1
|
Zhong J, Xu Z, Peng J, Guan L, Li J, Zhou Z, Zhang Y, Zhang J, Liu S, Yang Y, Hao X. A CRISPR/Cas13a system based on a dumbbell-shaped hairpin combined with DNA-PAINT to establish the DCP-platform for highly sensitive detection of Hantaan virus RNA. Talanta 2025; 291:127852. [PMID: 40054218 DOI: 10.1016/j.talanta.2025.127852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Rapid and sensitive detection of specific RNA sequences is crucial for clinical diagnosis, surveillance, and biotechnology applications. Currently, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) is the gold standard for RNA detection; however, it is associated with long processing time, complex procedures, and a high false-positive rate. To address these challenges, we developed a novel sensing platform based on CRISPR/Cas13a that incorporates a dumbbell-shaped hairpin and DNA-PAINT for rapid, highly specific, and sensitive RNA analysis. By leveraging the CRISPR/Cas13a system, this platform enables the cleavage of dumbbell-shaped hairpins, which subsequently allows the cleaved primers to initiate cyclic amplification of fluorescent signals. These signals are further enhanced by the binding and dissociation phenomena inherent to DNA-PAINT technology, ultimately achieving remarkable triple signal amplification. Additionally, the system effectively discriminates Hantaan virus RNA from Seoul virus in real samples. Importantly, the platform can be easily adapted for the detection of other RNAs by simply reconfiguring the hybridization region of crRNA. In conclusion, this platform represents a "five-in-one" RNA detection approach that integrates reliability, versatility, robustness, high specificity, and superior quantitative capabilities. It provides novel insights for direct RNA detection based on CRISPR/Cas13a and demonstrates significant potential for advancement in viral diagnostics.
Collapse
Affiliation(s)
- Jiamei Zhong
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Ziyue Xu
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Jiawei Peng
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Liwen Guan
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Jianxiong Li
- Laboratory of Viral Infectious Disease, The Key Laboratory of Important and Emerging Viral Infectious Diseases of Jiangxi Health Commission, Jiangxi Provincial Center for Disease Control and Prevention, Nanchang, Jiangxi, 330029, PR China
| | - Zhuoxun Zhou
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Yu Zhang
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Jie Zhang
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Shiwen Liu
- Laboratory of Viral Infectious Disease, The Key Laboratory of Important and Emerging Viral Infectious Diseases of Jiangxi Health Commission, Jiangxi Provincial Center for Disease Control and Prevention, Nanchang, Jiangxi, 330029, PR China.
| | - Yifei Yang
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China.
| | - Xian Hao
- School of Public Health &Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330031, PR China.
| |
Collapse
|
2
|
Hu H, Xue H, Dong K, Li Y, Liu P, Wang H, Li L, Xiao X, Chen H. Strand displacement-enhanced CRISPR-Cas13a system for ultra-specific detection of RNA single nucleotide variation. Biosens Bioelectron 2025; 280:117445. [PMID: 40194350 DOI: 10.1016/j.bios.2025.117445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/25/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
RNA plays a critical role in biological systems, mediating genetic information transfer and regulating gene expression. However, RNA is susceptible to variations from endogenous and exogenous sources, with potentially profound biological consequences. The CRISPR-Cas13a system has emerged as a promising tool for RNA variation detection due to its cost-effectiveness, sensitivity, and user-friendly nature. Despite this, designing a simple, universal system with high discrimination factor (DF) for single-nucleotide variations remains a challenge. Here, we present the strand displacement-enhanced Cas13a single-nucleotide variation detection assay (SECND), a sensitive, universal, and easy-to-implement method with a high DF for RNA variations. Using SECND, we detected 5 types of single-nucleotide variations, achieving a maximum DF of 1083.2. We validated the assay's effectiveness on miRNA and SARS-CoV-2 genomic RNA simulants, incorporating a 4-way strand displacement mechanism to enhance detection limits to 10 pmol/L and 50 pmol/L, and to identify variations at frequencies as low as 0.01 % and 0.1 %. Additionally, we demonstrated SECND's utility in quantifying single-nucleotide variants of miR-200b and miR-200c in ovarian cancer and retinal glioma cells. This versatile tool not only advances RNA variation detection but also has significant implications for disease research, diagnostics, and viral classification, enhancing our understanding of the CRISPR-Cas13a system and its potential applications.
Collapse
Affiliation(s)
- Hao Hu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Guangzhou, 510280, Guangdong, China; Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hanwen Xue
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kejun Dong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiyuan Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Pei Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haiyun Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Longjie Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xianjin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hui Chen
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
3
|
Zhao L, Zhao Z, Li N, Wang X. The nucleic acid detection using CRISPR/Cas biosensing system with micro-nano modality for point-of-care applications. Talanta 2025; 286:127457. [PMID: 39724853 DOI: 10.1016/j.talanta.2024.127457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Nucleic acid detection is considered the golden standard for diagnosing infectious diseases caused by various pathogens, including viruses, bacteria, and parasites. PCR and other amplification-based technologies are highly sensitive and specific, allowing for accurate detection and identification of low-level causative pathogens by targeting and amplifying their unique genetic segment (DNA or RNA). However, it is important to recognize that machinery-dependent diagnostic methods may only sometimes be available or practical in resource-limited settings, where direct implementation can be challenging. CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)-based diagnostics offer a promising alternative for nucleic acid detection. These methods provide gene sequence-specific targeting, multiplexing capability, rapid result disclosure, and ease of operation, making them suitable for point-of-care (POC) applications. CRISPR-Cas-based nucleic acid detection leverages the intrinsic gene-editing capabilities of CRISPR systems to detect specific DNA or RNA sequences with high precision, ensuring high specificity in identifying pathogens. When integrated with micro- and nano-technologies, CRISPR-based diagnostics gain additional benefits, including automated microfluidic processes, enhanced multiplexed detection, improved sensitivity through nanoparticle integration, and combined detection strategies. In this review, we analyze the motivations for tailoring the CRISPR-Cas system with microfluidic formats or nanoscale materials for nucleic acid biosensing and detection. We discuss and categorize current achievements in such systems, highlighting their differences, commonalities, and opportunities for addressing challenges, particularly for POC diagnostics. Micro- and nano-technologies can significantly enhance the practical utility of the CRISPR-Cas system, enabling more comprehensive diagnostic and surveillance capabilities. By integrating these technologies, CRISPR-based diagnostics can achieve higher levels of automation, sensitivity, and multiplexing, making them invaluable tools in the global effort to diagnose and control infectious diseases.
Collapse
Affiliation(s)
- Liang Zhao
- Center of Excellence for Environmental Safety and Biological Effects, Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| | - Zihao Zhao
- Center of Excellence for Environmental Safety and Biological Effects, Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Ning Li
- Center of Excellence for Environmental Safety and Biological Effects, Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Xiayan Wang
- Center of Excellence for Environmental Safety and Biological Effects, Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
4
|
Armbruster EG, Rani P, Lee J, Klusch N, Hutchings J, Hoffman LY, Buschkaemper H, Enustun E, Adler BA, Inlow K, VanderWal AR, Hoffman MY, Daksh D, Aindow A, Deep A, Rodriguez ZK, Morgan CJ, Ghassemian M, Laughlin TG, Charles E, Cress BF, Savage DF, Doudna JA, Pogliano K, Corbett KD, Villa E, Pogliano J. Sequential membrane- and protein-bound organelles compartmentalize genomes during phage infection. Cell Host Microbe 2025; 33:484-497.e6. [PMID: 40168997 DOI: 10.1016/j.chom.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Many eukaryotic viruses require membrane-bound compartments for replication, but no such organelles are known to be formed by prokaryotic viruses. Bacteriophages of the Chimalliviridae family sequester their genomes within a phage-generated organelle, the phage nucleus, which is enclosed by a lattice of the viral protein ChmA. We show that inhibiting phage nucleus formation arrests infections at an early stage in which the injected phage genome is enclosed within a membrane-bound early phage infection (EPI) vesicle. Early phage genes are expressed from the EPI vesicle, demonstrating its functionality as a prokaryotic, transcriptionally active, membrane-bound organelle. We also show that the phage nucleus is essential, with genome replication beginning after the injected DNA is transferred from the EPI vesicle to the phage nucleus. Our results show that Chimalliviridae require two sophisticated subcellular compartments of distinct compositions and functions that facilitate successive stages of the viral life cycle.
Collapse
Affiliation(s)
- Emily G Armbruster
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Phoolwanti Rani
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Jina Lee
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Niklas Klusch
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Joshua Hutchings
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Lizbeth Y Hoffman
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Hannah Buschkaemper
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Gene Center and Department of Biochemistry, Ludwig Maximilian University of Munich, 80539 Munich, Germany
| | - Eray Enustun
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Benjamin A Adler
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Koe Inlow
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Arica R VanderWal
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Madelynn Y Hoffman
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Daksh Daksh
- National Institute of Science, Education and Research (NISER), Bhubaneshwar 752050, Orissa, India
| | - Ann Aindow
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Zaida K Rodriguez
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Chase J Morgan
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Thomas G Laughlin
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Emeric Charles
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Brady F Cress
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David F Savage
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jennifer A Doudna
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Kit Pogliano
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Kevin D Corbett
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Elizabeth Villa
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA.
| | - Joe Pogliano
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA.
| |
Collapse
|
5
|
Luo H, Yao J, Zhang R. Harnessing RNA base editing for diverse applications in RNA biology and RNA therapeutics. ADVANCED BIOTECHNOLOGY 2025; 3:11. [PMID: 40198443 PMCID: PMC11979053 DOI: 10.1007/s44307-025-00063-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Recent advancements in molecular engineering have established RNA-based technologies as powerful tools for both fundamental research and translational applications. Among the various RNA-based technologies developed, RNA base editing has recently emerged as a groundbreaking advancement. It primarily involves the conversion of adenosine (A) to inosine (I) and cytidine (C) to uridine (U), which are mediated by ADAR and APOBEC enzymes, respectively. RNA base editing has been applied in both biological research and therapeutic contexts. It enables site-directed editing within target transcripts, offering reversible, dose-dependent effects, in contrast to the permanent or heritable changes associated with DNA base editing. Additionally, RNA editing-based profiling of RNA-binding protein (RBP) binding sites facilitates transcriptome-wide mapping of RBP-RNA interactions in specific tissues and at the single-cell level. Furthermore, RNA editing-based sensors have been utilized to express effector proteins in response to specific RNA species. As RNA base editing technologies continue to evolve, we anticipate that they will significantly drive advancements in RNA therapeutics, synthetic biology, and biological research.
Collapse
Affiliation(s)
- Hui Luo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
- Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Jing Yao
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
- Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Rui Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
- Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
6
|
Zilberzwige-Tal S, Altae-Tran H, Kannan S, Wilkinson ME, Vo SCDT, Strebinger D, Edmonds KK, Yao CCJ, Mears KS, Shmakov SA, Makarova KS, Macrae RK, Koonin EV, Zhang F. Reprogrammable RNA-targeting CRISPR systems evolved from RNA toxin-antitoxins. Cell 2025; 188:1925-1940.e20. [PMID: 39970912 DOI: 10.1016/j.cell.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 12/12/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025]
Abstract
Despite ongoing efforts to study CRISPR systems, the evolutionary origins giving rise to reprogrammable RNA-guided mechanisms remain poorly understood. Here, we describe an integrated sequence/structure evolutionary tracing approach to identify the ancestors of the RNA-targeting CRISPR-Cas13 system. We find that Cas13 likely evolved from AbiF, which is encoded by an abortive infection-linked gene that is stably associated with a conserved non-coding RNA (ncRNA). We further characterize a miniature Cas13, classified here as Cas13e, which serves as an evolutionary intermediate between AbiF and other known Cas13s. Despite this relationship, we show that their functions substantially differ. Whereas Cas13e is an RNA-guided RNA-targeting system, AbiF is a toxin-antitoxin (TA) system with an RNA antitoxin. We solve the structure of AbiF using cryoelectron microscopy (cryo-EM), revealing basic structural alterations that set Cas13s apart from AbiF. Finally, we map the key structural changes that enabled a non-guided TA system to evolve into an RNA-guided CRISPR system.
Collapse
Affiliation(s)
- Shai Zilberzwige-Tal
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Han Altae-Tran
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Soumya Kannan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Max E Wilkinson
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Samuel Chau-Duy-Tam Vo
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Strebinger
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - KeHuan K Edmonds
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chun-Chen Jerry Yao
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Molecular Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Kepler S Mears
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sergey A Shmakov
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Kira S Makarova
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Rhiannon K Macrae
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
7
|
Cheng ECK, Lam JKC, Kwon SC. Cytosolic CRISPR RNAs for efficient application of RNA-targeting CRISPR-Cas systems. EMBO Rep 2025; 26:1891-1912. [PMID: 40011676 PMCID: PMC11976971 DOI: 10.1038/s44319-025-00399-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein (CRISPR/Cas) technologies have evolved rapidly over the past decade with the continuous discovery of new Cas systems. In particular, RNA-targeting CRISPR-Cas13 proteins are promising single-effector systems to regulate target mRNAs without altering genomic DNA, yet the current Cas13 systems are restrained by suboptimal efficiencies. Here, we show that U1 promoter-driven CRISPR RNAs (crRNAs) increase the efficiency of various applications, including RNA knockdown and editing, without modifying the Cas13 protein effector. We confirm that U1-driven crRNAs are exported into the cytoplasm, while conventional U6 promoter-driven crRNAs are mostly confined to the nucleus. Furthermore, we reveal that the end positions of crRNAs expressed by the U1 promoter are consistent regardless of guide sequences and lengths. We also demonstrate that U1-driven crRNAs, but not U6-driven crRNAs, can efficiently repress the translation of target genes in combination with catalytically inactive Cas13 proteins. Finally, we show that U1-driven crRNAs can counteract the inhibitory effect of miRNAs. Our simple and effective engineering enables unprecedented cytosolic RNA-targeting applications.
Collapse
Affiliation(s)
- Ezra C K Cheng
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Joe K C Lam
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - S Chul Kwon
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
8
|
Chen J, Chen Y, Huang L, Lin X, Chen H, Xiang W, Liu L. Trans-nuclease activity of Cas9 activated by DNA or RNA target binding. Nat Biotechnol 2025; 43:558-568. [PMID: 38811761 DOI: 10.1038/s41587-024-02255-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 04/18/2024] [Indexed: 05/31/2024]
Abstract
Type V and type VI CRISPR-Cas systems have been shown to cleave nonspecific single-stranded DNA (ssDNA) or single-stranded RNA (ssRNA) in trans, but this has not been observed in type II CRISPR-Cas systems using single guide RNA. We show here that the type II CRISPR-Cas9 systems directed by CRISPR RNA and trans-activating CRISPR RNA dual RNAs show RuvC domain-dependent trans-cleavage activity for both ssDNA and ssRNA substrates. Cas9 possesses sequence preferences for trans-cleavage substrates, preferring to cleave T- or C-rich ssDNA substrates. We find that the trans-cleavage activity of Cas9 can be activated by target ssDNA, double-stranded DNA and ssRNA. The crystal structure of Cas9 in complex with guide RNA and target RNA provides a structural basis for the binding of target RNA to activate Cas9. Based on the trans-cleavage activity of Cas9 and nucleic acid amplification technology, we develop the nucleic acid detection platforms DNA-activated Cas9 detection and RNA-activated Cas9 detection, which are capable of detecting DNA and RNA samples with high sensitivity and specificity.
Collapse
Affiliation(s)
- Jiyun Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Linglong Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xiaofeng Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Hong Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Wenwen Xiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Liang Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
9
|
Pei J, Li L, Li C, Li Z, Wu Y, Kuang H, Ma P, Huang L, Liu J, Tian G. Dumbbell probe-bridged CRISPR/Cas13a and nicking-mediated DNA cascade reaction for highly sensitive detection of colorectal cancer-related microRNAs. Biosens Bioelectron 2025; 273:117190. [PMID: 39862677 DOI: 10.1016/j.bios.2025.117190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths globally, necessitating the development of sensitive and minimally invasive diagnostic approaches. In this study, we present a novel diagnostic strategy by integrating dumbbell probe-mediated CRISPR/Cas13a with nicking-induced DNA cascade reaction (DP-bridged Cas13a/NDCR) for highly sensitive microRNA (miRNA) detection. Target miRNA triggers Cas13a-mediated cleavage of the dumbbell probe, releasing an intermediate strand that hybridizes with a methylene blue-labeled hairpin probe on the electrode surface. Nicking enzyme cleaves the formed duplex DNA, triggering a cascade reaction that amplifies the electrochemical signal. Under optimized conditions, the method demonstrates a detection limit of 8.26 fM for miRNA-21, with reliable specificity and long-term stability. Furthermore, integration with machine learning models using multiple miRNA markers improved diagnostic accuracy, differentiating CRC from colorectal polyps and healthy controls with 100% accuracy in clinical validation cohorts. This study highlights the potential of DP-bridged Cas13a/NDCR as a sensitive and accurate diagnostic tool for CRC.
Collapse
Affiliation(s)
- Jingwen Pei
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Lan Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Chang Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Zongying Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Wu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Haiyang Kuang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Pan Ma
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Li Huang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China.
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China.
| |
Collapse
|
10
|
Cao L, Chen W, Kang W, Lei C, Nie Z. Engineering stimuli-responsive CRISPR-Cas systems for versatile biosensing. Anal Bioanal Chem 2025; 417:1699-1711. [PMID: 39601843 DOI: 10.1007/s00216-024-05678-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
The precise target recognition and nuclease-mediated effective signal amplification capacities of CRISPR-Cas systems have attracted considerable research interest within the biosensing field. Guided by insights into their structural and biochemical mechanisms, researchers have endeavored to engineer the key biocomponents of CRISPR-Cas systems with stimulus-responsive functionalities. By the incorporation of protein/nucleic acid engineering techniques, a variety of conditional CRISPR-Cas systems whose activities depend on the presence of target triggers have been established for the efficient detection of diverse types of non-nucleic acid analytes. In this review, we summarized recent research progress in engineering Cas proteins, guide RNA, and substrate nucleic acids to possess target analyte-responsive abilities for diverse biosensing applications. Furthermore, we also discussed the challenges and future possibilities of the stimulus-responsive CRISPR-Cas systems in versatile biosensing.
Collapse
Affiliation(s)
- Linxin Cao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Wenhui Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Wenyuan Kang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education & Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, Hainan, China
| | - Chunyang Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, Hunan, China.
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, Hunan, China.
| |
Collapse
|
11
|
Singh A, Yasheshwar, Kaushik NK, Kala D, Nagraik R, Gupta S, Kaushal A, Walia Y, Dhir S, Noorani MS. Conventional and cutting-edge advances in plant virus detection: emerging trends and techniques. 3 Biotech 2025; 15:100. [PMID: 40151342 PMCID: PMC11937476 DOI: 10.1007/s13205-025-04253-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
Plant viruses pose a significant threat to global agriculture. For a long time, conventional methods including detection based on visual symptoms, host range investigations, electron microscopy, serological assays (e.g., ELISA, Western blotting), and nucleic acid-based techniques (PCR, RT-PCR) have been used for virus identification. With increased sensitivity, speed, and specificity, new technologies like loop-mediated isothermal amplification (LAMP), high-throughput sequencing (HTS), nanotechnology-based biosensors, and CRISPR diagnostics have completely changed the way plant viruses are detected. Recent advances in detection techniques integrate artificial intelligence (AI), machine learning (ML), and the Internet of Things (IoT) for real-time monitoring. Innovations like hyperspectral imaging, deep learning, and cloud-based IoT platforms further support disease identification and surveillance. Nanotechnology-based lateral flow assays and CRISPR-Cas systems provide rapid, field-deployable solutions. Despite these advancements, challenges such as sequence limitations, multiplexing constraints, and environmental concerns remain. Future research should focus on refining portable on-site diagnostic kits, optimizing nanotechnology applications, and enhancing global surveillance systems. Interdisciplinary collaboration across molecular biology, bioinformatics, and engineering is essential to developing scalable, cost-effective solutions for plant virus detection, ensuring agricultural sustainability and ecosystem protection.
Collapse
Affiliation(s)
- Anjana Singh
- Plant Molecular Virology Lab, Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062 India
- Deshbandhu College, University of Delhi, New Delhi, 110019 India
| | - Yasheshwar
- Department of Botany, Acharya Narendra Dev College, University of Delhi, New Delhi, 110019 India
| | - Naveen K. Kaushik
- Department of Industrial Biotechnology, College of Biotechnology, Chaudhary Charan Singh Haryana Agricultural University, Hisar, Haryana 125004 India
| | - Deepak Kala
- NL-11 Centera Tetrahertz Laboratory, Institute of High-Pressure Physics, Polish Academy of Sciences, 29/37 Sokolowska Street, 01142 Warsaw, Poland
| | - Rupak Nagraik
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Shagun Gupta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207 India
| | - Ankur Kaushal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207 India
| | - Yashika Walia
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207 India
| | - Sunny Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207 India
| | - Md Salik Noorani
- Plant Molecular Virology Lab, Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062 India
| |
Collapse
|
12
|
Cao L, Wang Z, Lei C, Nie Z. Engineered CRISPR/Cas Ribonucleoproteins for Enhanced Biosensing and Bioimaging. Anal Chem 2025; 97:5866-5879. [PMID: 40066952 DOI: 10.1021/acs.analchem.4c06789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
CRISPR-Cas systems represent a highly programmable and precise nucleic acid-targeting platform, which has been strategically engineered as a versatile toolkit for biosensing and bioimaging applications. Nevertheless, their analytical performance is constrained by inherent functional and activity limitations of natural CRISPR/Cas systems, underscoring the critical role of molecular engineering in enhancing their capabilities. This review comprehensively examines recent advancements in engineering CRISPR/Cas ribonucleoproteins (RNPs) to enhance their functional capabilities for advanced molecular detection and cellular imaging. We explore innovative strategies for developing enhanced CRISPR/Cas RNPs, including Cas protein engineering through protein mutagenesis and fusion techniques, and guide RNA engineering via chemical and structural modifications. Furthermore, we evaluate these engineered RNPs' applications in sensitive biomarker detection and live-cell genomic DNA and RNA monitoring, while analyzing the current challenges and prospective developments in CRISPR-Cas RNP engineering for advanced biosensing and bioimaging.
Collapse
Affiliation(s)
- Linxin Cao
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemial Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Zeyuan Wang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemial Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Chunyang Lei
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemial Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Zhou Nie
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemial Biology, Hunan University, Changsha, 410082, Hunan, China
| |
Collapse
|
13
|
Karousi P, Kontos CK, Nikou ST, Carell T, Sideris DC, Scorilas A. Discovery of circular transcripts of the human BCL2-like 12 (BCL2L12) apoptosis-related gene, using targeted nanopore sequencing, provides new insights into circular RNA biology. Funct Integr Genomics 2025; 25:66. [PMID: 40106061 PMCID: PMC11923030 DOI: 10.1007/s10142-025-01578-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Circular RNAs (circRNAs) constitute an RNA type formed by back-splicing. BCL2-like 12 (BCL2L12) is an apoptosis-related gene comprising 7 exons. In this study, we used targeted nanopore sequencing to identify circular BCL2L12 transcripts in human colorectal cancer cells and investigated the effect of circRNA silencing on mRNA expression of the parental gene. In brief, nanopore sequencing following nested PCR amplification of cDNAs of BCL2L12 circRNAs from 7 colorectal cancer cell lines unraveled 46 BCL2L12 circRNAs, most of which described for the first time. Interestingly, 40 novel circRNAs are likely to form via back-splicing between non-canonical back-splice sites residing in highly similar regions of the primary transcripts. All back-splice junctions were validated using next-generation sequencing (NGS) after circRNA enrichment. Surprisingly, 2 novel circRNAs also comprised a poly(A) tract after BCL2L12 exon 7; this poly(A) tract was back-spliced to exon 1, in both cases. Furthermore, the selective silencing of a BCL2L12 circRNA resulted in a subsequent decrease of BCL2L12 mRNA levels in HCT 116 cells, thus providing evidence of parental gene expression regulation by circRNAs. In conclusion, our study led to the discovery of many circular transcripts from a single human gene and provided new insights into circRNA biogenesis and mode of action.
Collapse
Affiliation(s)
- Paraskevi Karousi
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| | - Stavroula T Nikou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Thomas Carell
- Department for Chemistry, Institute for Chemical Epigenetics, Ludwig Maximilian University of Munich, Munich, Germany
| | - Diamantis C Sideris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
Moreno-Sánchez I, Hernández-Huertas L, Nahón-Cano D, Martínez-García PM, Treichel AJ, Gómez-Marin C, Tomás-Gallardo L, da Silva Pescador G, Kushawah G, Egidy R, Perera A, Díaz-Moscoso A, Cano-Ruiz A, Walker JA, Muñoz MJ, Holden K, Galcerán J, Nieto MÁ, Bazzini AA, Moreno-Mateos MA. Enhanced RNA-targeting CRISPR-Cas technology in zebrafish. Nat Commun 2025; 16:2591. [PMID: 40091120 PMCID: PMC11911407 DOI: 10.1038/s41467-025-57792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
CRISPR-Cas13 RNA-targeting systems are widely used in basic and applied sciences. However, its application has recently generated controversy due to collateral activity in mammalian cells and mouse models. Moreover, its competence could be improved in vivo. Here, we optimized transient formulations as ribonucleoprotein complexes or mRNA-gRNA combinations to enhance the CRISPR-RfxCas13d system in zebrafish. We i) use chemically modified gRNAs to allow more penetrant loss-of-function phenotypes, ii) improve nuclear RNA targeting, and iii) compare different computational models and determine the most accurate to predict gRNA activity in vivo. Furthermore, we demonstrate that transient CRISPR-RfxCas13d can effectively deplete endogenous mRNAs in zebrafish embryos without inducing collateral effects, except when targeting extremely abundant and ectopic RNAs. Finally, we implement alternative RNA-targeting CRISPR-Cas systems such as CRISPR-Cas7-11 and CRISPR-DjCas13d. Altogether, these findings contribute to CRISPR-Cas technology optimization for RNA targeting in zebrafish through transient approaches and assist in the progression of in vivo applications.
Collapse
Grants
- F31 HD110268 NICHD NIH HHS
- R01 GM136849 NIGMS NIH HHS
- R21 OD034161 NIH HHS
- This work was supported by Ramon y Cajal (RyC-2017-23041), PID2021-127535NB-I00, CNS2022-135564 and CEX2020-001088-M grants funded by MICIU/AEI/ 10.13039/501100011033 by “ERDF A way of making Europe” (“ERDF/EU”), and by ESF Investing in your future from Ministerio de Ciencia, Innovación y Universidades and European Union (M.A.M.-M.). This work has also been co-financed by the Spanish Ministry of Science and Innovation with funds from the European Union NextGenerationEU (PRTR-C17.I1) and the Regional Ministry of University, Research and Innovation of the Autonomous Community of Andalusia within the framework of the Biotechnology Plan applied to Health. The Moreno-Mateos lab was also funded by European Regional Development Fund (FEDER 80% of the total funding) by the Ministry of Economy, Knowledge, Business and University, of the Government of Andalusia, within the framework of the FEDER Andalusia 2014-2020 operational program within the objective "Promotion and generation of frontier knowledge and knowledge oriented to the challenges of society, development of emerging technologies (grant UPO-1380590)” and by the Fondo Europeo de Desarrollo Regional (FEDER) and Consejería de Transformación Económica, Industria, Conocimiento y Universidades de la Junta de Andalucía, within the operative program FEDER Andalucía 2014-2020 (01 - Refuerzo de la investigación, el desarrollo tecnológico y la innovación, grant P20_00866). M.A.M.-M. was the recipient of the Genome Engineer Innovation 2019 Grant from Synthego. The CABD is an institution funded by University Pablo de Olavide, Consejo Superior de Investigaciones Científicas (CSIC), and Junta de Andalucía.
Collapse
Affiliation(s)
- Ismael Moreno-Sánchez
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
- Instituto de Neurociencias (CSIC-UMH), Alicante, Spain
| | - Luis Hernández-Huertas
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | - Daniel Nahón-Cano
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | - Pedro Manuel Martínez-García
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
| | | | - Carlos Gómez-Marin
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | - Laura Tomás-Gallardo
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Proteomics and Biochemistry Platform, Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
| | | | - Gopal Kushawah
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Rhonda Egidy
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Anoja Perera
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Alejandro Díaz-Moscoso
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Proteomics and Biochemistry Platform, Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Instituto de Investigaciones Químicas (IIQ-CICIC), CSIC-US, Seville, Spain
| | - Alejandra Cano-Ruiz
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | | | - Manuel J Muñoz
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain
| | | | - Joan Galcerán
- Instituto de Neurociencias (CSIC-UMH), Alicante, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - M Ángela Nieto
- Instituto de Neurociencias (CSIC-UMH), Alicante, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Ariel A Bazzini
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Miguel A Moreno-Mateos
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Seville, Spain.
- Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, Seville, Spain.
| |
Collapse
|
15
|
Chai HX, Bamert RS, Knott GJ. Methods for Cas13a expression and purification for use in CRISPR diagnostics. Methods Enzymol 2025; 712:225-244. [PMID: 40121074 DOI: 10.1016/bs.mie.2025.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The threat of emerging infectious diseases (e.g., SARS-CoV-2 the RNA virus responsible for the COVID-19 pandemic) has highlighted the importance of accurate and rapid testing for screening, patient diagnosis, and effective treatment of infectious disease. Nucleic acid diagnostic tools such as qPCR are considered the gold standard, providing a sensitive, accurate, and robust method of detection. However, these conventional diagnostic platforms are resource intensive, limited in some applications, and are almost always confined to laboratory settings. With the increasing demand for low-cost, rapid, and accurate point-of-care diagnostics, CRISPR-based systems have emerged as powerful tools to augment detection capabilities. Of note is the potent RNA detection enzyme, Leptotrichia buccalis (Lbu) Cas13a, which is capable of rapid RNA detection in complex mixtures with or without pre-amplification. To support its wide-spread use, we describe a detailed method for the expression, purification, and validation of LbuCas13a for use in molecular diagnostics.
Collapse
Affiliation(s)
- Her Xiang Chai
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Rebecca S Bamert
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Gavin J Knott
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
16
|
Zhao X, Yang L, Li P, Cheng Z, Jia Y, Luo L, Bi A, Xiong H, Zhang H, Xu H, Zhang J, Zhang Y. High-accuracy crRNA array assembly strategy for multiplex CRISPR. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102428. [PMID: 39897580 PMCID: PMC11787013 DOI: 10.1016/j.omtn.2024.102428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/10/2024] [Indexed: 02/04/2025]
Abstract
Simultaneous targeting of multiple loci with the CRISPR system, a tool known as multiplex CRISPR, offers greater feasibility for manipulating and elucidating the intricate and redundant endogenous networks underlying complex cellular functions. Owing to the versatility of continuously emerging Cas nucleases and the use of CRISPR arrays, multiplex CRISPR has been implemented in numerous in vitro and in vivo studies. However, a streamlined, practical strategy for CRISPR array assembly that is both convenient and accurate is lacking. Here, we present a novel, highly accurate, cost-, and time-saving strategy for CRISPR array assembly. Using this strategy, we efficiently assembled 12 CRISPR RNAs (crRNAs) (for AsCas12a) and 15 crRNAs (for RfxCas13d) in a single reaction. CRISPR arrays driven by Pol II promoters exhibited a distinct expression pattern compared with those driven by Pol III promoters, which could be exploited for specific distributions of CRISPR intensity. Improved approaches were subsequently designed and validated for expressing long CRISPR arrays. The study provides a flexible and powerful tool for the convenient implementation of multiplex CRISPR across DNA and RNA, facilitating the dissection of sophisticated cellular networks and the future realization of multi-target gene therapy.
Collapse
Affiliation(s)
- Xiangtong Zhao
- Henan Provincial Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, Henan, China
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Lixian Yang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peng Li
- Department of Gastroenterology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zijing Cheng
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongshi Jia
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Limin Luo
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Aihong Bi
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hanchu Xiong
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haibo Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hongen Xu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jinrui Zhang
- Henan Provincial Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, Henan, China
| | - Yaodong Zhang
- Henan Provincial Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, Henan, China
| |
Collapse
|
17
|
Dunkley ORS, Bell AG, Modi NH, Huang Y, Tseng S, Reiss R, Daivaa N, Davis JL, Vargas DA, Banada P, Xie YL, Myhrvold C. A Streamlined Point-of-Care CRISPR Test for Tuberculosis Detection Directly from Sputum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322517. [PMID: 40034782 PMCID: PMC11875272 DOI: 10.1101/2025.02.19.25322517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Mycobacterium tuberculosis (Mtb) is a major threat to global health and is responsible for over one million deaths each year. To stem the tide of cases and maximize opportunities for early interventions, there is an urgent need for affordable and simple means of tuberculosis diagnosis in under-resourced areas. We sought to develop a CRISPR-based isothermal assay coupled with a compatible, straightforward sample processing technique for point-of-care use. Here, we combine Recombinase Polymerase Amplification (RPA) with Cas13a and Cas12a, to create two parallelised one-pot assays that detect two conserved elements of Mtb (IS6110 and IS1081) and an internal control targeting human DNA. These assays were shown to be compatible with lateral flow and can be readily lyophilized. Our finalized assay exhibited sensitivity over a wide range of bacterial loads (105 to 102 CFU/mL) in sputum. The limit of detection (LoD) of the assay was determined to be 69.0 (51.0 - 86.9) CFU/mL for Mtb strain H37Rv spiked in sputum and 80.5 (59.4 - 101.6) CFU/mL for M. bovis BCG. Our assay showed no cross reactivity against a wide range of bacterial/fungal isolates. Clinical tests on 13 blinded sputum samples revealed 100% (6/6) sensitivity and 100% (7/7) specificity compared to culture. Our assay exhibited comparable sensitivity in clinical samples to the microbiological gold standard, TB culture, and to the nucleic acid state-of-the-art, GeneXpert MTB/RIF Ultra. This technology streamlines TB diagnosis from sample extraction to assay readout in a rapid and robust format, making it the first test to combine amplification and detection while being compatible with both lateral flow and lyophilization.
Collapse
Affiliation(s)
- Owen R. S. Dunkley
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Alexandra G. Bell
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Nisha H. Modi
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Yujia Huang
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Soleil Tseng
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Robert Reiss
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Naranjargal Daivaa
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - J. Lucian Davis
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Pulmonary, Critical Care, and Sleep Medicine Section, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Deninson Alejandro Vargas
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Padmapriya Banada
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Yingda L. Xie
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, New Jersey, 08544, USA
- Department of Chemistry, Princeton University, Princeton, New Jersey, 08544, USA
| |
Collapse
|
18
|
van Dongen JE, Segerink LI. Building the Future of Clinical Diagnostics: An Analysis of Potential Benefits and Current Barriers in CRISPR/Cas Diagnostics. ACS Synth Biol 2025; 14:323-331. [PMID: 39880685 PMCID: PMC11854988 DOI: 10.1021/acssynbio.4c00816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
Advancements in molecular diagnostics, such as polymerase chain reaction and next-generation sequencing, have revolutionized disease management and prognosis. Despite these advancements in molecular diagnostics, the field faces challenges due to high operational costs and the need for sophisticated equipment and highly trained personnel besides having several technical limitations. The emergent field of CRISPR/Cas sensing technology is showing promise as a new paradigm in clinical diagnostics, although widespread clinical adoption remains limited. This perspective paper discusses specific cases where CRISPR/Cas technology can surmount the challenges of existing diagnostic methods by stressing the significant role that CRISPR/Cas technology can play in revolutionizing clinical diagnostics. It underscores the urgency and importance of addressing the technological and regulatory hurdles that must be overcome to harness this technology effectively in clinical laboratories.
Collapse
Affiliation(s)
- Jeanne E. van Dongen
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical
Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University
of Twente, P.O. Box 217, 7500 AE Enschede, The
Netherlands
| | - Loes I. Segerink
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical
Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University
of Twente, P.O. Box 217, 7500 AE Enschede, The
Netherlands
| |
Collapse
|
19
|
Xia C, Colognori D, Jiang XS, Xu K, Doudna JA. Single-molecule live-cell RNA imaging with CRISPR-Csm. Nat Biotechnol 2025:10.1038/s41587-024-02540-5. [PMID: 39966655 DOI: 10.1038/s41587-024-02540-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 12/17/2024] [Indexed: 02/20/2025]
Abstract
Understanding the diverse dynamic behaviors of individual RNA molecules in single cells requires visualizing them at high resolution in real time. However, single-molecule live-cell imaging of unmodified endogenous RNA has not yet been achieved in a generalizable manner. Here, we present single-molecule live-cell fluorescence in situ hybridization (smLiveFISH), a robust approach that combines the programmable RNA-guided, RNA-targeting CRISPR-Csm complex with multiplexed guide RNAs for direct and efficient visualization of single RNA molecules in a range of cell types, including primary cells. Using smLiveFISH, we track individual native NOTCH2 and MAP1B transcripts in living cells and identify two distinct localization mechanisms including the cotranslational translocation of NOTCH2 mRNA at the endoplasmic reticulum and directional transport of MAP1B mRNA toward the cell periphery. This method has the potential to unlock principles governing the spatiotemporal organization of native transcripts in health and disease.
Collapse
Affiliation(s)
- Chenglong Xia
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - David Colognori
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Xueyang Stephen Jiang
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Ke Xu
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Jennifer A Doudna
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA.
- Innovative Genomics Institute, University of California, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Department of Chemistry, University of California, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
20
|
Álvarez-Rodríguez A, Li Z, Jin BK, Stijlemans B, Geldhof P, Magez S. A CRISPR-Cas-based recombinase polymerase amplification assay for ultra-sensitive detection of active Trypanosoma brucei evansi infections. Front Mol Biosci 2025; 12:1512970. [PMID: 40026698 PMCID: PMC11867955 DOI: 10.3389/fmolb.2025.1512970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Control of Trypanosoma brucei evansi (T. b. evansi) infections remains a significant challenge in managing Surra, a widespread veterinary disease affecting both wild and domestic animals. In the absence of an effective vaccine, accurate diagnosis followed by treatment is crucial for successful disease management. However, existing diagnostic methods often fail to detect active infections, particularly in field conditions. Recent advancements in CRISPR-Cas technology, combined with state-of-the-art isothermal amplification assays, offer a promising solution. This approach has led us to the development of a TevRPA-CRISPR assay, a highly sensitive and specific T. b. evansi diagnostic tool suitable for both laboratory and field settings. Methods First, the TevCRISPR-Cas12b cleavage assay was developed and optimized, and its analytical sensitivity was evaluated. Next, this technology was integrated with the TevRPA to create the TevRPA-CRISPR test, with the reaction conditions being optimized and its analytical sensitivity and specificity assessed. Finally, the test's accuracy in detecting both active and cured T. b. evansi infections was evaluated. Results The optimized TevCRISPR-Cas12b cleavage assay demonstrated the ability to detect T. b. evansi target DNA at picomolar concentrations. Integrating TevCRISPR-Cas12b with RPA in Two-Pot and One-Pot TevRPA-CRISPR tests achieved up to a 100-fold increase in analytical sensitivity over RPA alone, detecting attomolar concentrations of T. b. evansi target DNA, while maintaining analytical specificity for T. b. evansi. Both assays exhibited performance comparable to the gold standard TevPCR in experimental mouse infections, validating their effectiveness for detecting active infections and assessing treatment efficacy. Discussion The TevRPA-CRISPR tests prove highly effective for diagnosing active infections and assessing treatment efficacy, while being adaptable for both laboratory and field use. Thus, the TevRPA-CRISPR assays emerge as a promising addition to current diagnostic tools, offering efficient and reliable detection of active T. b. evansi infections.
Collapse
Affiliation(s)
- Andrés Álvarez-Rodríguez
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biochemistry and Microbiology (WE10), Ghent University, Ghent, Belgium
| | - Zeng Li
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Bo-Kyung Jin
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Benoit Stijlemans
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Peter Geldhof
- Laboratory for Parasitology and Parasitic Diseases, Department of Translational Physiology, Infectiology and Public Health (DI04), Ghent University, Ghent, Belgium
| | - Stefan Magez
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
21
|
Fry LE, Major L, Salman A, McDermott LA, Yang J, King AJ, McClements ME, MacLaren RE. Comparison of CRISPR-Cas13b RNA base editing approaches for USH2A-associated inherited retinal degeneration. Commun Biol 2025; 8:200. [PMID: 39922978 PMCID: PMC11807095 DOI: 10.1038/s42003-025-07557-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/15/2025] [Indexed: 02/10/2025] Open
Abstract
CRISPR-Cas13 systems have therapeutic promise for the precise correction of point mutations in RNA. Using adenosine deaminase acting on RNA (ADAR) effectors, A-I base conversions can be targeted using guide RNAs (gRNAs). We compare the Cas13 effectors PspCas13b and Cas13bt3 for the repair of the gene USH2A, a common cause of inherited retinal disease and Usher syndrome. In cultured cells, we demonstrate up to 80% efficiency for the repair of the common c.11864 G > A and its murine equivalent c.11840 G > A, across different gRNAs and promoters. We develop and characterize a mouse model of Usher syndrome carrying the c.11840 G > A mutation designed for the evaluation of base editors for inherited retinal disease. Finally, we compare Cas13 effectors delivered via AAV for the repair of Ush2a in photoreceptors. Mean RNA editing rates in photoreceptors across different constructs ranged from 0.32% to 2.04%, with greater efficiency in those injected with PspCas13b compared to Cas13bt3 constructs. In mice injected with PspCas13b constructs, usherin protein was successfully restored and correctly localized to the connecting cilium following RNA editing. These results support the development of transcriptome targeting gene editing therapies for retinal disease.
Collapse
Affiliation(s)
- Lewis E Fry
- Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Centre for Eye Research Australia, East Melbourne, VIC, Australia
| | - Lauren Major
- Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Ahmed Salman
- Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lucy A McDermott
- Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jun Yang
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Andrew J King
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Michelle E McClements
- Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Robert E MacLaren
- Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
22
|
Grimm MS, Myhrvold C. Using CRISPR for viral nucleic acid detection. Methods Enzymol 2025; 712:245-275. [PMID: 40121076 DOI: 10.1016/bs.mie.2025.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Pathogenic microorganisms, such as viruses, have threatened human health and will continue to contribute to future epidemics and pandemics, highlighting the importance of developing effective diagnostics. To contain viral outbreaks within populations, fast and early diagnosis of infected individuals is essential. Although current standard methods are highly sensitive and specific, like RT-qPCR, some can have slow turnaround times, which can hinder the prevention of viral transmission. The discovery of CRISPR-Cas systems in bacteria and archaea initially revolutionized the world of genome editing. Intriguingly, CRISPR-Cas enzymes also have the ability to detect nucleic acids with high sensitivity and specificity, which sparked the interest of researchers to also explore their potential in diagnosis of viral pathogens. In particular, the CRISPR-Cas13 system has been used as a tool for detecting viral nucleic acids. Cas13's capability to detect both target RNA and non-specific RNAs has led to the development of detection methods that leverage these characteristics through designing specific detection read-outs. Optimization of viral sample collection, amplification steps and the detection process within the Cas13 detection workflow has resulted in assays with high sensitivity, rapid turnaround times and the capacity for large-scale implementation. This review focuses on the significant innovations of various CRISPR-Cas13-based viral nucleic acid detection methods, comparing their strengths and weaknesses while highlighting Cas13's great potential as a tool for viral diagnostics.
Collapse
Affiliation(s)
- Maaike S Grimm
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States; Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ, United States; Department of Chemistry, Princeton University, Princeton, NJ, United States.
| |
Collapse
|
23
|
Low SJ, O'Neill M, Kerry WJ, Wild N, Krysiak M, Nong Y, Azzato F, Hor E, Williams L, Taiaroa G, Steinig E, Pasricha S, Williamson DA. PathoGD: an integrative genomics approach to primer and guide RNA design for CRISPR-based diagnostics. Commun Biol 2025; 8:147. [PMID: 39885339 PMCID: PMC11782503 DOI: 10.1038/s42003-025-07591-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025] Open
Abstract
Critical to the success of CRISPR-based diagnostic assays is the selection of a diagnostic target highly specific to the organism of interest, a process often requiring iterative cycles of manual selection, optimisation, and redesign. Here we present PathoGD, a bioinformatic pipeline for rapid and high-throughput design of RPA primers and gRNAs for CRISPR-Cas12a-based pathogen detection. PathoGD is fully automated, leverages publicly available sequences and is scalable to large datasets, allowing rapid continuous monitoring and validation of primer/gRNA sets to ensure ongoing assay relevance. We designed primers and gRNAs for five clinically relevant bacterial pathogens, and experimentally validated a subset of the designs for detecting Streptococcus pyogenes and/or Neisseria gonorrhoeae in assays with and without pre-amplification. We demonstrated high specificity of primers and gRNAs designed, with minimal off-target signal observed for all combinations. We anticipate PathoGD will be an important resource for assay design for current and emerging pathogens. PathoGD is available on GitHub at https://github.com/sjlow23/pathogd .
Collapse
Affiliation(s)
- Soo Jen Low
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | - Matthew O'Neill
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - William J Kerry
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - Natasha Wild
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - Marcelina Krysiak
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Yi Nong
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Francesca Azzato
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Eileen Hor
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Lewis Williams
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - George Taiaroa
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Eike Steinig
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Shivani Pasricha
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia.
| | - Deborah A Williamson
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- School of Medicine, University of St Andrews, Fife, Scotland
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| |
Collapse
|
24
|
Doctor Y, Sanghvi M, Mali P. A Manual for Genome and Transcriptome Engineering. IEEE Rev Biomed Eng 2025; 18:250-267. [PMID: 39514364 PMCID: PMC11875898 DOI: 10.1109/rbme.2024.3494715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Genome and transcriptome engineering have emerged as powerful tools in modern biotechnology, driving advancements in precision medicine and novel therapeutics. In this review, we provide a comprehensive overview of the current methodologies, applications, and future directions in genome and transcriptome engineering. Through this, we aim to provide a guide for tool selection, critically analyzing the strengths, weaknesses, and best use cases of these tools to provide context on their suitability for various applications. We explore standard and recent developments in genome engineering, such as base editors and prime editing, and provide insight into tool selection for change of function (knockout, deletion, insertion, substitution) and change of expression (repression, activation) contexts. Advancements in transcriptome engineering are also explored, focusing on established technologies like antisense oligonucleotides (ASOs) and RNA interference (RNAi), as well as recent developments such as CRISPR-Cas13 and adenosine deaminases acting on RNA (ADAR). This review offers a comparison of different approaches to achieve similar biological goals, and consideration of high-throughput applications that enable the probing of a variety of targets. This review elucidates the transformative impact of genome and transcriptome engineering on biological research and clinical applications that will pave the way for future innovations in the field.
Collapse
Affiliation(s)
| | | | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, CA 92039, USA
| |
Collapse
|
25
|
Jung J, Dreyer KS, Dray KE, Muldoon JJ, George J, Shirman S, Cabezas MD, d’Aquino AE, Verosloff MS, Seki K, Rybnicky GA, Alam KK, Bagheri N, Jewett MC, Leonard JN, Mangan NM, Lucks JB. Developing, Characterizing, and Modeling CRISPR-Based Point-of-Use Pathogen Diagnostics. ACS Synth Biol 2025; 14:129-147. [PMID: 39670656 PMCID: PMC11744932 DOI: 10.1021/acssynbio.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024]
Abstract
Recent years have seen intense interest in the development of point-of-care nucleic acid diagnostic technologies to address the scaling limitations of laboratory-based approaches. Chief among these are combinations of isothermal amplification approaches with CRISPR-based detection and readouts of target products. Here, we contribute to the growing body of rapid, programmable point-of-care pathogen tests by developing and optimizing a one-pot NASBA-Cas13a nucleic acid detection assay. This test uses the isothermal amplification technique NASBA to amplify target viral nucleic acids, followed by the Cas13a-based detection of amplified sequences. We first demonstrate an in-house formulation of NASBA that enables the optimization of individual NASBA components. We then present design rules for NASBA primer sets and LbuCas13a guide RNAs for the fast and sensitive detection of SARS-CoV-2 viral RNA fragments, resulting in 20-200 aM sensitivity. Finally, we explore the combination of high-throughput assay condition screening with mechanistic ordinary differential equation modeling of the reaction scheme to gain a deeper understanding of the NASBA-Cas13a system. This work presents a framework for developing a mechanistic understanding of reaction performance and optimization that uses both experiments and modeling, which we anticipate will be useful in developing future nucleic acid detection technologies.
Collapse
Affiliation(s)
- Jaeyoung
K. Jung
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Water Research, Northwestern University, Evanston, Illinois 60208, United States
| | - Kathleen S. Dreyer
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Kate E. Dray
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Joseph J. Muldoon
- Department
of Medicine, University of California, San
Francisco, San Francisco, California 94143, United States
- Gladstone-UCSF
Institute of Genomic Immunology, San Francisco, California 94158, United States
| | - Jithin George
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, Illinois 60208, United States
- NSF-Simons
Center for Quantitative Biology, Northwestern
University, Evanston, Illinois 60208, United States
| | - Sasha Shirman
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- NSF-Simons
Center for Quantitative Biology, Northwestern
University, Evanston, Illinois 60208, United States
| | - Maria D. Cabezas
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Illinois 60208, United States
| | - Anne E. d’Aquino
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Stemloop,
Inc., Evanston, Illinois 60201, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
| | - Matthew S. Verosloff
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
| | - Kosuke Seki
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Grant A. Rybnicky
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, Evanston, Illinois 60208, United States
| | | | - Neda Bagheri
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
- Departments
of Biology and Chemical Engineering, University
of Washington, Seattle, Washington 98195, United States
| | - Michael C. Jewett
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Joshua N. Leonard
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
| | - Niall M. Mangan
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, Illinois 60208, United States
- NSF-Simons
Center for Quantitative Biology, Northwestern
University, Evanston, Illinois 60208, United States
| | - Julius B. Lucks
- Department
of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
- Center
for Water Research, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, Evanston, Illinois 60208, United States
| |
Collapse
|
26
|
Xu X, Zhang Y, Liu J, Wei S, Li N, Yao X, Wang M, Su X, Jing G, Xu J, Liu Y, Lu Y, Cheng J, Xu Y. Concurrent Detection of Protein and miRNA at the Single Extracellular Vesicle Level Using a Digital Dual CRISPR-Cas Assay. ACS NANO 2025; 19:1271-1285. [PMID: 39688838 DOI: 10.1021/acsnano.4c13557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The simultaneous detection of proteins and microRNA (miRNA) at the single extracellular vesicle (EV) level shows great promise for precise disease profiling, owing to the heterogeneity and scarcity of tumor-derived EVs. However, a highly reliable method for multiple-target analysis of single EVs remains to be developed. In this study, a digital dual CRISPR-Cas-powered Single EV Evaluation (ddSEE) system was proposed to enable the concurrent detection of surface protein and inner miRNA of EVs at the single-molecule level. By optimizing simultaneous reaction conditions of CRISPR-Cas12a and CRISPR-Cas13a, the surface protein of EVs was detected by Cas12a using antibody-DNA conjugates to transfer the signal of the protein to DNA, while the inner miRNA was analyzed by Cas13a through EV-liposome fusion. A microfluidic chip containing 188,000 microwells was used to convert the CRISPR-Cas system into a digital assay format to enable the absolute quantification of miRNA/protein-positive EVs without bias through fluorescence imaging, which can detect as few as 214 EVs/μL. Finally, a total of 31 blood samples, 21 from breast cancer patients and 10 from healthy donors, were collected and tested, achieving a diagnostic accuracy of 92% in distinguishing patients with breast cancer from healthy donors. With its absolute quantification, ease of use, and multiplexed detection capability, the ddSEE system demonstrates its great potential for both EV research and clinical applications.
Collapse
Affiliation(s)
- Xun Xu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Yuanyue Zhang
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Jiajia Liu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- CapitalBio Technology, Beijing 101111, China
| | - Shujin Wei
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Nan Li
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Xintong Yao
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Muxue Wang
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaohan Su
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Gaoshan Jing
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Junquan Xu
- Iomics Biosciences, Beijing 101318, China
| | - Yan Liu
- Iomics Biosciences, Beijing 101318, China
| | - Ying Lu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102200, China
| | - Jing Cheng
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102200, China
| | - Youchun Xu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102200, China
| |
Collapse
|
27
|
Lin Y, Li C, Chen Y, Gao J, Li J, Huang C, Liu Z, Wang W, Zheng X, Song X, Wu J, Wu J, Luo OJ, Tu Z, Li S, Li XJ, Lai L, Yan S. RNA-Targeting CRISPR/CasRx system relieves disease symptoms in Huntington's disease models. Mol Neurodegener 2025; 20:4. [PMID: 39806441 PMCID: PMC11727607 DOI: 10.1186/s13024-024-00794-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND HD is a devastating neurodegenerative disorder caused by the expansion of CAG repeats in the HTT. Silencing the expression of mutated proteins is a therapeutic direction to rescue HD patients, and recent advances in gene editing technology such as CRISPR/CasRx have opened up new avenues for therapeutic intervention. METHODS The CRISPR/CasRx system was employed to target human HTT exon 1, resulting in an efficient knockdown of HTT mRNA. This therapeutic effect was substantiated in various models: HEK 293 T cell, the HD 140Q-KI mouse, and the HD-KI pig model. The efficiency of the knockdown was analyzed through Western blot and RT-qPCR. Additionally, neuropathological changes were examined using Western blot, immunostaining, and RNA sequencing. The impact on motor abilities was assessed via behavioral experiments, providing a comprehensive evaluation of the treatment's effectiveness. RESULTS CRISPR/CasRx system can significantly reduce HTT mRNA levels across various models, including HEK 293 T cells, HD 140Q-KI mice at various disease stages, and HD-KI pigs, and resulted in decreased expression of mHTT. Utilizing the CRISPR/CasRx system to knock down HTT RNA has shown to ameliorate gliosis in HD 140Q-KI mice and delay neurodegeneration in HD pigs. CONCLUSIONS These findings highlight the effectiveness of the RNA-targeting CRISPR/CasRx as a potential therapeutic strategy for HD. Furthermore, the success of this approach provides valuable insights and novel avenues for the treatment of other genetic disorders caused by gene mutations.
Collapse
Affiliation(s)
- Yingqi Lin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Caijuan Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yizhi Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiale Gao
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiawei Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Chunhui Huang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Zhaoming Liu
- China-New Zealand Joint Laboratory On Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Institutes of Biomedicine and Health , Chinese Academy of Sciences, Guangzhou, China
| | - Wei Wang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao Zheng
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xichen Song
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jianhao Wu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiaxi Wu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhuchi Tu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Liangxue Lai
- China-New Zealand Joint Laboratory On Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Institutes of Biomedicine and Health , Chinese Academy of Sciences, Guangzhou, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
- Department of Neurology, Faculty of Medical Science, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
28
|
Fei X, Lei C, Ren W, Liu C. 'Splice-at-will' Cas12a crRNA engineering enabled direct quantification of ultrashort RNAs. Nucleic Acids Res 2025; 53:gkaf002. [PMID: 39831307 PMCID: PMC11744192 DOI: 10.1093/nar/gkaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
We present a robust 'splice-at-will' CRISPR RNA (crRNA) engineering mechanism that overcomes the limitations of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system in directly detecting ultrashort RNAs. In this strategy, an intact Cas12a crRNA can be split from almost any site of the spacer region to obtain a truncated crRNA (tcrRNA) that cannot activate Cas12a even after binding an auxiliary DNA activator. While splicing tcrRNAs with a moiety of ultrashort RNA, the formed combination can work together to activate Cas12a efficiently, enabling 'splice-at-will' crRNA engineering. Importantly, the 'splice-at-will' crRNA exhibits almost the same trans-cleavage activation efficiency as that of a conventional intact crRNA. Therefore, by rationally designing a DNA auxiliary activator with a conserved tcrRNA-complementary sequence and an arbitrary short RNA-of-interest recognition domain, a general sensing system is established that directly utilizes traditional DNA-activated Cas12a to detect ultrashort RNAs. This 'splice-at-will' crRNA engineering strategy could faithfully detect ultrashort RNA sequences as short as 6-8 nt, which cannot be achieved by conventional Cas12a and Cas13a systems. Additionally, through flexible splicing site design, our method can precisely distinguish single-base differences in microRNA and other short RNA sequences. This work has significantly expanded the Cas12a-based diagnostic toolbox and opened new avenues for ultrashort RNA detection.
Collapse
Affiliation(s)
- Xinrui Fei
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 620 West Chang’an Avenue, Chang’an District, Xi’an, Shaanxi 710119, P.R. China
| | - Chao Lei
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 620 West Chang’an Avenue, Chang’an District, Xi’an, Shaanxi 710119, P.R. China
| | - Wei Ren
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 620 West Chang’an Avenue, Chang’an District, Xi’an, Shaanxi 710119, P.R. China
| | - Chenghui Liu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry & Chemical Engineering, Shaanxi Normal University, 620 West Chang’an Avenue, Chang’an District, Xi’an, Shaanxi 710119, P.R. China
| |
Collapse
|
29
|
Kempthorne L, Vaizoglu D, Cammack AJ, Carcolé M, Roberts MJ, Mikheenko A, Fisher A, Suklai P, Muralidharan B, Kroll F, Moens TG, Yshii L, Verschoren S, Hölbling BV, Moreira FC, Katona E, Coneys R, de Oliveira P, Zhang YJ, Jansen K, Daughrity LM, McGown A, Ramesh TM, Van Den Bosch L, Lignani G, Rahim AA, Coyne AN, Petrucelli L, Rihel J, Isaacs AM. Dual-targeting CRISPR-CasRx reduces C9orf72 ALS/FTD sense and antisense repeat RNAs in vitro and in vivo. Nat Commun 2025; 16:459. [PMID: 39779704 PMCID: PMC11711508 DOI: 10.1038/s41467-024-55550-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The most common genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) is an intronic G4C2 repeat expansion in C9orf72. The repeats undergo bidirectional transcription to produce sense and antisense repeat RNA species, which are translated into dipeptide repeat proteins (DPRs). As toxicity has been associated with both sense and antisense repeat-derived RNA and DPRs, targeting both strands may provide the most effective therapeutic strategy. CRISPR-Cas13 systems mature their own guide arrays, allowing targeting of multiple RNA species from a single construct. We show CRISPR-Cas13d variant CasRx effectively reduces overexpressed C9orf72 sense and antisense repeat transcripts and DPRs in HEK cells. In C9orf72 patient-derived iPSC-neuron lines, CRISPR-CasRx reduces endogenous sense and antisense repeat RNAs and DPRs and protects against glutamate-induced excitotoxicity. AAV delivery of CRISPR-CasRx to two distinct C9orf72 repeat mouse models significantly reduced both sense and antisense repeat-containing transcripts. This highlights the potential of RNA-targeting CRISPR systems as therapeutics for C9orf72 ALS/FTD.
Collapse
Affiliation(s)
- Liam Kempthorne
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Deniz Vaizoglu
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alexander J Cammack
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Martha J Roberts
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alla Mikheenko
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alessia Fisher
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Pacharaporn Suklai
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Bhavana Muralidharan
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, 560065, India
| | - François Kroll
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Thomas G Moens
- VIB-KU Center for Brain and Disease Research, Leuven, 3001, Belgium
| | - Lidia Yshii
- VIB-KU Center for Brain and Disease Research, Leuven, 3001, Belgium
| | - Stijn Verschoren
- VIB-KU Center for Brain and Disease Research, Leuven, 3001, Belgium
| | - Benedikt V Hölbling
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Francisco C Moreira
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Eszter Katona
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Rachel Coneys
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Paula de Oliveira
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Karen Jansen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Alexander McGown
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Tennore M Ramesh
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | | | - Gabriele Lignani
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alyssa N Coyne
- Department of Neurology, Johns Hopkins University, Baltimore, USA
- Brain Science Institute, Johns Hopkins University, Baltimore, USA
| | | | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
30
|
Wang K, Yin H, Li S, Wan Y, Xiao M, Yuan X, Huang Z, Gao Y, Zhou J, Guo K, Wang J. Quantitative detection of circular RNA and microRNA at point-of-care using droplet digital CRISPR/Cas13a platform. Biosens Bioelectron 2025; 267:116825. [PMID: 39369515 DOI: 10.1016/j.bios.2024.116825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/03/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Circular RNA (circRNA) and microRNA (miRNA) are both non-coding RNAs (ncRNAs) that serve as biomarkers for cancer diagnosis and prognosis. Quantitative detection of these ncRNAs is of particular importance to elucidate the functional mechanisms and evaluate their potential as biomarkers. However, the inherent structures of circRNA and miRNA are different from the mRNA, conventional qRT-PCR is unsuitable for the detection of these ncRNAs. Here, we propose a sensitive method for quantitative detection of circRNA and miRNA using polydisperse droplet digital CRISPR/Cas13a (PddCas13a). It can achieve limits of detection (LOD) as low as ∼10 aM without polymerase-based amplification. To efficiently detect the circRNA and miRNA in real samples, we use a chemically modified crRNA to enhance the stability of crRNA and improve the performance of Cas13a in complex environments containing contaminants. By integrating an extraction-free procedure with PddCas13a, we experimentally demonstrate the applicability of PddCas13a by testing clinical samples. Furthermore, we develop an automated and portable instrument for PddCas13a and verify its applicability for the detection of circRNA and miRNA from exosomes in point-of-care (POC) setting. This is the first report to detect the circRNA and miRNA simultaneously in POC setting. We envision this platform could promote the research of ncRNAs.
Collapse
Affiliation(s)
- Ke Wang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Haofan Yin
- Department of Medical Laboratory, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Sheng Li
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yunzhu Wan
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Minmin Xiao
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaopeng Yuan
- Department of Medical Laboratory, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Zhenheng Huang
- R & D Department, Guangdong Forevergen Medical Technology Co., Ltd, Foshan, China
| | - Yunxin Gao
- R & D Department, Guangdong Forevergen Medical Technology Co., Ltd, Foshan, China
| | - Jianhua Zhou
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Keying Guo
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, Parkville, VIC, 3052, Australia.
| | - Jiasi Wang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
31
|
Yazdi ZF, Roshannezhad S, Sharif S, Abbaszadegan MR. Recent progress in prompt molecular detection of liquid biopsy using Cas enzymes: innovative approaches for cancer diagnosis and analysis. J Transl Med 2024; 22:1173. [PMID: 39741289 DOI: 10.1186/s12967-024-05908-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
Creating fast, non-invasive, precise, and specific diagnostic tests is crucial for enhancing cancer treatment outcomes. Among diagnostic methods, those relying on nucleic acid detection are highly sensitive and specific. Recent developments in diagnostic technologies, particularly those leveraging Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR), are revolutionizing cancer detection, providing accurate and timely results. In clinical oncology, liquid biopsy has become a noninvasive and early-detectable alternative to traditional biopsies over the last two decades. Analyzing the nucleic acid content of liquid biopsy samples, which include Circulating Tumor Cells (CTCs), Circulating Tumor DNA (ctDNA), Circulating Cell-Free RNA (cfRNA), and tumor extracellular vesicles, provides a noninvasive method for cancer detection and monitoring. In this review, we explore how the characteristics of various Cas (CRISPR-associated) enzymes have been utilized in diagnostic assays for cancer liquid biopsy and highlight their main applications of innovative approaches in monitoring, as well as early and rapid detection of cancers.
Collapse
Affiliation(s)
- Zahra Farshchian Yazdi
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Samaneh Sharif
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Mashhad University of Medical Sciences, Azadi Square, Mashhad, Iran.
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Mashhad University of Medical Sciences, Azadi Square, Mashhad, Iran.
| |
Collapse
|
32
|
Aguilar R, Mardones C, Moreno AA, Cepeda-Plaza M. A guide to RNA structure analysis and RNA-targeting methods. FEBS J 2024. [PMID: 39718192 DOI: 10.1111/febs.17368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
RNAs are increasingly recognized as promising therapeutic targets, susceptible to modulation by strategies that include targeting with small molecules, antisense oligonucleotides, deoxyribozymes (DNAzymes), or CRISPR/Cas13. However, while drug development for proteins follows well-established paths for rational design based on the accurate knowledge of their three-dimensional structure, RNA-targeting strategies are challenging since comprehensive RNA structures are yet scarce and challenging to acquire. Numerous methods have been developed to elucidate the secondary and three-dimensional structure of RNAs, including X-ray crystallography, cryo-electron microscopy, nuclear magnetic resonance, SHAPE, DMS, and bioinformatic methods, yet they have often revealed flexible transcripts and co-existing populations rather than single-defined structures. Thus, researchers aiming to target RNAs face a critical decision: whether to acquire the detailed structure of transcripts in advance or to adopt phenotypic screens or sequence-based approaches that are independent of the structure. Still, even in strategies that seem to rely only on the nucleotide sequence (like the design of antisense oligonucleotides), researchers may need information about the accessibility of the compounds to the folded RNA molecule. In this concise guide, we provide an overview for researchers interested in targeting RNAs: We start by revisiting current methodologies for defining secondary or three-dimensional RNA structure and then we explore RNA-targeting strategies that may or may not require an in-depth knowledge of RNA structure. We envision that complementary approaches may expedite the development of RNA-targeting molecules to combat disease.
Collapse
Affiliation(s)
- Rodrigo Aguilar
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
| | - Constanza Mardones
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
| | - Adrian A Moreno
- Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | | |
Collapse
|
33
|
Zhang X, Huang Z, Zhang Y, Wang W, Ye Z, Liang P, Sun K, Kang W, Tang Q, Yu X. Mitigating Antibiotic Resistance: The Utilization of CRISPR Technology in Detection. BIOSENSORS 2024; 14:633. [PMID: 39727898 DOI: 10.3390/bios14120633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Antibiotics, celebrated as some of the most significant pharmaceutical breakthroughs in medical history, are capable of eliminating or inhibiting bacterial growth, offering a primary defense against a wide array of bacterial infections. However, the rise in antimicrobial resistance (AMR), driven by the widespread use of antibiotics, has evolved into a widespread and ominous threat to global public health. Thus, the creation of efficient methods for detecting resistance genes and antibiotics is imperative for ensuring food safety and safeguarding human health. The clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (Cas) systems, initially recognized as an adaptive immune defense mechanism in bacteria and archaea, have unveiled their profound potential in sensor detection, transcending their notable gene-editing applications. CRISPR/Cas technology employs Cas enzymes and guides RNA to selectively target and cleave specific DNA or RNA sequences. This review offers an extensive examination of CRISPR/Cas systems, highlighting their unique attributes and applications in antibiotic detection. It outlines the current utilization and progress of the CRISPR/Cas toolkit for identifying both nucleic acid (resistance genes) and non-nucleic acid (antibiotic micromolecules) targets within the field of antibiotic detection. In addition, it examines the current challenges, such as sensitivity and specificity, and future opportunities, including the development of point-of-care diagnostics, providing strategic insights to facilitate the curbing and oversight of antibiotic-resistance proliferation.
Collapse
Affiliation(s)
- Xuejiao Zhang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Zhaojie Huang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Yanxia Zhang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Wen Wang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Zihong Ye
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Pei Liang
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, China
| | - Kai Sun
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Wencheng Kang
- Inner Mongolia Institute of Metrology and Testing, Hohhot 010030, China
| | - Qiao Tang
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Xiaoping Yu
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Science, China Jiliang University, Hangzhou 310018, China
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, China Jiliang University, Hangzhou 310018, China
| |
Collapse
|
34
|
Lamb CH, Pitré EM, Ajufo S, Rigby CV, Bisht K, Oade MS, Jalal H, Myhrvold C, Te Velthuis AJW. Quantification of influenza virus mini viral RNAs using Cas13. RNA (NEW YORK, N.Y.) 2024; 31:126-138. [PMID: 39419543 PMCID: PMC11648933 DOI: 10.1261/rna.080174.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
Influenza A virus (IAV) RNA synthesis produces full-length and deletion-containing RNA molecules, which include defective viral genomes (DVG) and mini viral RNAs (mvRNA). Sequencing approaches have shown that DVG and mvRNA species may be present during infection, and that they can vary in size, segment origin, and sequence. Moreover, a subset of aberrant RNA molecules can bind and activate host-pathogen receptor retinoic acid-inducible gene I (RIG-I), leading to innate immune signaling and the expression of type I and III interferons. Measuring the kinetics and distribution of these immunostimulatory aberrant RNA sequences is important for understanding their function in IAV infection. Here, we explored if IAV mvRNA molecules can be detected and quantified using amplification-free, CRISPR-LbuCas13a-based detection. We show that CRISPR-LbuCas13a can be used to measure the copy numbers of specific mvRNAs in samples from infected tissue culture cells. However, to efficiently detect mvRNAs in other samples, promiscuous CRISPR guide RNAs are required that activate LbuCas13a in the presence of multiple mvRNA sequences. One crRNA was able to detect full-length IAV segment 5 without amplification, allowing it to be used for general IAV infection detection nasopharyngeal swabs. Using CRISPR-LbuCas13a, we confirm that mvRNAs are present in ferret upper and lower respiratory tract tissue, as well as clinical nasopharyngeal swab extracts of hospitalized patients. Overall, CRISPR-LbuCas13a-based RNA detection is a useful tool for studying deletion-containing viral RNAs, and it complements existing amplification-based approaches.
Collapse
Affiliation(s)
- Caitlin H Lamb
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Emmanuelle M Pitré
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- University of Cambridge, Department of Pathology, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
| | - Sean Ajufo
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Charlotte V Rigby
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- University of Cambridge, Department of Pathology, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
- Public Health England, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
| | - Karishma Bisht
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Michael S Oade
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Hamid Jalal
- Public Health England, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, New Jersey 08544, USA
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, USA
| | | |
Collapse
|
35
|
Molina-Sánchez MD, Martínez-Abarca F, Millán V, Mestre MR, Stehantsev P, Stetsenko A, Guskov A, Toro N. Adaptive immunity of type VI CRISPR-Cas systems associated with reverse transcriptase-Cas1 fusion proteins. Nucleic Acids Res 2024; 52:14229-14243. [PMID: 39673266 DOI: 10.1093/nar/gkae1154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 12/16/2024] Open
Abstract
Cas13-containing type VI CRISPR-Cas systems specifically target RNA; however, the mechanism of spacer acquisition remains unclear. We have previously reported the association of reverse transcriptase-Cas1 (RT-Cas1) fusion proteins with certain types of VI-A systems. Here, we show that RT-Cas1 fusion proteins are also recruited by type VI-B systems in bacteria from gut microbiomes, constituting a VI-B1 variant system that includes a CorA-encoding locus in addition to the CRISPR array and the RT-Cas1/Cas2 adaptation module. We found that type VI RT-CRISPR systems were functional for spacer acquisition, CRISPR array processing and interference activity, demonstrating that adaptive immunity mediated by these systems can function independently of other in trans systems. We provide evidence that the RT associated with these systems enables spacer acquisition from RNA molecules. We also found that CorA encoded by type VI-B1 RT-associated systems can transport divalent metal ions and downregulate Cas13b-mediated RNA interference. These findings highlight the importance of RTs in RNA-targeting CRISPR-Cas systems, potentially enabling the integration of RNA-derived spacers into CRISPR arrays as a mechanism against RNA-based invaders in specific environments.
Collapse
Affiliation(s)
- María Dolores Molina-Sánchez
- Department of Soil and Plant Microbiology, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Structure, Dynamics and Function of Bacterial Genomes, Grupo de Ecología Genética de la Rizosfera, C/Profesor Albareda 1, 18008 Granada, Spain
| | - Francisco Martínez-Abarca
- Department of Soil and Plant Microbiology, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Structure, Dynamics and Function of Bacterial Genomes, Grupo de Ecología Genética de la Rizosfera, C/Profesor Albareda 1, 18008 Granada, Spain
| | - Vicenta Millán
- Department of Soil and Plant Microbiology, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Structure, Dynamics and Function of Bacterial Genomes, Grupo de Ecología Genética de la Rizosfera, C/Profesor Albareda 1, 18008 Granada, Spain
| | - Mario Rodríguez Mestre
- Department of Soil and Plant Microbiology, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Structure, Dynamics and Function of Bacterial Genomes, Grupo de Ecología Genética de la Rizosfera, C/Profesor Albareda 1, 18008 Granada, Spain
- Section of Microbiology, Department of Biology, University of Copenhagen, Universitetsparken 15, 2100 Copenhagen, Denmark
| | - Pavlo Stehantsev
- Groningen Biomolecular & Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Artem Stetsenko
- Groningen Biomolecular & Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Albert Guskov
- Groningen Biomolecular & Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Nicolás Toro
- Department of Soil and Plant Microbiology, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Structure, Dynamics and Function of Bacterial Genomes, Grupo de Ecología Genética de la Rizosfera, C/Profesor Albareda 1, 18008 Granada, Spain
| |
Collapse
|
36
|
Yu L, Zou J, Hussain A, Jia R, Fan Y, Liu J, Nie X, Zhang X, Jin S. Systemic evaluation of various CRISPR/Cas13 orthologs for knockdown of targeted transcripts in plants. Genome Biol 2024; 25:307. [PMID: 39639368 PMCID: PMC11619151 DOI: 10.1186/s13059-024-03448-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND CRISPR/Cas13 system, recognized for its compact size and specificity in targeting RNA, is currently employed for RNA degradation. However, the potential of various CRISPR/Cas13 subtypes, particularly concerning the knockdown of endogenous transcripts, remains to be comprehensively characterized in plants. RESULTS Here we present a full spectrum of editing profiles for seven Cas13 orthologs from five distinct subtypes: VI-A (LwaCas13a), VI-B (PbuCas13b), VI-D (RfxCas13d), VI-X (Cas13x.1 and Cas13x.2), and VI-Y (Cas13y.1 and Cas13y.2). A systematic evaluation of the knockdown effects on two endogenous transcripts (GhCLA and GhPGF in cotton) as well as an RNA virus (TMV in tobacco) reveals that RfxCas13d, Cas13x.1, and Cas13x.2 exhibit enhanced stability with editing efficiencies ranging from 58 to 80%, closely followed by Cas13y.1 and Cas13y.2. Notably, both Cas13x.1 and Cas13y.1 can simultaneously degrade two endogenous transcripts through a tRNA-crRNA cassette approach, achieving editing efficiencies of up to 50%. Furthermore, different Cas13 orthologs enable varying degrees of endogenous transcript knockdown with minimal off-target effects, generating germplasms that exhibit a diverse spectrum of mutant phenotypes. Transgenic tobacco plants show significant reductions in damage, along with mild oxidative stress and minimal accumulation of viral particles after TMV infection. CONCLUSIONS In conclusion, our study presents an efficient and reliable platform for transcriptome editing that holds promise for plant functional research and future crop improvement.
Collapse
Affiliation(s)
- Lu Yu
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiawei Zou
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Amjad Hussain
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ruoyu Jia
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yibo Fan
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jinhang Liu
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xinhui Nie
- Key Laboratory of Oasis Ecology Agricultural of Xinjiang Production and Construction Corps, Agricultural College, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Xianlong Zhang
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuangxia Jin
- Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
37
|
Zhang X, Bhattacharya A, Pu C, Dai Y, Liu J, Rao L, Tian C. A programmable CRISPR/dCas9-based epigenetic editing system enabling loci-targeted histone citrullination and precise transcription regulation. J Genet Genomics 2024; 51:1485-1493. [PMID: 38849111 DOI: 10.1016/j.jgg.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024]
Abstract
Histone citrullination, an important post-translational modification mediated by peptidyl arginine deiminases, is essential for many physiological processes and epigenetic regulation. However, the causal relationship between histone citrullination and specific gene regulation remains unresolved. In this study, we develop a programmable epigenetic editor by fusing the peptidyl arginine deiminase (PAD) PPAD from Porphyromonas gingivalis with dCas9. With the assistance of gRNA, PPAD-dCas9 can recruit PPADs to specific genomic loci, enabling direct manipulation of the epigenetic landscape and regulation of gene expression. Our citrullination editor allows for the site-specific manipulation of histone H3R2,8,17 and H3R26 at target human gene loci, resulting in the activation or suppression of different genes in a locus-specific manner. Moreover, the epigenetic effects of the citrullination editor are specific and sustained. This epigenetic editor offers an accurate and efficient tool for exploring gene regulation of histone citrullination.
Collapse
Affiliation(s)
- Xiaoya Zhang
- National Technology Innovation Center of Synthetic Biology, Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; School of Pharmacy, Jilin University, Changchun, Jilin 130012, China
| | - Abhisek Bhattacharya
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chunxiang Pu
- National Technology Innovation Center of Synthetic Biology, Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Yan Dai
- National Technology Innovation Center of Synthetic Biology, Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Jia Liu
- National Technology Innovation Center of Synthetic Biology, Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Lang Rao
- National Technology Innovation Center of Synthetic Biology, Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
| | - Chaoguang Tian
- National Technology Innovation Center of Synthetic Biology, Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
| |
Collapse
|
38
|
Wang Y, He D, Li W, Dong Y, Fang L, Liu D, Tang Y, Xiao S. Field-deployable porcine epidemic diarrhea virus diagnostics utilizing CRISPR-Cas13a. Virulence 2024; 15:2429022. [PMID: 39560197 PMCID: PMC11581157 DOI: 10.1080/21505594.2024.2429022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 09/04/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
Porcine epidemic diarrhoea virus (PEDV), a pathogenic microorganism that induces epidemic diarrhoea in swine, causes substantial economic damage to swine-farming nations. To prevent and control PEDV infections, the availability of upgraded and rapid virus detection techniques is crucial. The clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein (Cas)13a system, namely, programmability of CRISPR RNA (crRNA) and "collateral" promiscuous RNase activity of Cas13a after target RNA identification. In this study, we aimed to develop a recombinase polymerase amplification (RPA)-based CRISPR-Cas13a approach for PEDV diagnosis for the first time. The results showed that up to 10 copies of the target PEDV DNA standard/µL were detected after 40 min at 37 °C. PEDV detection exhibited remarkable specificity compared to that of other selected pathogens. Additionally, this RPA-based CRISPR-Cas13a approach could be used to clinical samples, with similar performance to that of reverse transcription-quantitative polymerase chain reaction (RT - qPCR). The results of our proposed approach were visualized using either lateral flow strips or fluorescence for field-deployable viral diagnostics, thereby facilitating its use in endemic regions. Overall, our proposed approach showed good reliability, sensitivity, and specificity, suggesting that it is applicable for detecting other viruses in diagnosing diseases and inspecting food safety.
Collapse
Affiliation(s)
- Yuanyuan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Animal Health Standards and Regulation, China Animal Health and Epidemiology Center, Qingdao 266000, Shandong Province,China
| | - Dalin He
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Weihua Li
- Department of Animal Health Standards and Regulation, China Animal Health and Epidemiology Center, Qingdao 266000, Shandong Province,China
| | - Yaqin Dong
- Department of Animal Health Standards and Regulation, China Animal Health and Epidemiology Center, Qingdao 266000, Shandong Province,China
| | - Linlin Fang
- Department of Animal Health Standards and Regulation, China Animal Health and Epidemiology Center, Qingdao 266000, Shandong Province,China
| | - Deju Liu
- Department of Animal Health Standards and Regulation, China Animal Health and Epidemiology Center, Qingdao 266000, Shandong Province,China
| | - Yi Tang
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Shaobo Xiao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
39
|
Lei R, Liu XP. Rapid one-pot human single nucleotide polymorphism genotyping platform with Cas13a nuclease. J Biosci Bioeng 2024; 138:469-477. [PMID: 39304484 DOI: 10.1016/j.jbiosc.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/12/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
Single nucleotide polymorphism (SNP), as one of the key components of the genetic factors, is important for disease detection and early screening of hereditary diseases. Current SNP genotyping methods require laboratory instruments or long operating times. To facilitate the diagnosis of hereditary diseases, we developed a new method referred to as the LwaCas13a-based SNP genotyping platform (Cas13a platform), which is useful for detecting disease-related SNPs. We report a CRISPR/Cas13a-based SNP genotyping platform that couples recombinase-aided amplification (RAA), T7 transcription, and Leptotrichia wadei Cas13a (LwaCas13a) detection for simple and fast genotyping of human disease-related SNPs. We used this Cas13a platform to identify 17 disease-related SNPs, demonstrating that position 2 in gRNA is suitable for the introduction of additional mismatches to achieve high discrimination in genotyping across a wide range of SNP targets. The discrimination specificity of 17 SNPs was improved 3.0-35.1-fold after introducing additional mismatches at position 2 from the 5'-end. We developed a method, which has a lower risk of cross-contamination and operational complexity, for genotyping SNPs using human saliva samples in an one-pot testing that delivers results within 60 min. Compared to TaqMan probe qPCR, RFLP, AS-PCR and other SNP genotyping methods, the Cas13a platform is simple, rapid and reliable, expanding the applications of the CRISPR/Cas system in nucleic acid detection and SNP genotyping.
Collapse
Affiliation(s)
- Rui Lei
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong-Chuan Road, Shanghai 200240, China
| | - Xi-Peng Liu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong-Chuan Road, Shanghai 200240, China; SJTU Yazhou Bay Institute of Deepsea Sci-Tech, Yongyou Industrial Park, Sanya 572024, China; Joint International Research Laboratory of Metabolic & Developmental Sciences (Ministry of Education), Shanghai Jiao Tong University, 800 Dong-Chuan Road, Shanghai 200240, China; State Key Laboratory of Ocean Engineering, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, 800 Dong-Chuan Road, Shanghai 200240, China.
| |
Collapse
|
40
|
Zhan X, Zhang F, Li N, Xu K, Wang X, Gao S, Yin Y, Yuan W, Chen W, Ren Z, Yao M, Wang F. CRISPR/Cas: An Emerging Toolbox for Engineering Virus Resistance in Plants. PLANTS (BASEL, SWITZERLAND) 2024; 13:3313. [PMID: 39683106 DOI: 10.3390/plants13233313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas have been recognized as powerful genome-editing tools in diverse eukaryotic species, including plants, and thus hold great promise for engineering virus resistance in plants. Nevertheless, further attention is required regarding various issues associated with applying new powerful technologies in the field. This mini-review focuses on the recent advances in using CRISPR/Cas9 and CRISPR/Cas13 systems to combat DNA and RNA viruses in plants. We explored the utility of CRISPR/Cas for targeting the viral genome and editing host susceptibility genes in plants. We also provide insights into the limitations and challenges of using CRISPR/Cas for plant virus interference and propose individual combinatorial solutions. In conclusion, CRISPR/Cas technology has the potential to offer innovative and highly efficient approaches for controlling viruses in important crops in the near future.
Collapse
Affiliation(s)
- Xiaohui Zhan
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Fengjuan Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Ning Li
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Kai Xu
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Xiaodi Wang
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Shenghua Gao
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Yanxu Yin
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Weiling Yuan
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Weifang Chen
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Zhiyong Ren
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
| | - Minghua Yao
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Fei Wang
- Hubei Key Laboratory of Vegetable Germplasm Innovation and Genetic Improvement, Cash Crops Research Institute, Hubei Academy of Agricultural Sciences, Wuhan 430062, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
| |
Collapse
|
41
|
Huang S, Du L, Liu S, Yang Q, Lei C, Wang H, Yang L, Yang X. Development and Validation of RAA-CRISPR/Cas12a-Based Assay for Detecting Porcine Rotavirus. Animals (Basel) 2024; 14:3387. [PMID: 39682353 DOI: 10.3390/ani14233387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Piglet diarrhea poses significant economic losses to the pig industry, posing a worldwide challenge that urgently needs to be addressed in pig breeding practices. Porcine rotavirus (PoRV) is an important viral diarrhea pathogen in piglets, with a high incidence rate and a tendency to cause growth retardation. To enhance the sensitivity and specificity of PoRV detection, we sequenced the NSP3 gene of G5 and G9 genotypes of rotavirus A (RVA), enabling simultaneous detection of the two serotypes. Subsequently, we developed a rapid PoRV detection method using a combination of recombinase-aided amplification (RAA) and CRISPR/Cas12a. In this method, Cas12a binds to RAA amplification products, guided by CRISPR-derived RNA (crRNA), which activates its cleavage activity and releases fluorescence by cutting FAM-BHQ-labeled single-stranded DNA (ssDNA). In the optimized reaction system, the recombinant plasmid PoRV can achieve a highly sensitive reaction within 30 min at 37 °C, with a detection limit as low as 2.43 copies/μL, which is ten times higher in sensitivity compared to the qPCR method. Results from specificity testing indicate that no cross-reactivity was observed between the RAA-CRISPR/Cas12a analysis of PoRV and other viral pathogens, including PoRV G3, PoRV G4, porcine epidemic diarrhea virus (PEDV), porcine epidemic diarrhea (PDCoV), and porcine reproductive and respiratory syndrome virus (PRRSV). In the clinical sample detection using the RAA-CRISPR/Cas12a method and qPCR, Cohen's Kappa value reached as high as 0.952. Furthermore, this approach eliminates the need for large-scale instrumentation, offering a visual result under an ultraviolet lamp through fluorescence signal output.
Collapse
Affiliation(s)
- Siyu Huang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Longhuan Du
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Song Liu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Qingcheng Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Changwei Lei
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Liu Yang
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Xin Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
42
|
Li W, Jiang X, Wang W, Hou L, Cai R, Li Y, Gu Q, Chen Q, Ma P, Tang J, Guo M, Chuai G, Huang X, Zhang J, Liu Q. Discovering CRISPR-Cas system with self-processing pre-crRNA capability by foundation models. Nat Commun 2024; 15:10024. [PMID: 39562558 PMCID: PMC11576732 DOI: 10.1038/s41467-024-54365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
The discovery of CRISPR-Cas systems has paved the way for advanced gene editing tools. However, traditional Cas discovery methods relying on sequence similarity may miss distant homologs and aren't suitable for functional recognition. With protein large language models (LLMs) evolving, there is potential for Cas system modeling without extensive training data. Here, we introduce CHOOSER (Cas HOmlog Observing and SElf-processing scReening), an AI framework for alignment-free discovery of CRISPR-Cas systems with self-processing pre-crRNA capability using protein foundation models. By using CHOOSER, we identify 11 Casλ homologs, nearly doubling the known catalog. Notably, one homolog, EphcCasλ, is experimentally validated for self-processing pre-crRNA, DNA cleavage, and trans-cleavage, showing promise for CRISPR-based pathogen detection. This study highlights an innovative approach for discovering CRISPR-Cas systems with specific functions, emphasizing their potential in gene editing.
Collapse
Affiliation(s)
- Wenhui Li
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Xianyue Jiang
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Wuke Wang
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Liya Hou
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Runze Cai
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Yongqian Li
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Qiuxi Gu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | - Qinchang Chen
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Tang
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Menghao Guo
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Guohui Chuai
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- National Key Laboratory of Autonomous Intelligent Unmanned Systems, Frontiers Science Center for Intelligent Autonomous Systems, Ministry of Education, Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, China.
| | - Xingxu Huang
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang, China.
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China.
| | - Qi Liu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- National Key Laboratory of Autonomous Intelligent Unmanned Systems, Frontiers Science Center for Intelligent Autonomous Systems, Ministry of Education, Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, China.
| |
Collapse
|
43
|
Guermonprez P, Nioche P, Renaud L, Battaglini N, Sanaur S, Krejci E, Piro B. CRISPR-Cas Systems Associated with Electrolyte-Gated Graphene-Based Transistors: How They Work and How to Combine Them. BIOSENSORS 2024; 14:541. [PMID: 39590000 PMCID: PMC11592214 DOI: 10.3390/bios14110541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/27/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
In this review, recent advances in the combination of CRISPR-Cas systems with graphene-based electrolyte-gated transistors are discussed in detail. In the first part, the functioning of CRISPR-Cas systems is briefly explained, as well as the most common ways to convert their molecular activity into measurable signals. Other than optical means, conventional electrochemical transducers are also developed. However, it seems that the incorporation of CRISPR/Cas systems into transistor devices could be extremely powerful, as the former provides molecular amplification, while the latter provides electrical amplification; combined, the two could help to advance in terms of sensitivity and compete with conventional PCR assays. Today, organic transistors suffer from poor stability in biological media, whereas graphene materials perform better by being extremely sensitive to their chemical environment and being stable. The need for fast and inexpensive sensors to detect viral RNA arose on the occasion of the COVID-19 crisis, but many other RNA viruses are of interest, such as dengue, hepatitis C, hepatitis E, West Nile fever, Ebola, and polio, for which detection means are needed.
Collapse
Affiliation(s)
- Pierre Guermonprez
- ITODYS, CNRS, Université Paris Cité, F-75006 Paris, France; (P.G.); (N.B.)
| | - Pierre Nioche
- INSERM US 36|CNRS UAR 2009, Structural and Molecular Analysis Platform, Université Paris Cité, F-75006 Paris, France;
- INSERM U1124, Université Paris Cité, F-75006 Paris, France
| | - Louis Renaud
- Institut des Nanotechnologies de Lyon INL-UMR5270, Université Lyon 1, F-69622 Villeurbanne, France;
| | - Nicolas Battaglini
- ITODYS, CNRS, Université Paris Cité, F-75006 Paris, France; (P.G.); (N.B.)
| | - Sébastien Sanaur
- Department of Flexible Electronics, Institut Mines-Telecom, Mines Saint-Étienne, F-13541 Gardanne, France;
| | - Eric Krejci
- CNRS, ENS Paris Saclay, Centre Borelli UMR 9010, Université Paris Cité, F-75006 Paris, France;
| | - Benoît Piro
- ITODYS, CNRS, Université Paris Cité, F-75006 Paris, France; (P.G.); (N.B.)
| |
Collapse
|
44
|
Zhang L, Cao SM, Wu H, Yan M, Li J, Chen LL. A CRISPR/RfxCas13d-mediated strategy for efficient RNA knockdown in mouse embryonic development. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2297-2306. [PMID: 39110403 DOI: 10.1007/s11427-023-2572-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 10/22/2024]
Abstract
The growing variety of RNA classes, such as mRNAs, lncRNAs, and circRNAs, plays pivotal roles in both developmental processes and various pathophysiological conditions. Nonetheless, our comprehension of RNA functions in live organisms remains limited due to the absence of durable and effective strategies for directly influencing RNA levels. In this study, we combined the CRISPR-RfxCas13d system with sperm-like stem cell-mediated semi-cloning techniques, which enabled the suppressed expression of different RNA species. This approach was employed to interfere with the expression of three types of RNA molecules: Sfmbt2 mRNA, Fendrr lncRNA, and circMan1a2(2,3,4,5,6). The results confirmed the critical roles of these RNAs in embryonic development, as their loss led to observable phenotypes, including embryonic lethality, delayed embryonic development, and embryo resorption. In summary, our methodology offers a potent toolkit for silencing specific RNA targets in living organisms without introducing genetic alterations.
Collapse
Affiliation(s)
- Lin Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shi-Meng Cao
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hao Wu
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Meng Yan
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Ling-Ling Chen
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- New Cornerstone Science Laboratory, Shenzhen, 518054, China.
| |
Collapse
|
45
|
Hu W, Kumar A, Ahmed SF, Qi S, Ma DKG, Chen H, Singh GJ, Casan JML, Haber M, Voskoboinik I, McKay MR, Trapani JA, Ekert PG, Fareh M. Single-base tiled screen unveils design principles of PspCas13b for potent and off-target-free RNA silencing. Nat Struct Mol Biol 2024; 31:1702-1716. [PMID: 38951623 PMCID: PMC11564092 DOI: 10.1038/s41594-024-01336-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/15/2024] [Indexed: 07/03/2024]
Abstract
The development of precise RNA-editing tools is essential for the advancement of RNA therapeutics. CRISPR (clustered regularly interspaced short palindromic repeats) PspCas13b is a programmable RNA nuclease predicted to offer superior specificity because of its 30-nucleotide spacer sequence. However, its design principles and its on-target, off-target and collateral activities remain poorly characterized. Here, we present single-base tiled screening and computational analyses that identify key design principles for potent and highly selective RNA recognition and cleavage in human cells. We show that the de novo design of spacers containing guanosine bases at precise positions can greatly enhance the catalytic activity of inefficient CRISPR RNAs (crRNAs). These validated design principles (integrated into an online tool, https://cas13target.azurewebsites.net/ ) can predict highly effective crRNAs with ~90% accuracy. Furthermore, the comprehensive spacer-target mutagenesis revealed that PspCas13b can tolerate only up to four mismatches and requires ~26-nucleotide base pairing with the target to activate its nuclease domains, highlighting its superior specificity compared to other RNA or DNA interference tools. On the basis of this targeting resolution, we predict an extremely low probability of PspCas13b having off-target effects on other cellular transcripts. Proteomic analysis validated this prediction and showed that, unlike other Cas13 orthologs, PspCas13b exhibits potent on-target activity and lacks collateral effects.
Collapse
Affiliation(s)
- Wenxin Hu
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amit Kumar
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Diagnostic Genomics, Monash Health Pathology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Syed Faraz Ahmed
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shijiao Qi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - David K G Ma
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Honglin Chen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Gurjeet J Singh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joshua M L Casan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ilia Voskoboinik
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Matthew R McKay
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul G Ekert
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Mohamed Fareh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
46
|
Etemadzadeh A, Salehipour P, Motlagh FM, Khalifeh M, Asadbeigi A, Tabrizi M, Shirkouhi R, Modarressi MH. An Optimized CRISPR/Cas12a Assay to Facilitate the BRAF V600E Mutation Detection. J Clin Lab Anal 2024; 38:e25101. [PMID: 39445676 PMCID: PMC11555610 DOI: 10.1002/jcla.25101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/20/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Accurate detection of the BRAF V600E (1799T > A) mutation status can significantly contribute to selecting an optimal therapeutic strategy for diverse cancer types. CRISPR-based diagnostic platforms exhibit simple programming, cost-effectiveness, high sensitivity, and high specificity in detecting target sequences. The goal of this study is to develop a simple BRAF V600E mutation detection method. METHODS We combined the CRISPR/Cas12a system with recombinase polymerase amplification (RPA). Subsequently, several parameters related to CRISPR/Cas12a reaction efficiency were evaluated. Then, we conducted a comparative analysis of three distinct approaches toward identifying BRAF V600E mutations in the clinical samples. RESULTS Our data suggest that CRISPR/Cas detection is considerably responsive to variations in buffer conditions. Magnesium acetate (MgOAc) demonstrated superior performance compared to all other examined additive salts. It was observed using 150 nM guide RNA (gRNA) in an optimized reaction buffer containing 14 mM MgOAc, coupled with a reduction in the volumes of PCR and RPA products to 1 μL and 3 μL, respectively, resulted in an enhanced sensitivity. Detection time was decreased to 75 min with a 2% limit of detection (LOD), as evidenced by the results obtained from the blue light illuminator. The CRISPR/Cas12a assay confirmed the real-time PCR results in 31 of 32 clinical samples to identify the BRAF V600E mutation status, while Sanger sequencing detected BRAF V600E mutations with lower sensitivity. CONCLUSION We propose a potential diagnostic approach that is facile, fast, and affordable with high fidelity. This method can detect BRAF V600E mutation with a 2% LOD without the need for a thermocycler.
Collapse
Affiliation(s)
- Azadeh Etemadzadeh
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Pouya Salehipour
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Fatemeh Movahedi Motlagh
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Masoomeh Khalifeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Adnan Asadbeigi
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Mina Tabrizi
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
- Department of Molecular & Medical GeneticsOregon Health & Science UniversityPortlandOregonUSA
- Knight Diagnostic LaboratoriesOregon Health & Science UniversityPortlandOregonUSA
| | - Reza Shirkouhi
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | | |
Collapse
|
47
|
Zheng W, Tang H, Ye B, Lin J, Wang H, Liu Y, Wang D, Wu Z, Xie W, Dong WF, Zan M. Fast, portable and sensitive detection of group B streptococcus DNA using one-pot MIRA-CRISPR system with suboptimal PAM. Talanta 2024; 279:126574. [PMID: 39029179 DOI: 10.1016/j.talanta.2024.126574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
The group B Streptococcus (GBS) can generate vertical transmission to infants during delivery, has been seriously threatening the health of infants. Rapid and accurate prenatal GBS diagnosis for pregnant women is a deterministic blueprint to avoid infant viruses. Here, we developed an extraction-free nucleic acid isothermal amplification/CRISPR-Cas12a cutting one-pot system for GBS diagnostic assay by using suboptimal protospacer adjacent motifs, effectively avoiding multiple handling steps and uncapping contamination. The GBS diagnosis assay based on a one-pot system was validated by using fluorescent technique and lateral flow assay strips, exhibited fantastic specificity, accuracy and sensitivity with a limit of detection of 32 copies per reaction (0.64 copies/μL). Moreover, a portable device was constructed and integrated with the one-pot system to realize the GBS detection without professional and scene restrictions, it showed excellent performance in clinical sample detection, which achieved optical and portable GBS detection for point-of-care testing or home-self testing.
Collapse
Affiliation(s)
- Weigang Zheng
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, China; Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Huamei Tang
- Medical Laboratory of Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Benchen Ye
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Jiasheng Lin
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Huihui Wang
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Ying Liu
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, China; Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Dong Wang
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, China; Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Zaihui Wu
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, China; Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Wei Xie
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
| | - Wen-Fei Dong
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, China.
| | - Minghui Zan
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, China; Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China.
| |
Collapse
|
48
|
Núñez-Álvarez Y, Espie-Caullet T, Buhagiar G, Rubio-Zulaika A, Alonso-Marañón J, Luna-Pérez E, Blazquez L, Luco R. A CRISPR-dCas13 RNA-editing tool to study alternative splicing. Nucleic Acids Res 2024; 52:11926-11939. [PMID: 39162234 PMCID: PMC11514487 DOI: 10.1093/nar/gkae682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alternative splicing allows multiple transcripts to be generated from the same gene to diversify the protein repertoire and gain new functions despite a limited coding genome. It can impact a wide spectrum of biological processes, including disease. However, its significance has long been underestimated due to limitations in dissecting the precise role of each splicing isoform in a physiological context. Furthermore, identifying key regulatory elements to correct deleterious splicing isoforms has proven equally challenging, increasing the difficulty of tackling the role of alternative splicing in cell biology. In this work, we take advantage of dCasRx, a catalytically inactive RNA targeting CRISPR-dCas13 ortholog, to efficiently switch alternative splicing patterns of endogenous transcripts without affecting overall gene expression levels cost-effectively. Additionally, we demonstrate a new application for the dCasRx splice-editing system to identify key regulatory RNA elements of specific splicing events. With this approach, we are expanding the RNA toolkit to better understand the regulatory mechanisms underlying alternative splicing and its physiological impact in various biological processes, including pathological conditions.
Collapse
Affiliation(s)
- Yaiza Núñez-Álvarez
- Institut de Génétique Humaine, Université de Montpellier, CNRS UMR9002, Montpellier, France
| | - Tristan Espie-Caullet
- Institut de Génétique Humaine, Université de Montpellier, CNRS UMR9002, Montpellier, France
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| | - Géraldine Buhagiar
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| | - Ane Rubio-Zulaika
- Department of Neurosciences, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain
| | - Josune Alonso-Marañón
- Department of Neurosciences, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain
| | - Elvira Luna-Pérez
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| | - Lorea Blazquez
- Department of Neurosciences, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), 28031 Madrid, Spain
| | - Reini F Luco
- Institut de Génétique Humaine, Université de Montpellier, CNRS UMR9002, Montpellier, France
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| |
Collapse
|
49
|
Armbruster EG, Rani P, Lee J, Klusch N, Hutchings J, Hoffman LY, Buschkaemper H, Enustun E, Adler BA, Inlow K, VanderWal AR, Hoffman MY, Daksh D, Aindow A, Deep A, Rodriguez ZK, Morgan CJ, Ghassemian M, Laughlin TG, Charles E, Cress BF, Savage DF, Doudna JA, Pogliano K, Corbett KD, Villa E, Pogliano J. A transcriptionally active lipid vesicle encloses the injected Chimalliviridae genome in early infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.20.558163. [PMID: 37781618 PMCID: PMC10541120 DOI: 10.1101/2023.09.20.558163] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Many eukaryotic viruses require membrane-bound compartments for replication, but no such organelles are known to be formed by prokaryotic viruses1-3. Bacteriophages of the Chimalliviridae family sequester their genomes within a phage-generated organelle, the phage nucleus, which is enclosed by a lattice of the viral protein ChmA4-10. Previously, we observed lipid membrane-bound vesicles in cells infected by Chimalliviridae, but due to the paucity of genetics tools for these viruses it was unknown if these vesicles represented unproductive, abortive infections or a bona fide stage in the phage life cycle. Using the recently-developed dRfxCas13d-based knockdown system CRISPRi-ART11 in combination with fluorescence microscopy and cryo-electron tomography, we show that inhibiting phage nucleus formation arrests infections at an early stage in which the injected phage genome is enclosed within a membrane-bound early phage infection (EPI) vesicle. We demonstrate that early phage genes are transcribed by the virion-associated RNA polymerase from the genome within the compartment, making the EPI vesicle the first known example of a lipid membrane-bound organelle that separates transcription from translation in prokaryotes. Further, we show that the phage nucleus is essential for the phage life cycle, with genome replication only beginning after the injected DNA is transferred from the EPI vesicle to the newly assembled phage nucleus. Our results show that Chimalliviridae require two sophisticated subcellular compartments of distinct compositions and functions that facilitate successive stages of the viral life cycle.
Collapse
Affiliation(s)
- Emily G. Armbruster
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- These authors contributed equally: Emily G. Armbruster and Phoolwanti Rani
| | - Phoolwanti Rani
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
- These authors contributed equally: Emily G. Armbruster and Phoolwanti Rani
| | - Jina Lee
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Niklas Klusch
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Joshua Hutchings
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Lizbeth Y. Hoffman
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Hannah Buschkaemper
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Eray Enustun
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Benjamin A. Adler
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA
| | - Koe Inlow
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Arica R. VanderWal
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Madelynn Y. Hoffman
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Daksh Daksh
- National Institute of Science, Education and Research (NISER) Bhubaneshwar, Orissa 752050, India
| | - Ann Aindow
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Zaida K. Rodriguez
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Chase J. Morgan
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, La Jolla, CA 92093, USA
| | - Thomas G. Laughlin
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Emeric Charles
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Brady F. Cress
- Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA
| | - David F. Savage
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Jennifer A. Doudna
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Kit Pogliano
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Kevin D. Corbett
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Elizabeth Villa
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Joe Pogliano
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
50
|
Wu S, Yu W, Fu X, Yu X, Ye Z, Zhang M, Qiu Y, Ma B. Advances in Virus Detection Techniques Based on Recombinant Polymerase Amplification. Molecules 2024; 29:4972. [PMID: 39459340 PMCID: PMC11510534 DOI: 10.3390/molecules29204972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Recombinase polymerase amplification (RPA) has emerged as a rapid, efficient, and highly sensitive method for nucleic acid amplification, thus becoming a focal point of research in the field of virus detection. This paper provides an overview of RPA, emphasizing its unique double-stranded DNA synthesis mechanism, rapid amplification efficiency, and capability to operate at room temperature, among other advantages. In addition, strategies and case studies of RPA in combination with other technologies are detailed to explore the advantages and potential of these integrated approaches for virus detection. Finally, the development prospect of RPA technology is prospected.
Collapse
Affiliation(s)
| | | | - Xianshu Fu
- Key Laboratory of Microbiological Metrology, Measurement & Bio-product Quality Security, State Administration for Market Regulation, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (S.W.); (W.Y.); (X.Y.); (Z.Y.); (M.Z.); (Y.Q.); (B.M.)
| | | | | | | | | | | |
Collapse
|