid and highly sensitive detection of nucleic acids, such as viral RNA or DNA, including from pathogens like SARS-CoV-2 [101]. This system integrates two CRISPR-Cas12a complexes in a single reaction, enhancing the sensitivity and specificity of detection. It enables the simultaneous amplification and detection of target sequences, making it a powerful, efficient, and user-friendly tool for diagnosing infectious diseases. The AIOD-CRISPR system is also low-cost and suitable for use in point-of-care settings, offering a versatile and scalable solution for widespread diagnostic applications [101].
CRISPR-Cas Type | Technique | Action Mode | Advantages | Disadvantages | Detection Method | Limit of Detection | Sensitivity |
---|---|---|---|---|---|---|---|
N/A | Antigen–Antibody Serum Reactions [102,103] | Detects viral antigens or host antibodies using immunoassays (e.g., ELISA) to identify the presence of COVID-19 proteins or immune response. | Rapid results (within minutes to hours) Easy to perform Suitable for point-of-care testing | False negatives/positives are more common Dependent on viral load or immune response, may miss early or asymptomatic cases | Enzyme-linked immunosorbent assay (ELISA), lateral flow assay (LFA) | Typically, nanogram to picogram levels of antigen | Lower sensitivity, especially in early stages of infection |
N/A | PCR (Polymerase Chain Reaction) [102,103] | Amplifies viral RNA through reverse transcription followed by DNA amplification, enabling detection of viral genetic material. | Considered the gold standard for COVID-19 detection Detects early infection | Requires laboratory equipment and trained personnel Time-consuming (hours) Expensive Sample preparation needed | Fluorescence (via qPCR), amplification of target DNA/RNA | ~1–10 copies of RNA per reaction (~10 femtomolar) | High sensitivity |
Cas12a | DETECTR [62] | Utilizes CRISPR-Cas12a to bind to target DNA. Cas12a cleaves nearby reporter molecules upon activation, generating a detectable signal. | Produces results within 30 min Portable and low-cost Suitable for point-of-care | Requires nucleic acid extraction and amplification Fluorescence-based detection may need specialized equipment | Lateral flow visual readout | 6.75 copies per µL of nasopharyngeal specimens | High sensitivity (100%) |
Cas13a | SHERLOCK [61,63] | Uses CRISPR-Cas13 to target viral RNA. Cas13’s collateral cleavage releases a reporter signal, indicating target presence. | Portable and suitable for point-of-care Rapid detection Can detect low levels of RNA | Requires RNA extraction and amplification Needs equipment for signal detection May not be cost-effective in some settings | Fluorescence-based and lateral flow assay | 42 RNA copies per reaction of nasopharyngeal swab | Highly sensitive (100%) |
Cas12a | AOID-CRISPR [104] | Combines two CRISPR-Cas12a complexes in a single reaction for simultaneous amplification and detection of target DNA or RNA sequences. | Combines amplification and detection in a single reaction Low cost Minimal equipment needed for detection | Sample preparation steps still required Dual CRISPR system adds complexity Less widely validated in the field compared to SHERLOCK and DETECTR | Fluorescence-based assay | 4.6 copies per µL | Ultrasensitive (100%) |
Cas12a | HOLMES [105] | Uses CRISPR-Cas12 or LAMP technology for nucleic acid amplification and detection. Cas12 cleaves a reporter molecule to signal target presence within one hour. | Rapid detection within one hour Low-cost Suitable for resource-limited settings Scalable for point-of-care use | Requires amplification prior to detection Limited to specific viral RNA/DNA sequences Not as widely implemented as PCR-based methods | Fluorescence-based assay | 10aM | 100% sensitive |
10. CRISPR-Based Therapeutics Against COVID-19
- (a)
- CRISPR-Cas13d nanotherapy is an innovative therapeutic approach designed to combat severe COVID-19 by targeting host proteases like Ctsl, which are crucial for viral entry into cells. By selectively delivering Cas13d, a smaller and more versatile CRISPR enzyme, to the lungs, this therapy specifically targets and knocks down the expression of Ctsl, hindering the virus’s ability to infect cells [107]. In animal models, lung-selective delivery of Cas13d targeting Ctsl has shown promising results in significantly reducing viral load and alleviating inflammation, making it a treatment option for severe cases of COVID-19 [106,108]. This strategy offers a targeted and precise method to mitigate the effects of the virus, reducing the severity of the disease [106,107,108].
- (b)
- Prophylactic Antiviral CRISPR in huMAN cells (PAC-MAN) is a novel therapeutic approach that utilizes the CRISPR-Cas13 system to target and degrade viral RNA within human cells, providing a proactive defense against viral infections like COVID-19 (Figure 6) [109]. The PAC-MAN approach involves designing gRNAs that direct the Cas13 enzyme to cleave specific regions of the SARS-CoV-2 genome, particularly targeting conserved areas of the viral RNA to ensure effectiveness against various strains. For example, gRNAs can be designed to target the N gene of the coronavirus, which is crucial for viral replication and assembly. By cleaving these regions, PAC-MAN effectively prevents the virus from replicating and spreading [109]. This approach can be used as both a prophylactic (preventative) and therapeutic measure, offering broad-spectrum protection against various coronaviruses and other RNA viruses [110]. In pre-clinical models, PAC-MAN has demonstrated significant potential by effectively reducing viral loads, making it a promising tool for combating current and future viral pandemics [108].
11. Pre-Clinical Trials of CRISPR
12. Delivery of CRISPR/Cas Components
13. Challenges and Limitations of the CRISPR/Cas
- (a)
- Safe and Effective In Vivo Delivery: The most pressing challenge is the lack of a reliable and safe method for delivering CRISPR components into human respiratory tract cells. Although AAV appears to be a feasible option due to the small size of the Cas13d enzyme, the possibility of an adaptive immune response against AAV remains a concern. Exploring alternative delivery methods, such as those discussed earlier, could provide a solution for CRISPR-based antiviral delivery in humans, potentially through a nasal spray or nebulizer system.
- (b)
- Off-Target Effects: Another significant concern is the potential for off-target effects. The specificity of crRNAs needs to be thoroughly evaluated using whole transcriptome RNA sequencing to ensure that the CRISPR system does not inadvertently target unintended sites within the genome. Reducing off-target activity is crucial for the safe and effective use of CRISPR technologies in therapeutic applications. Advanced techniques such as high-fidelity Cas9 variants (e.g., SpCas9-HF1) and gRNA design algorithms are being improved to enhance specificity and reduce these off-target impacts [120].
- (c)
- Validation in Pre-Clinical Models: The studies conducted so far need to be validated in pre-clinical animal models, such as macaques or ferrets, to assess the specificity and efficacy of CRISPR-based antiviral strategies. Pre-clinical testing is essential to demonstrate that these technologies can work effectively and safely in living organisms before they can be considered for human use.
- (d)
- Limitations of Current Studies: One of the most advanced studies on CRISPR-based antiviral strategies, known as PAC-MAN, was conducted on synthetic constructs of the virus rather than live SARS-CoV-2. While this study provided valuable insights, further research is needed to validate these findings using live viruses. Understanding the exact effects of the CRISPR/Cas13 system in real-world conditions is critical to its development as a therapeutic tool.
- (e)
- Immune Response and Evasion: The Cas9 protein, derived from bacteria, can be recognized as foreign by the human immune system, potentially leading to an immune response that diminishes the therapy’s efficacy or causes adverse effects. Researchers are investigating strategies such as transient immunosuppression, the use of Cas9 proteins from less immunogenic bacterial species, or engineering humanized Cas9 variants to mitigate immune responses [64,121].
- (f)
- Ethical and Regulatory Considerations: The potential for germline editing raises significant ethical concerns, particularly regarding the long-term effects and possible unintended consequences on future generations. Establishing comprehensive regulatory guidelines to ensure the safety, efficacy, and ethical application of CRISPR therapies is crucial. This involves rigorous clinical trials and oversight by regulatory bodies like the FDA and EMA.
- (g)
- Viral Diversity and Mutation Rates: Viruses, particularly RNA viruses like HIV and influenza, exhibit high mutation rates, which can lead to the rapid emergence of resistant variants that evade CRISPR-mediated targeting. Designing gRNAs that target conserved regions of viral genomes can help minimize the likelihood of escape mutants [121,122].
14. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Kumar, S.; Tao, Q.; Weaver, S.; Sanderford, M.; Caraballo-Ortiz, M.A.; Sharma, S.; Pond, S.L.K.; Miura, S. An Evolutionary Portrait of the Progenitor SARS-CoV-2 and Its Dominant Offshoots in COVID-19 Pandemic. Mol. Biol. Evol. 2021, 38, 3046–3059. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Lai, S.; Gao, G.F.; Shi, W. The emergence, genomic diversity and global spread of SARS-CoV-2. Nature 2021, 600, 408–418. [Google Scholar] [CrossRef] [PubMed]
- Rosenthal, S.H.; Gerasimova, A.; Ruiz-Vega, R.; Livingston, K.; Kagan, R.M.; Liu, Y.; Anderson, B.; Owen, R.; Bernstein, L.; Smolgovsky, A.; et al. Development and validation of a high throughput SARS-CoV-2 whole genome sequencing workflow in a clinical laboratory. Sci. Rep. 2022, 12, 2054. [Google Scholar] [CrossRef] [PubMed]
- Ozer, E.A.; Simons, L.M.; Adewumi, O.M.; Fowotade, A.A.; Omoruyi, E.C.; Adeniji, J.A.; Olayinka, O.A.; Dean, T.J.; Zayas, J.; Bhimalli, P.P.; et al. Multiple expansions of globally uncommon SARS-CoV-2 lineages in Nigeria. Nat. Commun. 2022, 13, 688. [Google Scholar] [CrossRef]
- Hart, W.S.; Miller, E.; Andrews, N.J.; Waight, P.; Maini, P.K.; Funk, S.; Thompson, R.N. Generation time of the alpha and delta SARS-CoV-2 variants: An epidemiological analysis. Lancet Infect. Dis. 2022, 22, 603–610. [Google Scholar] [CrossRef]
- Gu, H.; Xie, R.; Adam, D.C.; Tsui, J.L.; Chu, D.K.; Chang, L.D.J.; Cheuk, S.S.Y.; Gurung, S.; Krishnan, P.; Ng, D.Y.M.; et al. Genomic epidemiology of SARS-CoV-2 under an elimination strategy in Hong Kong. Nat. Commun. 2022, 13, 736. [Google Scholar] [CrossRef]
- Dhar, M.S.; Marwal, R.; Vs, R.; Ponnusamy, K.; Jolly, B.; Bhoyar, R.C.; Sardana, V.; Naushin, S.; Rophina, M.; Mellan, T.A.; et al. Genomic characterization and epidemiology of an emerging SARS-CoV-2 variant in Delhi, India. Science 2021, 374, 995–999. [Google Scholar] [CrossRef]
- Vohringer, H.S.; Sanderson, T.; Sinnott, M.; De Maio, N.; Nguyen, T.; Goater, R.; Schwach, F.; Harrison, I.; Hellewell, J.; Ariani, C.V.; et al. Genomic reconstruction of the SARS-CoV-2 epidemic in England. Nature 2021, 600, 506–511. [Google Scholar] [CrossRef]
- Davies, N.G.; Jarvis, C.I.; Group, C.C.-W.; Edmunds, W.J.; Jewell, N.P.; Diaz-Ordaz, K.; Keogh, R.H. Increased mortality in community-tested cases of SARS-CoV-2 lineage B.1.1.7. Nature 2021, 593, 270–274. [Google Scholar] [CrossRef]
- Karim, S.S.A.; Karim, Q.A. Omicron SARS-CoV-2 variant: A new chapter in the COVID-19 pandemic. Lancet 2021, 398, 2126–2128. [Google Scholar] [CrossRef]
- Burki, T.K. Omicron variant and booster COVID-19 vaccines. Lancet Respir. Med. 2022, 10, e17. [Google Scholar] [CrossRef] [PubMed]
- Araf, Y.; Akter, F.; Tang, Y.D.; Fatemi, R.; Parvez, M.S.A.; Zheng, C.; Hossain, M.G. Omicron variant of SARS-CoV-2: Genomics, transmissibility, and responses to current COVID-19 vaccines. J. Med. Virol. 2022, 94, 1825–1832. [Google Scholar] [CrossRef] [PubMed]
- Andrews, N.; Stowe, J.; Kirsebom, F.; Toffa, S.; Rickeard, T.; Gallagher, E.; Gower, C.; Kall, M.; Groves, N.; O’Connell, A.M.; et al. COVID-19 Vaccine Effectiveness against the Omicron (B.1.1.529) Variant. N. Engl. J. Med. 2022, 386, 1532–1546. [Google Scholar] [CrossRef]
- Andre, M.; Lau, L.S.; Pokharel, M.D.; Ramelow, J.; Owens, F.; Souchak, J.; Akkaoui, J.; Ales, E.; Brown, H.; Shil, R.; et al. From Alpha to Omicron: How Different Variants of Concern of the SARS-Coronavirus-2 Impacted the World. Biology 2023, 12, 1267. [Google Scholar] [CrossRef] [PubMed]
- Kaku, Y.; Yo, M.S.; Tolentino, J.E.; Uriu, K.; Okumura, K.; Genotype to Phenotype Japan, C.; Ito, J.; Sato, K. Virological characteristics of the SARS-CoV-2 KP.3, LB.1, and KP.2.3 variants. Lancet Infect. Dis. 2024, 24, e482–e483. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Faraone, J.N.; Hsu, C.C.; Chamblee, M.; Zheng, Y.M.; Carlin, C.; Bednash, J.S.; Horowitz, J.C.; Mallampalli, R.K.; Saif, L.J.; et al. Characteristics of JN.1-derived SARS-CoV-2 subvariants SLip, FLiRT, and KP.2 in neutralization escape, infectivity and membrane fusion. bioRxiv 2024. [Google Scholar] [CrossRef]
- Msemburi, W.; Karlinsky, A.; Knutson, V.; Aleshin-Guendel, S.; Chatterji, S.; Wakefield, J. The WHO estimates of excess mortality associated with the COVID-19 pandemic. Nature 2023, 613, 130–137. [Google Scholar] [CrossRef]
- Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef]
- Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.R.; Zhu, Y.; Li, B.; Huang, C.L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef]
- Malik, Y.S.; Kumar, N.; Sircar, S.; Kaushik, R.; Bhat, S.; Dhama, K.; Gupta, P.; Goyal, K.; Singh, M.P.; Ghoshal, U.; et al. Coronavirus Disease Pandemic (COVID-19): Challenges and a Global Perspective. Pathogens 2020, 9, 519. [Google Scholar] [CrossRef]
- Sexton, N.R.; Smith, E.C.; Blanc, H.; Vignuzzi, M.; Peersen, O.B.; Denison, M.R. Homology-Based Identification of a Mutation in the Coronavirus RNA-Dependent RNA Polymerase That Confers Resistance to Multiple Mutagens. J. Virol. 2016, 90, 7415–7428. [Google Scholar] [CrossRef] [PubMed]
- Malik, Y.S.; Sircar, S.; Bhat, S.; Ansari, M.I.; Pande, T.; Kumar, P.; Mathapati, B.; Balasubramanian, G.; Kaushik, R.; Natesan, S.; et al. How artificial intelligence may help the Covid-19 pandemic: Pitfalls and lessons for the future. Rev. Med. Virol. 2021, 31, 1–11. [Google Scholar] [CrossRef]
- Udugama, B.; Kadhiresan, P.; Kozlowski, H.N.; Malekjahani, A.; Osborne, M.; Li, V.Y.C.; Chen, H.; Mubareka, S.; Gubbay, J.B.; Chan, W.C.W. Diagnosing COVID-19: The Disease and Tools for Detection. ACS Nano 2020, 14, 3822–3835. [Google Scholar] [CrossRef]
- Corman, V.M.; Landt, O.; Kaiser, M.; Molenkamp, R.; Meijer, A.; Chu, D.K.; Bleicker, T.; Brunink, S.; Schneider, J.; Schmidt, M.L.; et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Euro Surveill. 2020, 25, 2000045. [Google Scholar] [CrossRef] [PubMed]
- Rando, H.M.; Brueffer, C.; Lordan, R.; Dattoli, A.A.; Manheim, D.; Meyer, J.G.; Mundo, A.I.; Perrin, D.; Mai, D.; Wellhausen, N.; et al. Molecular and Serologic Diagnostic Technologies for SARS-CoV-2. arXiv 2022, arXiv:2204.12598. [Google Scholar]
- Vandenberg, O.; Martiny, D.; Rochas, O.; van Belkum, A.; Kozlakidis, Z. Considerations for diagnostic COVID-19 tests. Nat. Rev. Microbiol. 2021, 19, 171–183. [Google Scholar] [CrossRef]
- Ng, T.I.; Correia, I.; Seagal, J.; DeGoey, D.A.; Schrimpf, M.R.; Hardee, D.J.; Noey, E.L.; Kati, W.M. Antiviral Drug Discovery for the Treatment of COVID-19 Infections. Viruses 2022, 14, 961. [Google Scholar] [CrossRef]
- Gupta, A.; Pradhan, A.; Maurya, V.K.; Kumar, S.; Theengh, A.; Puri, B.; Saxena, S.K. Therapeutic approaches for SARS-CoV-2 infection. Methods 2021, 195, 29–43. [Google Scholar] [CrossRef] [PubMed]
- Strzelec, M.; Detka, J.; Mieszczak, P.; Sobocinska, M.K.; Majka, M. Immunomodulation-a general review of the current state-of-the-art and new therapeutic strategies for targeting the immune system. Front. Immunol. 2023, 14, 1127704. [Google Scholar] [CrossRef]
- Yuan, Y.; Jiao, B.; Qu, L.; Yang, D.; Liu, R. The development of COVID-19 treatment. Front. Immunol. 2023, 14, 1125246. [Google Scholar] [CrossRef]
- Xu, Y.; Li, Z. CRISPR-Cas systems: Overview, innovations and applications in human disease research and gene therapy. Comput. Struct. Biotechnol. J. 2020, 18, 2401–2415. [Google Scholar] [CrossRef] [PubMed]
- Ishino, Y.; Krupovic, M.; Forterre, P. History of CRISPR-Cas from Encounter with a Mysterious Repeated Sequence to Genome Editing Technology. J. Bacteriol. 2018, 200, e00580-17. [Google Scholar] [CrossRef] [PubMed]
- Shabbir, M.A.; Hao, H.; Shabbir, M.Z.; Hussain, H.I.; Iqbal, Z.; Ahmed, S.; Sattar, A.; Iqbal, M.; Li, J.; Yuan, Z. Survival and Evolution of CRISPR-Cas System in Prokaryotes and Its Applications. Front. Immunol. 2016, 7, 375. [Google Scholar] [CrossRef] [PubMed]
- Makarova, K.S.; Wolf, Y.I.; Iranzo, J.; Shmakov, S.A.; Alkhnbashi, O.S.; Brouns, S.J.J.; Charpentier, E.; Cheng, D.; Haft, D.H.; Horvath, P.; et al. Evolutionary classification of CRISPR-Cas systems: A burst of class 2 and derived variants. Nat. Rev. Microbiol. 2020, 18, 67–83. [Google Scholar] [CrossRef]
- Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
- Deol, P.; Madhwal, A.; Sharma, G.; Kaushik, R.; Malik, Y.S. CRISPR use in diagnosis and therapy for COVID-19. Methods Microbiol. 2022, 50, 123–150. [Google Scholar] [CrossRef]
- Bhardwaj, P.; Kant, R.; Behera, S.P.; Dwivedi, G.R.; Singh, R. Next-Generation Diagnostic with CRISPR/Cas: Beyond Nucleic Acid Detection. Int. J. Mol. Sci. 2022, 23, 6052. [Google Scholar] [CrossRef]
- Curti, L.A.; Pereyra-Bonnet, F.; Repizo, G.D.; Fay, J.V.; Salvatierra, K.; Blariza, M.J.; Ibanez-Alegre, D.; Rinflerch, A.R.; Miretti, M.; Gimenez, C.A. CRISPR-based platform for carbapenemases and emerging viruses detection using Cas12a (Cpf1) effector nuclease. Emerg. Microbes Infect. 2020, 9, 1140–1148. [Google Scholar] [CrossRef]
- Shen, Y.; Hu, K.; Yuan, M.; Duan, G.; Guo, Y.; Chen, S. Progress and bioapplication of CRISPR-based one-step, quantitative and multiplexed infectious disease diagnostics. J. Appl. Microbiol. 2023, 134, lxad035. [Google Scholar] [CrossRef]
- Abudayyeh, O.O.; Gootenberg, J.S. CRISPR diagnostics. Science 2021, 372, 914–915. [Google Scholar] [CrossRef]
- Ghouneimy, A.; Mahas, A.; Marsic, T.; Aman, R.; Mahfouz, M. CRISPR-Based Diagnostics: Challenges and Potential Solutions toward Point-of-Care Applications. ACS Synth. Biol. 2023, 12, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Koonin, E.V.; Gootenberg, J.S.; Abudayyeh, O.O. Discovery of Diverse CRISPR-Cas Systems and Expansion of the Genome Engineering Toolbox. Biochemistry 2023, 62, 3465–3487. [Google Scholar] [CrossRef] [PubMed]
- Meeske, A.J.; Nakandakari-Higa, S.; Marraffini, L.A. Cas13-induced cellular dormancy prevents the rise of CRISPR-resistant bacteriophage. Nature 2019, 570, 241–245. [Google Scholar] [CrossRef]
- Swarts, D.C.; van der Oost, J.; Jinek, M. Structural Basis for Guide RNA Processing and Seed-Dependent DNA Targeting by CRISPR-Cas12a. Mol. Cell 2017, 66, 221–233.e224. [Google Scholar] [CrossRef]
- Kaminski, M.M.; Abudayyeh, O.O.; Gootenberg, J.S.; Zhang, F.; Collins, J.J. CRISPR-based diagnostics. Nat. Biomed. Eng. 2021, 5, 643–656. [Google Scholar] [CrossRef]
- Hillary, V.E.; Ceasar, S.A. A Review on the Mechanism and Applications of CRISPR/Cas9/Cas12/Cas13/Cas14 Proteins Utilized for Genome Engineering. Mol. Biotechnol. 2023, 65, 311–325. [Google Scholar] [CrossRef] [PubMed]
- Chakraborty, J.; Chaudhary, A.A.; Khan, S.U.; Rudayni, H.A.; Rahaman, S.M.; Sarkar, H. CRISPR/Cas-Based Biosensor As a New Age Detection Method for Pathogenic Bacteria. ACS Omega 2022, 7, 39562–39573. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.Y.; Du, Y.C.; Wang, D.X.; Ma, J.Y.; Tang, A.N.; Kong, D.M. Signal amplification and output of CRISPR/Cas-based biosensing systems: A review. Anal. Chim. Acta 2021, 1185, 338882. [Google Scholar] [CrossRef]
- Tang, Y.; Gao, L.; Feng, W.; Guo, C.; Yang, Q.; Li, F.; Le, X.C. The CRISPR-Cas toolbox for analytical and diagnostic assay development. Chem. Soc. Rev. 2021, 50, 11844–11869. [Google Scholar] [CrossRef]
- Chen, J.S.; Ma, E.; Harrington, L.B.; Da Costa, M.; Tian, X.; Palefsky, J.M.; Doudna, J.A. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science 2018, 360, 436–439. [Google Scholar] [CrossRef]
- Yuan, M.; Ding, R.; Chen, S.; Duan, G. Advances in Field Detection Based on CRISPR/Cas System. ACS Synth. Biol. 2021, 10, 2824–2832. [Google Scholar] [CrossRef] [PubMed]
- Mao, X.; Zhao, Y.; Jiang, J.; Du, Q.; Tu, B.; Li, J.; Wang, F. Sensitive and high-accuracy detection of Salmonella based on CRISPR/Cas12a combined with recombinase polymerase amplification. Lett. Appl. Microbiol. 2022, 75, 899–907. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Cheng, N. Sensitive and Portable Signal Readout Strategies Boost Point-of-Care CRISPR/Cas12a Biosensors. ACS Sens. 2023, 8, 3988–4007. [Google Scholar] [CrossRef]
- Huang, Z.; Liu, S.; Pei, X.; Li, S.; He, Y.; Tong, Y.; Liu, G. Fluorescence Signal-Readout of CRISPR/Cas Biosensors for Nucleic Acid Detection. Biosensors 2022, 12, 779. [Google Scholar] [CrossRef]
- Wu, J.; Mukama, O.; Wu, W.; Li, Z.; Habimana, J.D.; Zhang, Y.; Zeng, R.; Nie, C.; Zeng, L. A CRISPR/Cas12a Based Universal Lateral Flow Biosensor for the Sensitive and Specific Detection of African Swine-Fever Viruses in Whole Blood. Biosensors 2020, 10, 203. [Google Scholar] [CrossRef]
- Ali, Z.; Sanchez, E.; Tehseen, M.; Mahas, A.; Marsic, T.; Aman, R.; Sivakrishna Rao, G.; Alhamlan, F.S.; Alsanea, M.S.; Al-Qahtani, A.A.; et al. Bio-SCAN: A CRISPR/dCas9-Based Lateral Flow Assay for Rapid, Specific, and Sensitive Detection of SARS-CoV-2. ACS Synth. Biol. 2022, 11, 406–419. [Google Scholar] [CrossRef] [PubMed]
- Wachholz Junior, D.; Kubota, L.T. CRISPR-based electrochemical biosensors: An alternative for point-of-care diagnostics? Talanta 2024, 278, 126467. [Google Scholar] [CrossRef]
- Priya Swetha, P.D.; Sonia, J.; Sapna, K.; Prasad, K.S. Towards CRISPR powered electrochemical sensing for smart diagnostics. Curr. Opin. Electrochem. 2021, 30, 100829. [Google Scholar] [CrossRef]
- Vealan, K.; Joseph, N.; Alimat, S.; Karumbati, A.S.; Thilakavathy, K. Lateral flow assay: A promising rapid point-of-care testing tool for infections and non-communicable diseases. Asian Biomed. (Res. Rev. News) 2023, 17, 250–266. [Google Scholar] [CrossRef]
- Zhou, J.; Ren, X.M.; Wang, X.; Li, Z.; Xian, C.J. Recent advances and challenges of the use of the CRISPR/Cas system as a non-nucleic acid molecular diagnostic. Heliyon 2023, 9, e22767. [Google Scholar] [CrossRef]
- Rahman, M.R.; Hossain, M.A.; Mozibullah, M.; Mujib, F.A.; Afrose, A.; Shahed-Al-Mahmud, M.; Apu, M.A.I. CRISPR is a useful biological tool for detecting nucleic acid of SARS-CoV-2 in human clinical samples. Biomed. Pharmacother. 2021, 140, 111772. [Google Scholar] [CrossRef] [PubMed]
- Broughton, J.P.; Deng, X.; Yu, G.; Fasching, C.L.; Servellita, V.; Singh, J.; Miao, X.; Streithorst, J.A.; Granados, A.; Sotomayor-Gonzalez, A.; et al. CRISPR-Cas12-based detection of SARS-CoV-2. Nat. Biotechnol. 2020, 38, 870–874. [Google Scholar] [CrossRef] [PubMed]
- Kellner, M.J.; Koob, J.G.; Gootenberg, J.S.; Abudayyeh, O.O.; Zhang, F. SHERLOCK: Nucleic acid detection with CRISPR nucleases. Nat. Protoc. 2019, 14, 2986–3012, Erratum in Nat. Protoc. 2020, 15, 1311. [Google Scholar] [CrossRef]
- Zhou, L.; Simonian, A.L. CRISPR/Cas Technology: The Unique Synthetic Biology Genome-Editing Tool Shifting the Paradigm in Viral Diagnostics, Defense, and Therapeutics. Annu. Rev. Biomed. Eng. 2024, 26, 247–272. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Lei, R.; Zhang, H.; Chen, W.; Jia, Q.; Guo, X.; Zhang, Y.; Wu, P.; Wang, X. Rapid and sensitive detection of two fungal pathogens in soybeans using the recombinase polymerase amplification/CRISPR-Cas12a method for potential on-site disease diagnosis. Pest. Manag. Sci. 2024, 80, 1168–1181. [Google Scholar] [CrossRef]
- Li, M.; Qin, Z.; Yin, K.; Zheng, B. Application of CRISPR/Cas systems in the nucleic acid detection of pathogens: A review. Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi 2023, 35, 98–103. [Google Scholar] [CrossRef]
- Gopikrishnan, M.; Haryini, S.; Doss C, G.P. Emerging strategies and therapeutic innovations for combating drug resistance in Staphylococcus aureus strains: A comprehensive review. J. Basic. Microbiol. 2024, 64, e2300579. [Google Scholar] [CrossRef]
- de Puig, H.; Bosch, I.; Collins, J.J.; Gehrke, L. Point-of-Care Devices to Detect Zika and Other Emerging Viruses. Annu. Rev. Biomed. Eng. 2020, 22, 371–386. [Google Scholar] [CrossRef]
- Gootenberg, J.S.; Abudayyeh, O.O.; Lee, J.W.; Essletzbichler, P.; Dy, A.J.; Joung, J.; Verdine, V.; Donghia, N.; Daringer, N.M.; Freije, C.A.; et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science 2017, 356, 438–442. [Google Scholar] [CrossRef]
- Joung, J.; Ladha, A.; Saito, M.; Segel, M.; Bruneau, R.; Huang, M.W.; Kim, N.G.; Yu, X.; Li, J.; Walker, B.D.; et al. Point-of-care testing for COVID-19 using SHERLOCK diagnostics. medRxiv 2020. [Google Scholar] [CrossRef]
- Notomi, T.; Okayama, H.; Masubuchi, H.; Yonekawa, T.; Watanabe, K.; Amino, N.; Hase, T. Loop-mediated isothermal amplification of DNA. Nucleic Acids Res. 2000, 28, E63. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Ji, S.; Koh, H.R. CRISPR as a Diagnostic Tool. Biomolecules 2021, 11, 1162. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Bai, L.; Wang, G.; Han, J.; Wu, L.; Chen, X.; Zhang, H.; Feng, J.; Wang, Y.; Wang, R.; et al. SATCAS: A CRISPR/Cas13a-based simultaneous amplification and testing platform for one-pot RNA detection and SNPs distinguish in clinical diagnosis. Biosens. Bioelectron. 2024, 263, 116636. [Google Scholar] [CrossRef]
- Habimana, J.D.; Huang, R.; Muhoza, B.; Kalisa, Y.N.; Han, X.; Deng, W.; Li, Z. Mechanistic insights of CRISPR/Cas nucleases for programmable targeting and early-stage diagnosis: A review. Biosens. Bioelectron. 2022, 203, 114033. [Google Scholar] [CrossRef]
- Zhang, X. Development of CRISPR-Mediated Nucleic Acid Detection Technologies and Their Applications in the Livestock Industry. Genes 2022, 13, 2007. [Google Scholar] [CrossRef]
- Tanny, T.; Sallam, M.; Soda, N.; Nguyen, N.T.; Alam, M.; Shiddiky, M.J.A. CRISPR/Cas-Based Diagnostics in Agricultural Applications. J. Agric. Food Chem. 2023, 71, 11765–11788. [Google Scholar] [CrossRef]
- Liu, X.; Hussain, M.; Dai, J.; Li, Y.; Zhang, L.; Yang, J.; Ali, Z.; He, N.; Tang, Y. Programmable Biosensors Based on RNA-Guided CRISPR/Cas Endonuclease. Biol. Proced. Online 2022, 24, 2. [Google Scholar] [CrossRef]
- Kumaran, A.; Jude Serpes, N.; Gupta, T.; James, A.; Sharma, A.; Kumar, D.; Nagraik, R.; Kumar, V.; Pandey, S. Advancements in CRISPR-Based Biosensing for Next-Gen Point of Care Diagnostic Application. Biosensors 2023, 13, 202. [Google Scholar] [CrossRef] [PubMed]
- Huang, Z.; Fang, J.; Zhou, M.; Gong, Z.; Xiang, T. CRISPR-Cas13: A new technology for the rapid detection of pathogenic microorganisms. Front. Microbiol. 2022, 13, 1011399. [Google Scholar] [CrossRef]
- Hu, T.; Ke, X.; Li, W.; Lin, Y.; Liang, A.; Ou, Y.; Chen, C. CRISPR/Cas12a-Enabled Multiplex Biosensing Strategy Via an Affordable and Visual Nylon Membrane Readout. Adv. Sci. 2023, 10, e2204689. [Google Scholar] [CrossRef]
- Palaz, F.; Kalkan, A.K.; Tozluyurt, A.; Ozsoz, M. CRISPR-based tools: Alternative methods for the diagnosis of COVID-19. Clin. Biochem. 2021, 89, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharjee, G.; Gohil, N.; Lam, N.L.; Singh, V. CRISPR-based diagnostics for detection of pathogens. Prog. Mol. Biol. Transl. Sci. 2021, 181, 45–57. [Google Scholar] [CrossRef] [PubMed]
- Strohmaier-Nguyen, D.; Horn, C.; Baeumner, A.J. Innovations in one-step point-of-care testing within microfluidics and lateral flow assays for shaping the future of healthcare. Biosens. Bioelectron. 2024, 267, 116795. [Google Scholar] [CrossRef] [PubMed]
- Huang, Z.; Lyon, C.J.; Wang, J.; Lu, S.; Hu, T.Y. CRISPR Assays for Disease Diagnosis: Progress to and Barriers Remaining for Clinical Applications. Adv. Sci. 2023, 10, e2301697. [Google Scholar] [CrossRef] [PubMed]
- Guo, C.; Ma, X.; Gao, F.; Guo, Y. Off-target effects in CRISPR/Cas9 gene editing. Front. Bioeng. Biotechnol. 2023, 11, 1143157. [Google Scholar] [CrossRef] [PubMed]
- Nafian, F.; Nafian, S.; Kamali Doust Azad, B.; Hashemi, M. CRISPR-Based Diagnostics and Microfluidics for COVID-19 Point-of-Care Testing: A Review of Main Applications. Mol. Biotechnol. 2023, 65, 497–508. [Google Scholar] [CrossRef]
- Wang, L.; Zhou, J.; Wang, Q.; Wang, Y.; Kang, C. Rapid design and development of CRISPR-Cas13a targeting SARS-CoV-2 spike protein. Theranostics 2021, 11, 649–664. [Google Scholar] [CrossRef]
- Yang, Z.; Li, J.; Li, J.; Zheng, H.; Li, H.; Lai, Q.; Chen, Y.; Qin, L.; Zuo, Y.; Guo, L.; et al. Engagement of the G3BP2-TRIM25 Interaction by Nucleocapsid Protein Suppresses the Type I Interferon Response in SARS-CoV-2-Infected Cells. Vaccines 2022, 10, 2042. [Google Scholar] [CrossRef]
- Oh, S.J.; Shin, O.S. SARS-CoV-2 Nucleocapsid Protein Targets RIG-I-Like Receptor Pathways to Inhibit the Induction of Interferon Response. Cells 2021, 10, 530. [Google Scholar] [CrossRef]
- Tofaute, M.J.; Weller, B.; Grass, C.; Halder, H.; Dohai, B.; Falter-Braun, P.; Krappmann, D. SARS-CoV-2 NSP14 MTase activity is critical for inducing canonical NF-kappaB activation. Biosci. Rep. 2024, 44, BSR20231418. [Google Scholar] [CrossRef]
- Shen, Z.; Ratia, K.; Cooper, L.; Kong, D.; Lee, H.; Kwon, Y.; Li, Y.; Alqarni, S.; Huang, F.; Dubrovskyi, O.; et al. Design of SARS-CoV-2 PLpro Inhibitors for COVID-19 Antiviral Therapy Leveraging Binding Cooperativity. J. Med. Chem. 2022, 65, 2940–2955. [Google Scholar] [CrossRef] [PubMed]
- Singh, L.; Bajaj, S.; Gadewar, M.; Verma, N.; Ansari, M.N.; Saeedan, A.S.; Kaithwas, G.; Singh, M. Modulation of Host Immune Response Is an Alternative Strategy to Combat SARS-CoV-2 Pathogenesis. Front. Immunol. 2021, 12, 660632. [Google Scholar] [CrossRef] [PubMed]
- Emmi, A.; Tushevski, A.; Sinigaglia, A.; Barbon, S.; Sandre, M.; Stocco, E.; Macchi, V.; Antonini, A.; Barzon, L.; Porzionato, A.; et al. ACE2 Receptor and TMPRSS2 Protein Expression Patterns in the Human Brainstem Reveal Anatomical Regions Potentially Vulnerable to SARS-CoV-2 Infection. ACS Chem. Neurosci. 2023, 14, 2089–2097. [Google Scholar] [CrossRef]
- Kandhaya-Pillai, R.; Yang, X.; Tchkonia, T.; Martin, G.M.; Kirkland, J.L.; Oshima, J. TNF-alpha/IFN-gamma synergy amplifies senescence-associated inflammation and SARS-CoV-2 receptor expression via hyper-activated JAK/STAT1. Aging Cell 2022, 21, e13646. [Google Scholar] [CrossRef]
- Artika, I.M.; Dewi, Y.P.; Nainggolan, I.M.; Siregar, J.E.; Antonjaya, U. Real-Time Polymerase Chain Reaction: Current Techniques, Applications, and Role in COVID-19 Diagnosis. Genes. 2022, 13, 2387. [Google Scholar] [CrossRef]
- Vatankhah, M.; Azizi, A.; Sanajouyan Langeroudi, A.; Ataei Azimi, S.; Khorsand, I.; Kerachian, M.A.; Motaei, J. CRISPR-based biosensing systems: A way to rapidly diagnose COVID-19. Crit. Rev. Clin. Lab. Sci. 2021, 58, 225–241. [Google Scholar] [CrossRef]
- Kumar, P.; Malik, Y.S.; Ganesh, B.; Rahangdale, S.; Saurabh, S.; Natesan, S.; Srivastava, A.; Sharun, K.; Yatoo, M.I.; Tiwari, R.; et al. CRISPR-Cas System: An Approach With Potentials for COVID-19 Diagnosis and Therapeutics. Front. Cell Infect. Microbiol. 2020, 10, 576875. [Google Scholar] [CrossRef]
- Kostyusheva, A.; Brezgin, S.; Babin, Y.; Vasilyeva, I.; Glebe, D.; Kostyushev, D.; Chulanov, V. CRISPR-Cas systems for diagnosing infectious diseases. Methods 2022, 203, 431–446. [Google Scholar] [CrossRef] [PubMed]
- Fapohunda, F.O.; Qiao, S.; Pan, Y.; Wang, H.; Liu, Y.; Chen, Q.; Lu, P. CRISPR Cas system: A strategic approach in detection of nucleic acids. Microbiol. Res. 2022, 259, 127000. [Google Scholar] [CrossRef]
- Li, L.; Li, S.; Wu, N.; Wu, J.; Wang, G.; Zhao, G.; Wang, J. HOLMESv2: A CRISPR-Cas12b-Assisted Platform for Nucleic Acid Detection and DNA Methylation Quantitation. ACS Synth. Biol. 2019, 8, 2228–2237. [Google Scholar] [CrossRef]
- Ding, X.; Yin, K.; Li, Z.; Lalla, R.V.; Ballesteros, E.; Sfeir, M.M.; Liu, C. Ultrasensitive and visual detection of SARS-CoV-2 using all-in-one dual CRISPR-Cas12a assay. Nat. Commun. 2020, 11, 4711. [Google Scholar] [CrossRef] [PubMed]
- Rong, G.; Zheng, Y.; Li, X.; Guo, M.; Su, Y.; Bian, S.; Dang, B.; Chen, Y.; Zhang, Y.; Shen, L.; et al. A high-throughput fully automatic biosensing platform for efficient COVID-19 detection. Biosens. Bioelectron. 2023, 220, 114861. [Google Scholar] [CrossRef] [PubMed]
- Jayamohan, H.; Lambert, C.J.; Sant, H.J.; Jafek, A.; Patel, D.; Feng, H.; Beeman, M.; Mahmood, T.; Nze, U.; Gale, B.K. SARS-CoV-2 pandemic: A review of molecular diagnostic tools including sample collection and commercial response with associated advantages and limitations. Anal. Bioanal. Chem. 2021, 413, 49–71. [Google Scholar] [CrossRef] [PubMed]
- 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 2020. [Google Scholar] [CrossRef]
- Li, S.Y.; Cheng, Q.X.; Wang, J.M.; Li, X.Y.; Zhang, Z.L.; Gao, S.; Cao, R.B.; Zhao, G.P.; Wang, J. CRISPR-Cas12a-assisted nucleic acid detection. Cell Discov. 2018, 4, 20. [Google Scholar] [CrossRef]
- Xue, Y.; Chen, Z.; Zhang, W.; Zhang, J. Engineering CRISPR/Cas13 System against RNA Viruses: From Diagnostics to Therapeutics. Bioengineering 2022, 9, 291. [Google Scholar] [CrossRef]
- Zhou, Q.; Chen, Y.; Wang, R.; Jia, F.; He, F.; Yuan, F. Advances of CRISPR-Cas13 system in COVID-19 diagnosis and treatment. Genes. Dis. 2022, 10, 2414–2424. [Google Scholar] [CrossRef]
- Lin, X.; Liu, Y.; Chemparathy, A.; Pande, T.; La Russa, M.; Qi, L.S. A comprehensive analysis and resource to use CRISPR-Cas13 for broad-spectrum targeting of RNA viruses. Cell Rep. Med. 2021, 2, 100245. [Google Scholar] [CrossRef]
- Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R.; et al. Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza. Cell 2020, 181, 865–876.e812. [Google Scholar] [CrossRef]
- Dash, P.; Mohapatra, S.; Ghosh, S.; Nayak, B. A Scoping Insight on Potential Prophylactics, Vaccines and Therapeutic Weaponry for the Ongoing Novel Coronavirus (COVID-19) Pandemic- A Comprehensive Review. Front. Pharmacol. 2020, 11, 590154. [Google Scholar] [CrossRef]
- Lee, C.Y.; Lowen, A.C. Animal models for SARS-CoV-2. Curr. Opin. Virol. 2021, 48, 73–81. [Google Scholar] [CrossRef] [PubMed]
- Ni, W.; Yang, X.; Yang, D.; Bao, J.; Li, R.; Xiao, Y.; Hou, C.; Wang, H.; Liu, J.; Yang, D.; et al. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit. Care 2020, 24, 422. [Google Scholar] [CrossRef] [PubMed]
- Lutz, C.; Maher, L.; Lee, C.; Kang, W. COVID-19 preclinical models: Human angiotensin-converting enzyme 2 transgenic mice. Hum. Genomics 2020, 14, 20. [Google Scholar] [CrossRef]
- Sun, S.H.; Chen, Q.; Gu, H.J.; Yang, G.; Wang, Y.X.; Huang, X.Y.; Liu, S.S.; Zhang, N.N.; Li, X.F.; Xiong, R.; et al. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis. Cell Host Microbe 2020, 28, 124–133.e124. [Google Scholar] [CrossRef]
- Zhao, F.; Zhang, T.; Sun, X.; Zhang, X.; Chen, L.; Wang, H.; Li, J.; Fan, P.; Lai, L.; Sui, T.; et al. A strategy for Cas13 miniaturization based on the structure and AlphaFold. Nat. Commun. 2023, 14, 5545. [Google Scholar] [CrossRef]
- Handumrongkul, C.; Ye, A.L.; Chmura, S.A.; Soroceanu, L.; Mack, M.; Ice, R.J.; Thistle, R.; Myers, M.; Ursu, S.J.; Liu, Y.; et al. Durable multitransgene expression in vivo using systemic, nonviral DNA delivery. Sci. Adv. 2019, 5, eaax0217. [Google Scholar] [CrossRef]
- Krishnamurthy, S.; Wohlford-Lenane, C.; Kandimalla, S.; Sartre, G.; Meyerholz, D.K.; Theberge, V.; Hallee, S.; Duperre, A.M.; Del’Guidice, T.; Lepetit-Stoffaes, J.P.; et al. Engineered amphiphilic peptides enable delivery of proteins and CRISPR-associated nucleases to airway epithelia. Nat. Commun. 2019, 10, 4906. [Google Scholar] [CrossRef]
- Shahbazi, R.; Sghia-Hughes, G.; Reid, J.L.; Kubek, S.; Haworth, K.G.; Humbert, O.; Kiem, H.P.; Adair, J.E. Targeted homology-directed repair in blood stem and progenitor cells with CRISPR nanoformulations. Nat. Mater. 2019, 18, 1124–1132. [Google Scholar] [CrossRef] [PubMed]
- Guan, S.; Munder, A.; Hedtfeld, S.; Braubach, P.; Glage, S.; Zhang, L.; Lienenklaus, S.; Schultze, A.; Hasenpusch, G.; Garrels, W.; et al. Self-assembled peptide-poloxamine nanoparticles enable in vitro and in vivo genome restoration for cystic fibrosis. Nat. Nanotechnol. 2019, 14, 287–297. [Google Scholar] [CrossRef]
- Baddeley, H.J.E.; Isalan, M. The Application of CRISPR/Cas Systems for Antiviral Therapy. Front. Genome Ed. 2021, 3, 745559. [Google Scholar] [CrossRef]
- Walter, M.; Perrone, R.; Verdin, E. Targeting Conserved Sequences Circumvents the Evolution of Resistance in a Viral Gene Drive against Human Cytomegalovirus. J. Virol. 2021, 95, e0080221. [Google Scholar] [CrossRef] [PubMed]
- Martinez, M.A.; Franco, S. Therapy Implications of Hepatitis C Virus Genetic Diversity. Viruses 2020, 13, 41. [Google Scholar] [CrossRef] [PubMed]
Parameter | SHERLOCK | DETECTR |
---|---|---|
Full name | Specific High-sensitivity Enzymatic Reporter unLOCKing [63,69,70] | DNA Endonuclease Targeted CRISPR Trans Reporter [50,62,71] |
Developed by | SHERLOCK Biosciences | Mammoth Biosciences |
CRISPR enzyme | Cas13 | Cas12 |
Approval | FDA EUA | FDA EUA |
Detection method | Fluorescent signal via Cas13 activation | Fluorescent or colorimetric readout via Cas12 |
Turnaround | 1 h | 30 min to 1 h |
Readout format | Fluorescent | Fluorescent or visual color change |
Parameter | Standard COVID-19 Treatment | CRISPR-Tx (CRISPR-Based Therapeutics) |
---|---|---|
Mode of action | Antivirals like Remdesivir inhibit viral replication by targeting viral RNA polymerase. Monoclonal antibodies neutralize the virus by binding to its spike protein, preventing it from entering host cells. Corticosteroids reduce inflammation and immune response in severe cases. | CRISPR-based therapeutics use gene-editing tools like Cas proteins to directly target and cut viral RNA/DNA. Cas13a, for instance, can be programmed to cleave SARS-CoV-2 RNA, destroying the viral genome before replication. |
Advantages | Widely available and already FDA-approved. Effective in reducing viral load and hospitalization when administered early. Broad range of options depending on disease severity (mild, moderate, severe). Readily accessible in emergency use settings. Well studied with established clinical protocols. | High specificity to the viral RNA, minimizing off-target effects. Potential for rapid adaptation to viral variants due to programmable nature. Could offer a one-time treatment that completely eradicates viral RNA. Diagnostic tool (CRISPR-Dx) has the potential for real-time, at-home detection of viral RNA. |
Limitations | Effectiveness can be limited by viral mutations (e.g., antibody resistance in variants). Antiviral drugs must be administered within a specific time window to be effective. Possible side effects, including liver toxicity and immune suppression (e.g., steroids). High cost for advanced therapies like monoclonal antibodies. | Still in experimental stages with no wide-scale human trials yet for COVID-19. Delivery of CRISPR components into cells poses challenges. Long-term safety of gene-editing approaches is unknown. Regulatory hurdles and ethical concerns surrounding gene-editing technologies. Higher complexity in production and distribution. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Hadi, R.; Poddar, A.; Sonnaila, S.; Bhavaraju, V.S.M.; Agrawal, S. Advancing CRISPR-Based Solutions for COVID-19 Diagnosis and Therapeutics. Cells 2024, 13, 1794. https://doi.org/10.3390/cells13211794
Hadi R, Poddar A, Sonnaila S, Bhavaraju VSM, Agrawal S. Advancing CRISPR-Based Solutions for COVID-19 Diagnosis and Therapeutics. Cells. 2024; 13(21):1794. https://doi.org/10.3390/cells13211794
Chicago/Turabian StyleHadi, Roaa, Abhishek Poddar, Shivakumar Sonnaila, Venkata Suryanarayana Murthy Bhavaraju, and Shilpi Agrawal. 2024. "Advancing CRISPR-Based Solutions for COVID-19 Diagnosis and Therapeutics" Cells 13, no. 21: 1794. https://doi.org/10.3390/cells13211794
APA StyleHadi, R., Poddar, A., Sonnaila, S., Bhavaraju, V. S. M., & Agrawal, S. (2024). Advancing CRISPR-Based Solutions for COVID-19 Diagnosis and Therapeutics. Cells, 13(21), 1794. https://doi.org/10.3390/cells13211794