1
|
He Y, Tian R, Shen W, Zhang J, Tao C. An autocatalytic hybridization circuit-based FRET aptasensor for detection of low-abundant sulfameter in human serum. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 326:125241. [PMID: 39388936 DOI: 10.1016/j.saa.2024.125241] [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: 06/02/2024] [Revised: 09/03/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Exposure to antibiotics is considered a potential risk factor for human health. Yet, the extensive and cost-effective detection of low-abundant antibiotics in complex matrices remains a significant challenge. Herein, an aptamer and an autocatalytic hybridization circuit (AHC) were used to fabricate a fluorescence resonance energy transfer (FRET) platform to detect sulfameter (SME) in human serum. The AHC system comprised two mutually motivated hybridization chain reactions (HCR) modules, ultimately producing long-branched DNA copolymeric nanowires. This mutually reciprocal activation of two HCR modules enables continuous signal amplification, providing the AHC system with wide linear range and high sensitivity for the SME detection. Compared to the HCR-based aptasensor, the AHC-based aptasensor exhibited a wider linear range and improved sensitivity (3.3 times greater). Under optimal conditions, the fluorescent AHC-based aptasensor demonstrated a linear range (R2 was 0.996) from 0.5 to 2000 nM, with a low detection limit of 0.301 nM (S/N = 3). The fluorescent aptasensor was also validated by SME-spiked human serum samples, showing average recoveries ranging from 96.40 % to 109.30 %, with a relative standard deviation below 10.45 %. Furthermore, when tested on six human serum samples, the aptasensor results were consistent with those obtained from the commercial ELISA method. These findings demonstrate that the proposed aptasensor provides a promising approach for the practical monitoring of low-abundant SME in human serum.
Collapse
Affiliation(s)
- Yanping He
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China.
| | - Ruifen Tian
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Weili Shen
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Jingrui Zhang
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Chen Tao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| |
Collapse
|
2
|
Khan A, Khan H, He N, Li Z, Alyahya HK, Bin Jardan YA. Colorimetric aptasensor coupled with a deep-learning-powered smartphone app for programmed death ligand-1 expressing extracellular vesicles. Front Immunol 2025; 15:1479403. [PMID: 39916963 PMCID: PMC11798968 DOI: 10.3389/fimmu.2024.1479403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/21/2024] [Indexed: 02/09/2025] Open
Abstract
Lung cancer is a devastating public health threat and a leading cause of cancer-related deaths. Therefore, it is imperative to develop sophisticated techniques for the non-invasive detection of lung cancer. Extracellular vesicles expressing programmed death ligand-1 (PD-L1) markers (PD-L1@EVs) in the blood are reported to be indicative of lung cancer and response to immunotherapy. Our approach is the development of a colorimetric aptasensor by combining the rapid capturing efficiency of (Fe3O4)-SiO2-TiO2 for EV isolation with PD-L1 aptamer-triggered enzyme-linked hybridization chain reaction (HCR) for signal amplification. The numerous HRPs catalyze their substrate dopamine (colorless) into polydopamine (blackish brown). Change in chromaticity directly correlates with the concentration of PD-L1@EVs in the sample. The colorimetric aptasensor was able to detect PD-L1@EVs at concentrations as low as 3.6×102 EVs/mL with a wide linear range from 103 to 1010 EVs/mL with high specificity and successfully detected lung cancer patients' serum from healthy volunteers' serum. To transform the qualitative colorimetric approach into a quantitative operation, we developed an intelligent convolutional neural network (CNN)-powered quantitative analyzer for chromaticity in the form of a smartphone app named ExoP, thereby achieving the intelligent analysis of chromaticity with minimal user intervention or additional hardware attachments for the sensitive and specific quantification of PD-L1@EVs. This combined approach offers a simple, sensitive, and specific tool for lung cancer detection using PD-L1@EVs. The addition of a CNN-powered smartphone app further eliminates the need for specialized equipment, making the colorimetric aptasensor more accessible for low-resource settings.
Collapse
Affiliation(s)
- Adeel Khan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Department of Biotechnology, University of Science and Technology, Bannu, Pakistan
| | - Haroon Khan
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhiyang Li
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Heba Khalil Alyahya
- Department of Exercise Physiology, College of Sport Science and Physical Activity, King Saud University, Riyadh, Saudi Arabia
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
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
|
4
|
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 2024; 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] [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
|
5
|
Balaraman AK, Babu MA, Moglad E, Mandaliya V, Rekha MM, Gupta S, Prasad GVS, Kumari M, Chauhan AS, Ali H, Goyal K. Exosome-mediated delivery of CRISPR-Cas9: A revolutionary approach to cancer gene editing. Pathol Res Pract 2024; 266:155785. [PMID: 39708520 DOI: 10.1016/j.prp.2024.155785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Several molecular strategies based on targeted gene delivery systems have been developed in recent years; however, the CRISPR-Cas9 technology introduced a new era of targeted gene editing, precisely modifying oncogenes, tumor suppressor genes, and other regulatory genes involved in carcinogenesis. However, efficiently and safely delivering CRISPR-Cas9 to cancer cells across the cell membrane and the nucleus is still challenging. Using viral vectors and nanoparticles presents issues of immunogenicity, off-target effects, and low targeting affinity. Naturally, extracellular vesicles called exosomes have garnered the most attention as delivery vehicles in oncology-related CRISPR-Cas9 calls due to their biocompatibility, loading capacity, and inherent targeting features. The following review discusses the current progress in using exosomes to deliver CRISPR-Cas9 components, the approaches to load the CRISPR components into exosomes, and the modification of exosomes to increase stability and tumor-targeted delivery. We discuss the latest strategies in targeting recently accomplished in the exosome field, including modifying the surface of exosomes to enhance their internalization by cancer cells, as well as the measures taken to overcome the impacts of TME on delivery efficiency. Focusing on in vitro and in vivo experimentation, this review shows that exosome-mediated CRISPR-Cas9 can potentially treat cancer types, including pancreatic, lymphoma, and leukemia, for given gene targets. This paper compares exosome-mediated delivery and conventional vectors regarding safety, immune response, and targeting ability. Last but not least, we present the major drawbacks and potential development of the seemingly promising field of exosome engineering in gene editing, with references to CRISPR technologies and applications that may help make the target exosomes therapeutic in oncology.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| |
Collapse
|
6
|
Song SF, Zhang XW, Chen S, Shu Y, Yu YL, Wang JH. CRISPR-based dual-aptamer proximity ligation coupled hybridization chain reaction for precise detection of tumor extracellular vesicles and cancer diagnosis. Talanta 2024; 280:126780. [PMID: 39191105 DOI: 10.1016/j.talanta.2024.126780] [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: 05/30/2024] [Revised: 08/16/2024] [Accepted: 08/24/2024] [Indexed: 08/29/2024]
Abstract
Tumor cell-derived extracellular vesicles (TEVs) contain numerous cellular molecules and are considered potential biomarkers for non-invasive liquid biopsy. However, due to the low abundance of TEVs secreted by tumor cells and their phenotypic heterogeneity, there is a lack of sensitive and specific methods to quantify TEVs. Here, we developed a dual-aptamer proximity ligation-coupled hybridization chain reaction (HCR) method for tracing TEVs, exploiting CRISPR to achieve highly sensitive detection. Taking advantage of the high binding affinity of aptamers, the two aptamers (AptEpCAM, AptHER2) exhibited the high selectivity for TEVs recognition. HCR generated long-repeated sequence containing multiple crRNA targetable barcodes, and the signals were further amplified by CRISPR upon recognizing the HCR sequences, thereby enhancing the sensitivity. Under optimal conditions, the developed method demonstrated a favorable linear relationship in the range of 2 × 103-107 particles/μL, with a limit of detection (LOD) of 3.3 × 102 particles/μL. We directly applied our assay to clinical plasma analysis, achieving 100 % accuracy in cancer diagnosis, thus demonstrating the potential clinical applications of TEVs. Due to its simplicity and rapidity, excellent sensitivity and specificity, this method has broad applications in clinical medicine.
Collapse
Affiliation(s)
- Shi-Fan Song
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Xue-Wei Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Yang Shu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| |
Collapse
|
7
|
Wang T, Huang W, Gao X, Deng Y, Huang J. Single extracellular vesicle research: From cell population to a single cell. Biochem Biophys Res Commun 2024; 734:150439. [PMID: 39083971 DOI: 10.1016/j.bbrc.2024.150439] [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: 06/14/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Extracellular vesicles (EVs) are secreted by cells with a membrane structure and complex components such as DNA, RNA and proteins. These biomolecules play an important role in cell communication, cell proliferation, cell migration, vascularization, immune response and other physiological and pathological processes. Most current research on EVs focused on populations of EVs. Heterogeneity of EVs is neglected. Considering the heterogeneity of single EVs may offer critical molecular insights into cell-cell interactions, it is necessary to enhance our understanding about molecular characteristics from EVs derived from cell population to a single EV of derived from a single cell. This transformation is expected to provide a new insight into the understanding of cellular biology and the accurate description of the law of disease progress. In this article, we review the current research progress of single EV analysis technology for single EVs derived from cell population (SECP) and discuss its main applications in biological and clinical medicine research. After that, we propose the development direction, main difficulties and application prospect of single EV analysis technology for single EVs derived from single cells (SESC) according to our own research work, to provide new perspectives for the field of EV research.
Collapse
Affiliation(s)
- Ting Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xu Gao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
8
|
Feng Y, Yang J, He Z, Liu X, Ma C. CRISPR-Cas-based biosensors for the detection of cancer biomarkers. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6634-6653. [PMID: 39258950 DOI: 10.1039/d4ay01446d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Along with discovering cancer biomarkers, non-invasive detection methods have played a critical role in early cancer diagnosis and prognostic improvement. Some traditional detection methods have been used for detecting cancer biomarkers, but they are time-consuming and involve materials and human costs. With great flexibility, sensitivity and specificity, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated system provides a wide range of application prospects in this field. Herein, we introduce the background of the CRISPR-Cas (CRISPR-associated) system and comprehensively summarize the diagnosis strategies of cancer mediated by the CRISPR-Cas system, including four kinds of biochemical-based markers: nucleic acid, enzyme, tumor-specific protein and exosome. Furthermore, we discuss the challenges in implementing the CRISPR-Cas system in clinical applications.
Collapse
Affiliation(s)
- Yuxin Feng
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jinmeng Yang
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Ziping He
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Xinfa Liu
- School of Life Sciences, Central South University, Changsha 410013, China.
| | - Changbei Ma
- School of Life Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
9
|
Guo Y, Zhou Y, Duan H, Xu D, Wei M, Wu Y, Xiong Y, Chen X, Wang S, Liu D, Huang X, Xin H, Xiong Y, Tang BZ. CRISPR/Cas-mediated "one to more" lighting-up nucleic acid detection using aggregation-induced emission luminogens. Nat Commun 2024; 15:8560. [PMID: 39362874 PMCID: PMC11450156 DOI: 10.1038/s41467-024-52931-0] [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: 12/07/2023] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
CRISPR diagnostics are effective but suffer from low signal transduction efficiency, limited sensitivity, and poor stability due to their reliance on the trans-cleavage of single-stranded nucleic acid fluorescent reporters. Here, we present CrisprAIE, which integrates CRISPR/Cas reactions with "one to more" aggregation-induced emission luminogen (AIEgen) lighting-up fluorescence generated by the trans-cleavage of Cas proteins to AIEgen-incorporated double-stranded DNA labeled with single-stranded nucleic acid linkers and Black Hole Quencher groups at both ends (Q-dsDNA/AIEgens-Q). CrisprAIE demonstrates superior performance in the clinical nucleic acid detection of norovirus and SARS-CoV-2 regardless of amplification. Moreover, the diagnostic potential of CrisprAIE is further enhanced by integrating it with spherical nucleic acid-modified AIEgens (SNA/AIEgens) and a portable cellphone-based readout device. The improved CrisprAIE system, utilizing Q-dsDNA/AIEgen-Q and SNA/AIEgen reporters, exhibits approximately 80- and 270-fold improvements in sensitivity, respectively, compared to conventional CRISPR-based diagnostics. We believe CrisprAIE can be readily extended as a universal signal generation strategy to significantly enhance the detection efficiency of almost all existing CRISPR-based diagnostics.
Collapse
Affiliation(s)
- Yuqian Guo
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yaofeng Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hong Duan
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University, Beijing, China
| | - Derong Xu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Min Wei
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Ying Xiong
- National Engineering Research Center of Rice and Byproducts Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Xirui Chen
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Siyuan Wang
- Key Laboratory of Agricultural Information Acquisition Technology, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Daofeng Liu
- Jiangxi Provincial Center for Disease Control and Prevention, Nanchang, China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China.
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, China.
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| |
Collapse
|
10
|
Razavi Z, Soltani M, Souri M, Pazoki-Toroudi H. CRISPR-Driven Biosensors: A New Frontier in Rapid and Accurate Disease Detection. Crit Rev Anal Chem 2024:1-25. [PMID: 39288095 DOI: 10.1080/10408347.2024.2400267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
This comprehensive review delves into the advancements and challenges in biosensing, with a strong emphasis on the transformative potential of CRISPR technology for early and rapid detection of infectious diseases. It underscores the versatility of CRISPR/Cas systems, highlighting their ability to detect both nucleic acids and non-nucleic acid targets, and their seamless integration with isothermal amplification techniques. The review provides a thorough examination of the latest developments in CRISPR-based biosensors, detailing the unique properties of CRISPR systems, such as their high specificity and programmability, which make them particularly effective for detecting disease-associated nucleic acids. While the review focuses on nucleic acid detection due to its critical role in diagnosing infectious diseases, it also explores the broader applications of CRISPR technology in detecting non-nucleic acid targets, thereby acknowledging the technology's broader potential. Additionally, the review identifies existing challenges, such as the need for improved signal amplification and real-world applicability, and offers future perspectives aimed at overcoming these hurdles. The ultimate goal is to advance the development of highly sensitive and specific CRISPR-based biosensors that can be used widely for improving human health, particularly in point-of-care settings and resource-limited environments.
Collapse
Affiliation(s)
- ZahraSadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran
- Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
- Centre for Sustainable Business, International Business University, Toronto, Canada
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
11
|
Huang Z, Deng C, Ma C, He G, Tao J, Zhang L, Hu X, Mo Y, Qiu L, Zhang N, Luo C, Xing S, Xie J, Yin H. Identification and validation of the surface proteins FIBG, PDGF-β, and TGF-β on serum extracellular vesicles for non-invasive detection of colorectal cancer: experimental study. Int J Surg 2024; 110:4672-4687. [PMID: 38704642 PMCID: PMC11326011 DOI: 10.1097/js9.0000000000001533] [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: 02/29/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVES The absence of non-invasive biomarkers for the early diagnosis of colorectal cancer (CRC) has contributed to poor prognosis. Extracellular vesicles (EVs) have emerged as promising candidates for cancer monitoring using liquid biopsy. However, the complexity of EVs isolation procedures and the absence of clear targets for detecting serum-derived EVs have hindered the clinical application of EVs in early CRC diagnosis. METHODS In the discovery phase, we conducted a comprehensive 4D-DIA proteomic analysis of serum-derived EVs samples from 37 individuals, performing an initial screening of EVs surface proteins. In the technical validation phase, we developed an extraction-free CRC-EVArray microarray to assess the expression of these potential EVs surface proteins in a multi-centre study comprising 404 individuals. In the application phase, the authors evaluated the diagnostic efficacy of the CRC-EVArray model based on machine-learning algorithms. RESULTS Through 4D-DIA proteomic analysis, the authors identified seven potential EVs surface proteins showing significantly differential expression in CRC patients compared to healthy controls. Utilizing our developed high-throughput CRC-EVArray microarray, we further confirmed the differential expression of three EVs surface proteins, FIBG, PDGF-β and TGF-β, in a large sample population. Moreover, we established an optimal CRC-EVArray model using the NNET algorithm, demonstrating superior diagnostic efficacy with an area under the curve (AUC) of 0.882 in the train set and 0.937 in the test set. Additionally, we predicted the functions and potential origins of these EVs-derived proteins through a series of multi-omics approaches. CONCLUSIONS Our systematic exploration of surface protein expression profiles on serum-derived EVs has identified FIBG, PDGF-β, and TGF-β as novel diagnostic biomarkers for CRC. The development of CRC-EVArray diagnostic model based on these findings provided an effective tool for the large-scale CRC screening, thus facilitating its translation into clinical practice.
Collapse
Affiliation(s)
- Zhijian Huang
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University
| | - Cuncan Deng
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University
| | - Caiqi Ma
- Department of Oncology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University
| | - Guirong He
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Jian Tao
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Lijun Zhang
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Xiaoyun Hu
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Yanfang Mo
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Lumei Qiu
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Ningfang Zhang
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| | - Chuanghua Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shan Xing
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center
| | - Jinye Xie
- Department of Laboratory Medicine, Zhongshan City People's Hospital, Zhongshan
| | - Haofan Yin
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University
- Department of Laboratory Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen
| |
Collapse
|
12
|
Song R, Chen Z, Xiao H, Wang H. The CRISPR-Cas system in molecular diagnostics. Clin Chim Acta 2024; 561:119820. [PMID: 38901631 DOI: 10.1016/j.cca.2024.119820] [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: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Robust, sensitive, and rapid molecular detection tools are essential prerequisites for disease diagnosis and epidemiological control. However, the current mainstream tests necessitate expensive equipment and specialized operators, impeding the advancement of molecular diagnostics. The CRISPR-Cas system is an integral component of the bacterial adaptive immune system, wherein Cas proteins recognize PAM sequences by binding to CRISPR RNA, subsequently triggering DNA or RNA cleavage. The discovery of the CRISPR-Cas system has invigorated molecular diagnostics. With further in-depth research on this system, its application in molecular diagnosis is flourishing. In this review, we provide a comprehensive overview of recent research progress on the CRISPR-Cas system, specifically focusing on its application in molecular diagnosis.
Collapse
Affiliation(s)
- Rao Song
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Zhongyi Chen
- Department of Pathology, Suining Central Hospital, Suining 629000, China
| | - Hongtao Xiao
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Haojun Wang
- Department of Pathology, Suining Central Hospital, Suining 629000, China.
| |
Collapse
|
13
|
Zhang XW, Qi GX, Chen S, Yu YL, Wang JH. Ultrasensitive and Wash-Free Detection of Tumor Extracellular Vesicles by Aptamer-Proximity-Ligation-Activated Rolling Circle Amplification Coupled to Single Particle ICP-MS. Anal Chem 2024; 96:10800-10808. [PMID: 38904228 DOI: 10.1021/acs.analchem.4c02066] [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: 06/22/2024]
Abstract
Tumor-derived extracellular vesicles (TEVs) are rich in cellular information and hold great promise as a biomarker for noninvasive cancer diagnosis. However, accurate measurement of TEVs presents challenges due to their low abundance and potential interference from a high number of EVs derived from normal cells. Herein, an aptamer-proximity-ligation-activated rolling circle amplification (RCA) method for EV membrane recognition, coupled with single particle inductively coupled plasma mass spectrometry (sp-ICP-MS) for the quantification of TEVs, is developed. When DNA-labeled ultrasmall gold nanoparticle (AuNP) probes bind to the long chains formed by RCA, they aggregate to form large particles. Notably, small AuNPs scarcely produce pulse signals in sp-ICP-MS, thereby detecting TEVs in a wash-free manner. By leveraging the strong binding affinity of aptamers, dual aptamers for EpCAM and PD-L1 recognition, and the sp-ICP-MS technique, this method offers remarkable sensitivity and selectivity in tracing TEVs. Under optimized conditions, the present method shows a favorable linear relationship between the pulse signal frequency of sp-ICP-MS and TEV concentration within the range of 105-107 particles/mL, along with a detection limit of 1.1 × 104 particles/mL. The pulse signals from sp-ICP-MS combined with machine learning algorithms are used to discriminate cancer patients from healthy donors with 100% accuracy. Due to its simple and fast operation and excellent sensitivity and accuracy, this approach holds significant potential for diverse applications in life sciences and personalized medicine.
Collapse
Affiliation(s)
- Xue-Wei Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Gong-Xiang Qi
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
14
|
Yang G, Li Z, Usman R, Liu Y, Li S, Chen Z, Chen H, Deng Y, Fang Y, He N. From biogenesis to aptasensors: advancements in analysis for tumor-derived extracellular vesicles research. Theranostics 2024; 14:4161-4183. [PMID: 38994022 PMCID: PMC11234286 DOI: 10.7150/thno.95885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Extracellular vesicles (EVs) are enclosed by a nanoscale phospholipid bilayer membrane and typically range in size from 30 to 200 nm. They contain a high concentration of specific proteins, nucleic acids, and lipids, reflecting but not identical to the composition of the parent cell. The inherent characteristics and variety of EVs give them extensive and unique advantages in the field of cancer identification and treatment. Recently, EVs have been recognized as potential tumor markers for the detection of cancer. Aptamers, which are molecules of single-stranded DNA or RNA, demonstrate remarkable specificity and affinity for their targets by adopting distinct tertiary structures. Aptamers offer various advantages over their protein counterparts, such as reduced immunogenicity, the ability for convenient large-scale synthesis, and straightforward chemical modification. In this review, we summarized EVs biogenesis, sample collection, isolation, storage and characterization, and finally provided a comprehensive survey of analysis techniques for EVs detection that are based on aptamers.
Collapse
Affiliation(s)
- Gaojian Yang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhiyang Li
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Rabia Usman
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuan Liu
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
- Institute for Future Sciences, University of South China, Changsha Hunan 410000, China
| | - Song Li
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
- Institute for Future Sciences, University of South China, Changsha Hunan 410000, China
| | - Zhu Chen
- China Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, PR China
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
- Institute for Future Sciences, University of South China, Changsha Hunan 410000, China
| | - Hui Chen
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
- Institute for Future Sciences, University of South China, Changsha Hunan 410000, China
| | - Yan Deng
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
- Institute for Future Sciences, University of South China, Changsha Hunan 410000, China
| | - Yile Fang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Nongyue He
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- China Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, PR China
| |
Collapse
|
15
|
Wu X, Cui J, Sun Q, Wang X, Chen J, Liu Y, Chen JH, Jiang D, Zhou Z, Zhou H. Organic photoelectrochemical transistor based on cascaded DNA network structure modulated ZnIn 2S 4/MXene Schottky junction for sensitive ATP detection. Talanta 2024; 274:125992. [PMID: 38552479 DOI: 10.1016/j.talanta.2024.125992] [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: 01/09/2024] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 05/04/2024]
Abstract
Organic photoelectrochemical transistor (OPECT) biosensor is now appearing in perspective of public, which characterized by amplified the grating electrode potential by ion transport. In this study, the DNA network formed by the hybridization chain reaction (HCR) detects the target adenosine triphosphate (ATP) by adjusting the surface potential of the new heterojunction of ZnIn2S4/MXene. The formation of DNA network amplifies the detection signal of ATP. Significantly, OPECT biosensor could further amplify the signal, which calculated the gain achieved 103, which is consistent with the gain signal of the previously reported OPECT biosensor. Furthermore, the OPECT biosensor achieved a highly sensitivity detection of the target ATP, which the linear detection range is 0.03 pM-30 nM, and the detection limit is 0.03 pM, and illustrated a high selectivity to ATP. The proposed OPECT biosensor achieved signal amplification by adjusting the surface potential of ZnIn2S4/MXene through cascade DNA network, which provides a new direction for the detection of biomolecules.
Collapse
Affiliation(s)
- Xiaodi Wu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Jiayi Cui
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Qihao Sun
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Xue Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Jiahe Chen
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Yue Liu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Jia-Hao Chen
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Degang Jiang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Zhongmin Zhou
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| | - Hong Zhou
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
16
|
Ren Y, Ge K, Tang Q, Liang X, Fan L, Ye K, Wang M, Yao B. Dual-Recognition-Mediated Autocatalytic Amplification Assay for the Subpopulations of PD-L1 Positive Extracellular Vesicle. Anal Chem 2024; 96:9585-9592. [PMID: 38816678 DOI: 10.1021/acs.analchem.4c01111] [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: 06/01/2024]
Abstract
The PD-L1 protein on extracellular vesicles (EVs) is a promising biomarker for tumor immunotherapy. However, PD-L1+ EVs have various cell origins, so further analysis of the subpopulations is essential to help understand better their relationship with tumor immunotherapy. Different from the previous work which focus on the level of total PD-L1+ EVs expression, we, herein, report a dual-recognition mediated autocatalytic amplification (DRMAA) assay to detect the PD-L1 derived from tumors (EpCAM+), immune T cells (CD3+), and total (Lipids) EVs, respectively. The DRMAA assay employed proximity hybridization to construct a complete trigger sequence and then catalyzed the cross-hybridization of three hairpin probes, producing a three-way DNA junction (3-WJ) structure carrying the newly exposed trigger sequence. The 3-WJ complex subsequently initiated an autocatalytic amplification reaction and higher sensitivity than the traditional catalytic hairpin assembly assay was obtained. It was found that the EpCAM+ and PD-L1+ EVs were more effective than others in distinguishing lung cancer patients from healthy people. Surprisingly, the CD3+ and PD-L1+ EVs in lung cancer patients were also upregulated, indicating that immune cell-derived PD-L1+ EVs are also non-negligible marker in a tumor microenvironment. Our results suggested that the DRMAA assay would improve the study of subpopulations of PD-L1+ EVs to provide new insights for cancer immunotherapies.
Collapse
Affiliation(s)
- Yongan Ren
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Ke Ge
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - QiaoQiao Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Xiaoxuan Liang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Linlin Fan
- Jining First People's Hospital, Jining 272002, China
| | - Kai Ye
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Min Wang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
17
|
Lim W, Lee S, Koh M, Jo A, Park J. Recent advances in chemical biology tools for protein and RNA profiling of extracellular vesicles. RSC Chem Biol 2024; 5:483-499. [PMID: 38846074 PMCID: PMC11151817 DOI: 10.1039/d3cb00200d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized vesicles secreted by cells that contain various cellular components such as proteins, nucleic acids, and lipids from the parent cell. EVs are abundant in body fluids and can serve as circulating biomarkers for a variety of diseases or as a regulator of various biological processes. Considering these characteristics of EVs, analysis of the EV cargo has been spotlighted for disease diagnosis or to understand biological processes in biomedical research. Over the past decade, technologies for rapid and sensitive analysis of EVs in biofluids have evolved, but detection and isolation of targeted EVs in complex body fluids is still challenging due to the unique physical and biological properties of EVs. Recent advances in chemical biology provide new opportunities for efficient profiling of the molecular contents of EVs. A myriad of chemical biology tools have been harnessed to enhance the analytical performance of conventional assays for better understanding of EV biology. In this review, we will discuss the improvements that have been achieved using chemical biology tools.
Collapse
Affiliation(s)
- Woojeong Lim
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Soyeon Lee
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Minseob Koh
- Department of Chemistry, Pusan National University Busan 46241 Republic of Korea
| | - Ala Jo
- Center for Nanomedicine, Institute for Basic Science Seoul 03722 Republic of Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University Chuncheon 24341 Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University Chuncheon 24341 Republic of Korea
| |
Collapse
|
18
|
Deng J, Liu C, Sun J. DNA-Based Nanomaterials for Analysis of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303092. [PMID: 38016069 DOI: 10.1002/adma.202303092] [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: 04/03/2023] [Revised: 07/21/2023] [Indexed: 11/30/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanovesicles comprising a myriad of molecular cargo such as proteins and nucleic acids, playing essential roles in intercellular communication and physiological and pathological processes. EVs have received substantial attention as noninvasive biomarkers for disease diagnosis and prognosis. Owing to their ability to recognize protein and nucleic acid targets, DNA-based nanomaterials with excellent programmability and modifiability provide a promising tool for the sensitive and accurate detection of molecular cargo carried by EVs. In this perspective, recent advancements in EV analysis using a variety of DNA-based nanomaterials are summarized, which can be broadly classified into three categories: linear DNA probes, DNA nanostructures, and hybrid DNA nanomaterials. The design, construction, advantages, and disadvantages of different types of DNA nanomaterials, as well as their performance for detecting EVs are reviewed. The challenges and opportunities in the field of EV analysis by DNA nanomaterials are also discussed.
Collapse
Affiliation(s)
- Jinqi Deng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Liu
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
19
|
Wang ZY, Teng SQ, Zhao NN, Han Y, Li DL, Zhang CY. Ligase detection reaction amplification-activated CRISPR-Cas12a for single-molecule counting of FEN1 in breast cancer tissues. Chem Commun (Camb) 2024; 60:3075-3078. [PMID: 38404229 DOI: 10.1039/d4cc00408f] [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: 02/27/2024]
Abstract
We construct a simple fluorescent biosensor for single-molecule counting of flap endonuclease 1 (FEN1) based on ligase detection reaction (LDR) amplification-activated CRISPR-Cas12a. This biosensor exhibits excellent selectivity and high sensitivity with a detection limit (LOD) of 1.31 × 10-8 U. Moreover, it can be employed to screen the FEN1 inhibitors and quantitatively measure the FEN1 activity in human cells and breast cancer tissues, holding great promise in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Zi-Yue Wang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Shuang-Qian Teng
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Ning-Ning Zhao
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Yun Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Dong-Ling Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Chun-Yang Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
20
|
Wu C, Yue Y, Huang B, Ji H, Wu L, Huang H. CRISPR-powered microfluidic biosensor for preamplification-free detection of ochratoxin A. Talanta 2024; 269:125414. [PMID: 37992484 DOI: 10.1016/j.talanta.2023.125414] [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: 08/15/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
The CRISPR technology, which does not require complex instruments, expensive reagents or professional operators, has attracted a lot of attention. When utilizing the CRISPR-Cas system for detection, the pre-amplification step is often necessary to enhance sensitivity. However, this approach tends to introduce complexity and prolong the time required. To address this issue, we employed Pd@PCN-222 nanozyme to label single-stranded DNA, referred to as Pd@PCN-222 CRISPR nanozyme, which serves as the reporter of the CRISPR system. Pd@PCN-222 nanozyme possess exceptional catalytic activity for the reduction of H2O2. Compared with traditional electrochemical probe ferrocene and methylene blue without catalytic activity, there is a significant amplification of the electrochemical signal. So the need for pre-amplification was eliminated. In this study, we constructed a CRISPR-Cas system for ochratoxin A, utilizing the Pd@PCN-222 CRISPR nanozyme to amplified signal avoiding pre-amplification with outstanding detection of 1.21 pg/mL. Furthermore, we developed a microfluidic electrochemical chip for the on-site detection of ochratoxin A. This achievement holds significant promise in establishing a practical on-site detection platform for identifying food safety hazards.
Collapse
Affiliation(s)
- Chengyuan Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yuanyuan Yue
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | | | - Hanxu Ji
- Key Laboratory of Biotoxin Analysis & Assessment for State Market Regulation, Nanjing Institute of Product Quality Inspection & Testing, Nanjing, 210019, China
| | - Lina Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - He Huang
- Food Laboratory of Zhongyuan, Luohe, 462300, Henan, China.
| |
Collapse
|
21
|
Kong H, Yi K, Mintz RL, Wang B, Xu Y, Lao YH, Tao Y, Li M. CRISPR/Cas detection with nanodevices: moving deeper into liquid biopsy. Chem Commun (Camb) 2024; 60:2301-2319. [PMID: 38251733 DOI: 10.1039/d3cc05375j] [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: 01/23/2024]
Abstract
The emerging field of liquid biopsy has garnered significant interest in precision diagnostics, offering a non-invasive and repetitive method for analyzing bodily fluids to procure real-time diagnostic data. The precision and accuracy offered by the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (CRISPR/Cas) technology have advanced and broadened the applications of liquid biopsy. Significantly, when combined with swiftly advancing nanotechnology, CRISPR/Cas-mediated nanodevices show vast potential in precise liquid biopsy applications. However, persistent challenges are still associated with off-target effects, and the current platforms also constrain the performance of the assays. In this review, we highlight the merits of CRISPR/Cas systems in liquid biopsy, tracing the development of CRISPR/Cas systems and their current applications in disease diagnosis particularly in liquid biopsies. We also outline ongoing efforts to design nanoscale devices with improved sensing and readout capabilities, aiming to enhance the performance of CRISPR/Cas detectors in liquid biopsy. Finally, we identify the critical obstacles hindering the widespread adoption of CRISPR/Cas liquid biopsy and explore potential solutions. This feature article presents a comprehensive overview of CRISPR/Cas-mediated liquid biopsies, emphasizing the progress in integrating nanodevices to improve specificity and sensitivity. It also sheds light on future research directions in employing nanodevices for CRISPR/Cas-based liquid biopsies in the realm of precision medicine.
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Bin Wang
- Department of Infectious Diseases, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130061, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
22
|
Zeng D, Jiao J, Mo T. Combination of nucleic acid amplification and CRISPR/Cas technology in pathogen detection. Front Microbiol 2024; 15:1355234. [PMID: 38380103 PMCID: PMC10877009 DOI: 10.3389/fmicb.2024.1355234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024] Open
Abstract
Major health events caused by pathogenic microorganisms are increasing, seriously jeopardizing human lives. Currently PCR and ITA are widely used for rapid testing in food, medicine, industry and agriculture. However, due to the non-specificity of the amplification process, researchers have proposed the combination of nucleic acid amplification technology with the novel technology CRISPR for detection, which improves the specificity and credibility of results. This paper summarizes the research progress of nucleic acid amplification technology in conjunction with CRISPR/Cas technology for the detection of pathogens, which provides a reference and theoretical basis for the subsequent application of nucleic acid amplification technology in the field of pathogen detection.
Collapse
Affiliation(s)
| | | | - Tianlu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
23
|
Lee I, Kwon SJ, Heeger P, Dordick JS. Ultrasensitive ImmunoMag-CRISPR Lateral Flow Assay for Point-of-Care Testing of Urinary Biomarkers. ACS Sens 2024; 9:92-100. [PMID: 38141036 PMCID: PMC11090086 DOI: 10.1021/acssensors.3c01694] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Rapid, accurate, and noninvasive detection of biomarkers in saliva, urine, or nasal fluid is essential for the identification, early diagnosis, and monitoring of cancer, organ failure, transplant rejection, vascular diseases, autoimmune disorders, and infectious diseases. We report the development of an Immuno-CRISPR-based lateral flow assay (LFA) using antibody-DNA barcode complexes with magnetic enrichment of the target urinary biomarkers CXCL9 and CXCL10 for naked eye detection (ImmunoMag-CRISPR LFA). An intermediate approach involving a magnetic bead-based Immuno-CRISPR assay (ImmunoMag-CRISPR) resulted in a limit of detection (LOD) of 0.6 pg/mL for CXCL9. This value surpasses the detection limits achieved by previously reported assays. The highly sensitive detection method was then re-engineered into an LFA format with an LOD of 18 pg/mL for CXCL9, thereby enabling noninvasive early detection of acute kidney transplant rejection. The ImmunoMag-CRISPR LFA was tested on 42 clinical urine samples from kidney transplant recipients, and the assay could determine 11 positive and 31 negative urinary samples through a simple visual comparison of the test line and the control line of the LFA strip. The LFA system was then expanded to quantify the CXCL9 and CXCL10 levels in clinical urine samples from images. This approach has the potential to be extended to a wide range of point-of-care tests for highly sensitive biomarker detection.
Collapse
Affiliation(s)
- Inseon Lee
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States
| | - Seok-Joon Kwon
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States
| | - Peter Heeger
- Comprehensive Transplant Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Jonathan S. Dordick
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States
| |
Collapse
|
24
|
Tan W, Zhang C, Cheng S, Hu X, Wang M, Xian Y. DNA Gate-Based CRISPR-Cas Exponential Amplification System for Ultrasensitive Small Extracellular Vesicle Detection to Enhance Breast Cancer Diagnosis. Anal Chem 2024; 96:1328-1335. [PMID: 38190500 DOI: 10.1021/acs.analchem.3c04873] [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: 01/10/2024]
Abstract
Tumor-derived small extracellular vesicles (tEVs) as potential biomarkers possess abundant surface proteins closely related to parent cells, which are crucial for noninvasive cancer diagnosis. However, tEVs exhibit phenotype heterogeneity and low abundance, posing a significant challenge for multiplex detection with a high sensitivity. Herein, we developed a DNA gate-based exponential amplification CRISPR-Cas (DGEAC) system for accurate and ultrasensitive detection of tEVs, which can greatly improve the accuracy of breast cancer (BC) diagnosis. Based on the coexpression of CD63 and vascular endothelial growth factor (VEGF) on BC-derived tEVs, we developed a dual-aptamer-based AND gate fluorescent probe by proximity hybridization. By integrating the target recognition and trans-cleavage activity of Cas12a, an autocatalysis-driven exponential amplification circuit was developed for ultrasensitive detection of CD63 and VEGF proteins on tEVs, which could avoid false negative signals from single protein or other interfering proteins. We achieved highly sensitive detection of tEVs over a linear range from 1.75 × 103 to 3.5 × 108 particles/mL with a detection limit as low as 1.02 × 103 particles/mL. Furthermore, the DGEAC system can distinguish tEVs from tEVs derived from different BC cell lines, including MDA-MB-231, MCF-7, SKBR3, and MCF-10A. Compared to linear amplification (AUC 90.0%), the DGEAC system effectively differentiates BC in different stages (AUC 98.3%).
Collapse
Affiliation(s)
- Wenqiao Tan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xinyu Hu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Man Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| |
Collapse
|
25
|
Wu YX, Tian BY, Ou XY, Wu M, Huang Q, Han RK, He X, Chen SL. A novel model for predicting prognosis and response to immunotherapy in nasopharyngeal carcinoma patients. Cancer Immunol Immunother 2024; 73:14. [PMID: 38236288 PMCID: PMC10796600 DOI: 10.1007/s00262-023-03626-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: 10/20/2023] [Accepted: 12/30/2023] [Indexed: 01/19/2024]
Abstract
Blood-based biomarkers of immune checkpoint inhibitors (ICIs) response in patients with nasopharyngeal carcinoma (NPC) are lacking, so it is necessary to identify biomarkers to select NPC patients who will benefit most or least from ICIs. The absolute values of lymphocyte subpopulations, biochemical indexes, and blood routine tests were determined before ICIs-based treatments in the training cohort (n = 130). Then, the least absolute shrinkage and selection operator (Lasso) Cox regression analysis was developed to construct a prediction model. The performances of the prediction model were compared to TNM stage, treatment, and Epstein-Barr virus (EBV) DNA using the concordance index (C-index). Progression-free survival (PFS) was estimated by Kaplan-Meier (K-M) survival curve. Other 63 patients were used for validation cohort. The novel model composed of histologic subtypes, CD19+ B cells, natural killer (NK) cells, regulatory T cells, red blood cells (RBC), AST/ALT ratio (SLR), apolipoprotein B (Apo B), and lactic dehydrogenase (LDH). The C-index of this model was 0.784 in the training cohort and 0.735 in the validation cohort. K-M survival curve showed patients with high-risk scores had shorter PFS compared to the low-risk groups. For predicting immune therapy responses, the receiver operating characteristic (ROC), decision curve analysis (DCA), net reclassifcation improvement index (NRI) and integrated discrimination improvement index (IDI) of this model showed better predictive ability compared to EBV DNA. In this study, we constructed a novel model for prognostic prediction and immunotherapeutic response prediction in NPC patients, which may provide clinical assistance in selecting those patients who are likely to gain long-lasting clinical benefits to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Ya-Xian Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Bo-Yu Tian
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Xin-Yuan Ou
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Meng Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Qi Huang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Run-Kun Han
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Xia He
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China.
| | - Shu-Lin Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China.
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
26
|
Yang G, Li W, Zhang S, Hu B, Huang Z. Highly-efficient selection of aptamers for detecting various HPV subtypes in clinical samples. Talanta 2024; 266:125039. [PMID: 37604070 DOI: 10.1016/j.talanta.2023.125039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023]
Abstract
Nucleic acid aptamers are of great potentials in diagnostic and therapeutic applications because of their unique molecular recognition capabilities. However, satisfactory aptamers with high affinity and specificity are still in short supply. Herein, we have developed new selection methods allowing the free interactions between the targets and potential aptamers in solution. In our selection system, the protein targets (biotinylated randomly or site-specifically) were first incubated with the random DNA library, followed by the pull-down with the streptavidin magnetic beads or biolayer-interferometry (BLI) sensors. By comparing the two biotinylation strategies (random or site-specific) and two states of the targets (free or immobilized), we have found that the combination of the site-specific biotinylation and free-target strategies was most successful. Based on these highly-efficient selection strategies, HPV L1 aptamers were obtained. By designing the sandwich aptasensor assisted with RCA and CRISPR/Cas12a, we have diagnosed various HPV subtypes in clinical samples, such as easily-collected urine samples. In summary, our new strategy can allow efficient selection of aptamers with high affinity and specificity for clinical applications.
Collapse
Affiliation(s)
- Guotai Yang
- Key Laboratory of Bio-Resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610064, PR China
| | - Wei Li
- Key Laboratory of Bio-Resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610064, PR China
| | - Shun Zhang
- Key Laboratory of Bio-Resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610064, PR China
| | - Bei Hu
- Key Laboratory of Bio-Resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610064, PR China
| | - Zhen Huang
- Key Laboratory of Bio-Resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, 610064, PR China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610000, PR China; SeNA Research Institute and Szostak-CDHT Large Nucleic Acids Institute, Chengdu, Sichuan, 610095, PR China.
| |
Collapse
|
27
|
Asleh K, Dery V, Taylor C, Davey M, Djeungoue-Petga MA, Ouellette RJ. Extracellular vesicle-based liquid biopsy biomarkers and their application in precision immuno-oncology. Biomark Res 2023; 11:99. [PMID: 37978566 PMCID: PMC10655470 DOI: 10.1186/s40364-023-00540-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
While the field of precision oncology is rapidly expanding and more targeted options are revolutionizing cancer treatment paradigms, therapeutic resistance particularly to immunotherapy remains a pressing challenge. This can be largely attributed to the dynamic tumor-stroma interactions that continuously alter the microenvironment. While to date most advancements have been made through examining the clinical utility of tissue-based biomarkers, their invasive nature and lack of a holistic representation of the evolving disease in a real-time manner could result in suboptimal treatment decisions. Thus, using minimally-invasive approaches to identify biomarkers that predict and monitor treatment response as well as alert to the emergence of recurrences is of a critical need. Currently, research efforts are shifting towards developing liquid biopsy-based biomarkers obtained from patients over the course of disease. Liquid biopsy represents a unique opportunity to monitor intercellular communication within the tumor microenvironment which could occur through the exchange of extracellular vesicles (EVs). EVs are lipid bilayer membrane nanoscale vesicles which transfer a plethora of biomolecules that mediate intercellular crosstalk, shape the tumor microenvironment, and modify drug response. The capture of EVs using innovative approaches, such as microfluidics, magnetic beads, and aptamers, allow their analysis via high throughput multi-omics techniques and facilitate their use for biomarker discovery. Artificial intelligence, using machine and deep learning algorithms, is advancing multi-omics analyses to uncover candidate biomarkers and predictive signatures that are key for translation into clinical trials. With the increasing recognition of the role of EVs in mediating immune evasion and as a valuable biomarker source, these real-time snapshots of cellular communication are promising to become an important tool in the field of precision oncology and spur the recognition of strategies to block resistance to immunotherapy. In this review, we discuss the emerging role of EVs in biomarker research describing current advances in their isolation and analysis techniques as well as their function as mediators in the tumor microenvironment. We also highlight recent lung cancer and melanoma studies that point towards their application as predictive biomarkers for immunotherapy and their potential clinical use in precision immuno-oncology.
Collapse
Affiliation(s)
- Karama Asleh
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada.
| | - Valerie Dery
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
| | - Catherine Taylor
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Michelle Davey
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | | | - Rodney J Ouellette
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- Dr Georges L. Dumont University Hospital, Vitalite Health Network, Moncton, New Brunswick, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| |
Collapse
|
28
|
Dey D, Ghosh S, Mirgh D, Panda SP, Jha NK, Jha SK. Role of exosomes in prostate cancer and male fertility. Drug Discov Today 2023; 28:103791. [PMID: 37777169 DOI: 10.1016/j.drudis.2023.103791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Prostate cancer (PCa) is the second most common and fifth most aggressive neoplasm among men worldwide. In the last decade, extracellular vesicle (EV) research has decoded multiple unsolved cancer-related mysteries. EVs can be classified as microvesicles, apoptotic bodies, and exosomes, among others. Exosomes play a key role in cellular signaling. Their internal cargos (nucleic acids, proteins, lipids) influence the recipient cell. In PCa, the exosome is the regulator of cancer progression. It is also a promising theranostics tool for PCa. Moreover, exosomes have strong participation in male fertility complications. This review aims to highlight the exosome theranostics signature in PCa and its association with male fertility.
Collapse
Affiliation(s)
- Dwaipayan Dey
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Rahara, West Bengal 700118, India
| | - Srestha Ghosh
- Department of Microbiology, Lady Brabourne College, Kolkata 700017, West Bengal, India
| | - Divya Mirgh
- Johns Hopkins University, Baltimore, MD 21218, USA
| | - Siva Parsad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal, University, Dehradun, India.
| |
Collapse
|
29
|
Zhu Y, Zhang M, Guo S, Xu H, Jie Z, Tao SC. CRISPR-based diagnostics of different biomolecules from nucleic acids, proteins, and small molecules to exosomes. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1539-1550. [PMID: 37528660 PMCID: PMC10577475 DOI: 10.3724/abbs.2023134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/29/2023] [Indexed: 08/03/2023] Open
Abstract
CRISPR-based detection technologies have been widely explored for molecular diagnostics. However, the challenge lies in converting the signal of different biomolecules, such as nucleic acids, proteins, small molecules, exosomes, and ions, into a CRISPR-based nucleic acid detection signal. Understanding the detection of different biomolecules using CRISPR technology can aid in the development of practical and promising detection approaches. Unfortunately, existing reviews rarely provide an overview of CRISPR-based molecular diagnostics from the perspective of different biomolecules. Herein, we first introduce the principles and characteristics of various CRISPR nucleases for molecular diagnostics. Then, we focus on summarizing and evaluating the latest advancements in CRISPR-based detection of different biomolecules. Through a comparison of different methods of amplification and signal readout, we discuss how general detection methods can be integrated with CRISPR. Finally, we conclude by identifying opportunities for the improvement of CRISPR in quantitative, amplification-free, multiplex, all-in-one, and point-of-care testing (POCT) purposes.
Collapse
Affiliation(s)
- Yuanshou Zhu
- Shanghai Center for Systems BiomedicineKey Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200240China
- School of Biomedical EngineeringMed-X Research InstituteShanghai Jiao Tong UniversityShanghai200030China
| | - Meng Zhang
- Department of Pulmonary and Critical Care MedicineShanghai Fifth People’s HospitalFudan UniversityShanghai200240China
| | - Shujuan Guo
- Shanghai Center for Systems BiomedicineKey Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200240China
| | - Hong Xu
- School of Biomedical EngineeringMed-X Research InstituteShanghai Jiao Tong UniversityShanghai200030China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care MedicineShanghai Fifth People’s HospitalFudan UniversityShanghai200240China
- Center of Community-Based Health ResearchFudan UniversityShanghai200240China
| | - Sheng-ce Tao
- Shanghai Center for Systems BiomedicineKey Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200240China
- School of Biomedical EngineeringMed-X Research InstituteShanghai Jiao Tong UniversityShanghai200030China
| |
Collapse
|
30
|
Mukherjee S, Nag S, Mukerjee N, Maitra S, Muthusamy R, Fuloria NK, Fuloria S, Adhikari MD, Anand K, Thorat N, Subramaniyan V, Gorai S. Unlocking Exosome-Based Theragnostic Signatures: Deciphering Secrets of Ovarian Cancer Metastasis. ACS OMEGA 2023; 8:36614-36627. [PMID: 37841156 PMCID: PMC10568589 DOI: 10.1021/acsomega.3c02837] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/21/2023] [Indexed: 10/17/2023]
Abstract
Ovarian cancer (OC) is a common gynecological cancer worldwide. Unfortunately, the lack of early detection methods translates into a substantial cohort of women grappling with the pressing health crisis. The discovery of extracellular vesicles (EVs) (their major subpopulation exosomes, microvesicles, and apoptotic bodies) has provided new insights into the understanding of cancer. Exosomes, a subpopulation of EVs, play a crucial role in cellular communication and reflect the cellular status under both healthy and pathological conditions. Tumor-derived exosomes (TEXs) dynamically influence ovarian cancer progression by regulating uncontrolled cell growth, immune suppression, angiogenesis, metastasis, and the development of drug and therapeutic resistance. In the field of OC diagnostics, TEXs offer potential biomarkers in various body fluids. On the other hand, exosomes have also shown promising abilities to cure ovarian cancer. In this review, we address the interlink between exosomes and ovarian cancer and explore their theragnostic signature. Finally, we highlight future directions of exosome-based ovarian cancer research.
Collapse
Affiliation(s)
- Sayantanee Mukherjee
- Centre
for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, Kerala, India
| | - Sagnik Nag
- Department
of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Road, Tamil Nadu 632014, India
| | - Nobendu Mukerjee
- Department
of Microbiology, West Bengal State University, West Bengal 700126, Kolkata, India
- Department
of Health Sciences, Novel Global Community
Educational Foundation, New South
Wales, Australia
| | - Swastika Maitra
- Department
of Microbiology, Adamas University, West Bengal 700126, Kolkata, India
| | - Raman Muthusamy
- Department
of Microbiology, Centre for Infectious Diseases, Saveetha Dental College, Chennai, Tamil Nadu 600077, India
| | - Neeraj Kumar Fuloria
- Faculty
of Pharmacy, & Centre of Excellence for Biomaterials Engineering, AIMST University, Semeling, Kedah 08100, Malaysia
| | - Shivkanya Fuloria
- Faculty
of Pharmacy, AIMST University, Semeling, Kedah 08100, Malaysia
| | - Manab Deb Adhikari
- Department
of Biotechnology, University of North Bengal
Raja Rammohunpur, Darjeeling, West Bengal 734013, India
| | - Krishnan Anand
- Department
of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Nanasaheb Thorat
- Limerick
Digital Cancer Research Centre and Department of Physics, Bernal Institute, University of Limerick, Castletroy Co. Limerick, Limerick V94T9PX, Ireland
| | - Vetriselvan Subramaniyan
- Jeffrey
Cheah School of Medicine and Health Sciences, Monash University, Malaysia, Jalan Lagoon Selatan, Bandar
Sunway, 47500 Selangor
Darul Ehsan, Malaysia
- Center
for Transdisciplinary Research, Department of Pharmacology, Saveetha
Dental College, Saveetha Institute of Medical
and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Sukhamoy Gorai
- Rush
University Medical Center, 1620 West Harrison Street, Chicago, Illinois 60612, United States
| |
Collapse
|
31
|
Bertrand P. Aptamers Targeting the PD-1/PD-L1 Axis: A Perspective. J Med Chem 2023; 66:10878-10888. [PMID: 37561598 DOI: 10.1021/acs.jmedchem.3c00551] [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: 08/12/2023]
Abstract
Aptamers have emerged in recent years as alternatives to antibodies or small molecules to interfere with the immune check points by blocking the PD-1/PD-L1 interactions and represent an interesting perspective for immuno-oncology. Aptamers are RNA or DNA nucleotides able to bind to a target with high affinity, with the target ranging from small molecules to proteins and up to cells. Aptamers are identified by the SELEX method that can be modified for specific purposes. The range of applications of aptamers covers therapy as well as new alternative assay technologies similar to ELISA. Aptamers' limited plasma stability can be managed using delivery strategies. The goal of this Perspective is to give an overview of the current development of aptamers targeting the most studied immune checkpoint modulators, PD-1 and PD-L1, and analogous strategies with aptamers for other immuno-related targets.
Collapse
Affiliation(s)
- Philippe Bertrand
- University of Poitiers, IC2MP UMR 7285 CNRS, 4 rue Michel Brunet B27, TSA 51106, 86073 Poitiers cedex 9, France
| |
Collapse
|
32
|
Wu YX, Xing S, Wang Y, Tian BY, Wu M, Wang XP, Huang Q, He X, Chen SL, Li XH, Zeng MS, Liu WL. Multiple TMA-aided CRISPR/Cas13a platform for highly sensitive detection of IL-15 to predict immunotherapeutic response in nasopharyngeal carcinoma. J Immunother Cancer 2023; 11:e006552. [PMID: 37536937 PMCID: PMC10401221 DOI: 10.1136/jitc-2022-006552] [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] [Accepted: 07/13/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs)-based treatments have been recommended as the first line for refractory recurrent and/or metastatic nasopharyngeal carcinoma (NPC) patients, yet responses vary, and predictive biomarkers are urgently needed. We selected serum interleukin-15 (sIL-15) out of four interleukins as a candidate biomarker, while most patients' sIL-15 levels were too low to be detected by conventional methods, so it was necessary to construct a highly sensitive method to detect sIL-15 in order to select NPC patients who would benefit most or least from ICIs. METHODS Combining a primer exchange reaction (PER), transcription-mediated amplification (TMA), and a immuno-PER-TMA-CRISPR/Cas13a system, we developed a novel multiple signal amplification platform with a detection limit of 32 fg/mL, making it 153-fold more sensitive than ELISA. RESULTS This platform demonstrated high specificity, repeatability, and versatility. When applied to two independent cohorts of 130 NPC sera, the predictive value of sIL-15 was accurate in both cohorts (area under the curve: training, 0.882; validation, 0.898). Additionally, lower sIL-15 levels were correlated with poorer progression-free survival (training, HR: 0.080, p<0.0001; validation, HR: 0.053, p<0.0001). CONCLUSION This work proposes a simple and sensitive approach for sIL-15 detection to provide insights for personalized immunotherapy of NPC patients.
Collapse
Affiliation(s)
- Ya-Xian Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Shan Xing
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Yu Wang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Bo-Yu Tian
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Meng Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xue-Ping Wang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Qi Huang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xia He
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Shu-Lin Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xiao-Hui Li
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Mu-Sheng Zeng
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Wan-Li Liu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| |
Collapse
|
33
|
Deng F, Li Y, Hall T, Vesey G, Goldys EM. Bi-functional antibody-CRISPR/Cas12a ribonucleoprotein conjugate for improved immunoassay performance. Anal Chim Acta 2023; 1259:341211. [PMID: 37100476 DOI: 10.1016/j.aca.2023.341211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023]
Abstract
Protein conjugates are commonly used in biochemistry, including diagnostic platforms such as antibody-based immunoassays. Antibodies can be bound to a variety of molecules creating conjugates with desirable functions, particularly for imaging and signal amplification. Cas12a is a recently discovered programable nuclease with the remarkable capability to amplify assay signals due to its trans-cleavage property. In this study, we directly conjugated antibody with Cas12a/gRNA ribonucleoprotein without the loss of function in either constituent. The conjugated antibody was suitable for immunoassays and the conjugated Cas12a was capable of amplifying the signal produced in an immunosensor without the need to change the original assay protocol. We applied the bi-functional antibody-Cas12a/gRNA conjugate to successfully detect two different types of targets, a whole pathogenic microorganism, Cryptosporidium, and a small protein, cytokine IFN-γ, with sensitivity reaching one single microorganism per sample and 10 fg/mL for IFN-γ, respectively. With simple substitution of the antibody conjugated with the Cas12a/gRNA RNP, this approach can potentially be applied to increase sensitivity of a variety of immunoassays for a broad range of different analytes.
Collapse
Affiliation(s)
- Fei Deng
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia; ARC Centre of Excellence in Nanoscale Biophotonics, University of New South Wales, Sydney, 2052, Australia.
| | - Yi Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia; ARC Centre of Excellence in Nanoscale Biophotonics, University of New South Wales, Sydney, 2052, Australia.
| | - Tim Hall
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Graham Vesey
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Ewa M Goldys
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia; ARC Centre of Excellence in Nanoscale Biophotonics, University of New South Wales, Sydney, 2052, Australia
| |
Collapse
|
34
|
Li M, Luo N, Liao X, Zou L. Proximity hybridization-regulated CRISPR/Cas12a-based dual signal amplification strategy for sensitive detection of circulating tumor DNA. Talanta 2023; 257:124395. [PMID: 36858011 DOI: 10.1016/j.talanta.2023.124395] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023]
Abstract
Circulating tumor DNA (ctDNA) is regarded as an ideal candidate biomarker for the non-invasive diagnosis of cancer. However, the lack of convenient and reliable detection methods for ctDNA restricts its clinical application. Herein, we developed a dual signal amplification strategy for sensitive detection of ctDNA based on hybridization chain reaction (HCR) and proximity hybridization-regulated CRISPR/Cas12a. The ctDNA initiates HCR through the continuous hybridization of two hairpin probes (H1 and H2), yielding long nicked double-stranded DNA nanowires composed of numerous split segments, which are successively connected to activate the trans-cleavage activity of CRISPR/Cas12a. In this case, the doubly labeled single-stranded DNA reporter can be cleaved to produce a strong fluorescent signal. Owing to the dual amplification of HCR and CRISPR/Cas12a, this strategy exhibits high sensitivity toward ctDNA with a low detection limit of 5.43 fM. Moreover, the proposed method was successfully applied for ctDNA detection in serum samples with satisfactory results, which has great potential in the clinical diagnosis of cancer.
Collapse
Affiliation(s)
- Mengyan Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Nian Luo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaofei Liao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Li Zou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, PR China.
| |
Collapse
|
35
|
Sagoe KO, Kyama MC, Maina N, Kamita M, Njokah M, Thiong'o K, Kanoi BN, Wandera EA, Ndegwa D, Kinyua DM, Gitaka J. Application of Hybridization Chain Reaction/CRISPR-Cas12a for the Detection of SARS-CoV-2 Infection. Diagnostics (Basel) 2023; 13:diagnostics13091644. [PMID: 37175035 PMCID: PMC10178590 DOI: 10.3390/diagnostics13091644] [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: 04/05/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Globally, the emergence of the coronavirus disease (COVID-19) has had a significant impact on life. The need for ongoing SARS-CoV-2 screening employing inexpensive and quick diagnostic approaches is undeniable, given the ongoing pandemic and variations in vaccine administration in resource-constrained regions. This study presents results as proof of concept to use hybridization chain reaction (HCR) and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas12a complex for detecting SARS-CoV-2. HCR hairpin probes were designed using the NUPACK web-based program and further used to amplify the SARS-CoV-2 N gene in archived nasopharyngeal samples. The results were visualized using agarose gels and CRISPR Cas12a-based lateral flow strips. The assay was evaluated using the gold standard, real-time polymerase chain reaction (RT-PCR), as recommended by the World Health Organization (WHO). The results show the comparative efficiency of HCR to RT-PCR. This study shows that HCR and CRISPR are viable alternatives for diagnosing SARS-CoV-2 in samples.
Collapse
Affiliation(s)
- Kate Obaayaa Sagoe
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology and Innovation (PAUSTI), Nairobi P.O. Box 62000-00200, Kenya
| | - Mutinda Cleophas Kyama
- Department of Medical Laboratory Science, College of Health Sciences, Jomo Kenyatta University of Agriculture & Technology, Nairobi P.O. Box 62000-00200, Kenya
| | - Naomi Maina
- Department of Biochemistry, College of Health Sciences, Jomo Kenyatta University of Agriculture & Technology, Nairobi P.O. Box 62000-00200, Kenya
| | - Moses Kamita
- Directorate of Research and Innovation, Mount Kenya University, Thika P.O. Box 342-01000, Kenya
| | - Muturi Njokah
- Department of Biochemistry, College of Health Sciences, Jomo Kenyatta University of Agriculture & Technology, Nairobi P.O. Box 62000-00200, Kenya
| | - Kelvin Thiong'o
- Center for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | - Bernard N Kanoi
- Directorate of Research and Innovation, Mount Kenya University, Thika P.O. Box 342-01000, Kenya
| | - Ernest Apondi Wandera
- Directorate of Research and Innovation, Mount Kenya University, Thika P.O. Box 342-01000, Kenya
- Center for Virus Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | - Davies Ndegwa
- Department of Medical Laboratory Sciences, Kenya Medical Training College, Nairobi P.O. Box 30195-00100, Kenya
| | - Dickson Mwenda Kinyua
- Department of Physical Sciences, Meru University of Science & Technology, Meru P.O. Box 972-60200, Kenya
- Department of Pure and Applied Sciences, Kirinyaga University, Kerugoya P.O. Box 143-10300, Kenya
| | - Jesse Gitaka
- Directorate of Research and Innovation, Mount Kenya University, Thika P.O. Box 342-01000, Kenya
| |
Collapse
|
36
|
Ma L, Liao D, Zhao Z, Kou J, Guo H, Xiong X, Man S. Sensitive Small Molecule Aptasensing based on Hybridization Chain Reaction and CRISPR/Cas12a Using a Portable 3D-Printed Visualizer. ACS Sens 2023; 8:1076-1084. [PMID: 36651835 DOI: 10.1021/acssensors.2c02097] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Next-generation biosensing tools based on CRISPR/Cas have revolutionized the molecular detection. A number of CRISPR/Cas-based biosensors have been reported for the detection of nucleic acid targets. The establishment of efficient methods for non-nucleic acid target detection would further broaden the scope of this technique, but up to now, the concerning research is limited. In the current study, we reported a versatile biosensing platform for non-nucleic acid small-molecule detection called SMART-Cas12a (small-molecule aptamer regulated test using CRISPR/Cas12a). Simply, hybridization chain reaction cascade signal amplification was first trigged by functional nucleic acid (aptamer) through target binding. Then, the CRISPR/Cas system was integrated to recognize the amplified products followed by activation of the trans-cleavage. As such, the target can be ingeniously converted to nucleic acid signals and then fluorescent signals that can be readily visualized and analyzed by a customized 3D-printed visualizer with the help of a home-made App-enabled smartphone. Adenosine triphosphate was selected as a model target, and under the optimized conditions, we achieved fine analytical performance with a linear range from 0.1 to 750 μM and a detection limit of 1.0 nM. The satisfactory selectivity and recoveries that we have obtained further demonstrated this method to be suitable for a complex sample environment. The sample-to-answer time was less than 100 min. Our work not only expanded the reach of the CRISPR-Cas system in biosensing but also provided a prototype method that can be generalized for detecting a wider range of analytes with desirable adaptability, sensitivity, specificity, and on-site capability.
Collapse
Affiliation(s)
- Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Dan Liao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Zhiying Zhao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jun Kou
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Haoyu Guo
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Xin Xiong
- College of Artificial Intelligence, Tianjin University of Science and Technology, Tianjin 3000457, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
37
|
Wang Z, Wei L, Ruan S, Chen Y. CRISPR/Cas12a-Assisted Chemiluminescence Sensor for Aflatoxin B 1 Detection in Cereal Based on Functional Nucleic Acid and In-Pipet Rolling Circle Amplification. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4417-4425. [PMID: 36853759 DOI: 10.1021/acs.jafc.3c00341] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Herein, we report a CRISPR/Cas12a-assisted chemiluminescence sensor for aflatoxin B1 (AFB1) detection based on functional nucleic-acid-mediated target recognition and in-pipet rolling circle amplification-mediated signal amplification. In this sensor, we performed rolling circle amplification on the inside of the pipet to enrich horseradish peroxidase (pipet-poly-HRP). When AFB1 is present, it interacts with functional nucleic acids and results in the release of the activator. The activator is designed to activate the CRISPR/Cas12a system, which cleaves the pipet-poly-HRP to liberate HRP. The freed HRP can then be measured by chemiluminescence to quantify AFB1. This CRISPR/Cas12a-assisted chemiluminescence sensor enables facile, highly sensitive, and specific detection of AFB1, with a linear range from 50 pg/mL to 100 ng/mL and a detection limit of 5.2 pg/mL. Furthermore, it exhibits satisfactory recovery and has successfully challenged AFB1 detection in cereal samples. The proposed sensor offers a novel rapid screening approach that holds great promise for food security monitoring.
Collapse
Affiliation(s)
- Zhilong Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, People's Republic of China
| | - Luyu Wei
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, People's Republic of China
| | - Shilong Ruan
- Daye Public Inspection and Test Center, Daye, Hubei 435100, People's Republic of China
| | - Yiping Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, People's Republic of China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, People's Republic of China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong 518120, People's Republic of China
| |
Collapse
|
38
|
Guo Y, Guo L, Su Y, Xiong Y. CRISPR-Cas system manipulating nanoparticles signal transduction for cancer diagnosis. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1851. [PMID: 36199268 DOI: 10.1002/wnan.1851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/30/2022] [Accepted: 08/10/2022] [Indexed: 11/05/2022]
Abstract
Early diagnosis of cancer is important to improve the survival rate and relieve patient pain. Sensitive detection of cancer related biomarkers in body fluids is a critical approach for the early diagnosis of cancer. The clustered regularly interspaced short palindromic repeat-associated protein (CRISPR-Cas) system has emerged as a molecular manipulation technology because of its simple detection procedure, high base resolution, and isothermal signal amplification. Recently, various nanomaterials with unique optical and electrical characteristics have been introduced as the novel signal transducers to enhance the detection performance of CRISPR-Cas-based nanosensors. This review summarizes the working mechanisms of the CRISPR-Cas system for biosensing. It also enumerates the strategies of CRISPR-manipulated nanosensors based on various signal models for cancer diagnosis, including colorimetric, fluorescence, electrochemical, electrochemiluminescence, pressure, and other signals. Finally, the prospects and challenges of CRISPR-Cas-based nanosensors for cancer diagnostic are also discussed. This article is categorized under: Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Yuqian Guo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China
| | - Liang Guo
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, People's Republic of China
| | - Yu Su
- School of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China.,School of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
39
|
Zhang J, Guan M, Ma C, Liu Y, Lv M, Zhang Z, Gao H, Zhang K. Highly Effective Detection of Exosomal miRNAs in Plasma Using Liposome-Mediated Transfection CRISPR/Cas13a. ACS Sens 2023; 8:565-575. [PMID: 36722721 DOI: 10.1021/acssensors.2c01683] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Exosomal miRNAs play a critical role in cancer biology and could be potential biomarkers for cancer diagnosis. However, due to the low abundance of miRNAs in the exosomes, recognizing and detecting disease-associated exosomal miRNAs in an easy-to-operate way remain a challenge. Herein, we used a liposome-mediated membrane fusion strategy (MFS) to transfect CRISPR/Cas13a into exosomes, termed MFS-CRISPR, directly measuring exosomal miRNAs in plasma. Using the MFS-CRISPR platform for detection of the exosomal miR-21, we achieve a linear range spanning four orders of magnitude (104-108 particles/mL) and the method is able to detect the exosomal miR-21 in as low as 1.2 × 103 particles/mL. The liposome-mediated MFS could confine fluorescent signals in fused vesicles, which can be used for exosome heterogeneity analysis. Moreover, MFS-CRISPR assay was evaluated by measuring clinical samples, and the difference of miR-21 expression of breast cancer patients and healthy donors was significant. Because of high sensitivity and simplicity, the proposed method could have promising clinical potential for cancer diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Junli Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.,Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Mengting Guan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Chihong Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yingying Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Min Lv
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.,Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Hua Gao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.,Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450052, China
| |
Collapse
|
40
|
Kadam US, Cho Y, Park TY, Hong JC. Aptamer-based CRISPR-Cas powered diagnostics of diverse biomarkers and small molecule targets. APPLIED BIOLOGICAL CHEMISTRY 2023; 66:13. [PMID: 36843874 PMCID: PMC9937869 DOI: 10.1186/s13765-023-00771-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 02/03/2023] [Indexed: 06/06/2023]
Abstract
CRISPR-Cas systems have been widely used in genome editing and transcriptional regulation. Recently, CRISPR-Cas effectors are adopted for biosensor construction due to its adjustable properties, such as simplicity of design, easy operation, collateral cleavage activity, and high biocompatibility. Aptamers' excellent sensitivity, specificity, in vitro synthesis, base-pairing, labeling, modification, and programmability has made them an attractive molecular recognition element for inclusion in CRISPR-Cas systems. Here, we review current advances in aptamer-based CRISPR-Cas sensors. We briefly discuss aptamers and the knowledge of Cas effector proteins, crRNA, reporter probes, analytes, and applications of target-specific aptamers. Next, we provide fabrication strategies, molecular binding, and detection using fluorescence, electrochemical, colorimetric, nanomaterials, Rayleigh, and Raman scattering. The application of CRISPR-Cas systems in aptamer-based sensing of a wide range of biomarkers (disease and pathogens) and toxic contaminants is growing. This review provides an update and offers novel insights into developing CRISPR-Cas-based sensors using ssDNA aptamers with high efficiency and specificity for point-of-care setting diagnostics.
Collapse
Affiliation(s)
- Ulhas Sopanrao Kadam
- Division of Life Science and Division of Applied Life Science (BK21 Four), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, Gyeongnam-do 52828 Republic of Korea
| | - Yuhan Cho
- Division of Life Science and Division of Applied Life Science (BK21 Four), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, Gyeongnam-do 52828 Republic of Korea
| | - Tae Yoon Park
- Graduate School of Education, Yonsei University, Seoul, 03722 Republic of Korea
| | - Jong Chan Hong
- Division of Life Science and Division of Applied Life Science (BK21 Four), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, Gyeongnam-do 52828 Republic of Korea
- Division of Plant Sciences, University of Missouri, Columbia, MO 65211 USA
| |
Collapse
|
41
|
Li L, Zhang L, Montgomery KC, Jiang L, Lyon CJ, Hu TY. Advanced technologies for molecular diagnosis of cancer: State of pre-clinical tumor-derived exosome liquid biopsies. Mater Today Bio 2023; 18:100538. [PMID: 36619206 PMCID: PMC9812720 DOI: 10.1016/j.mtbio.2022.100538] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Exosomes are membrane-defined extracellular vesicles (EVs) approximately 40-160 nm in diameter that are found in all body fluids including blood, urine, and saliva. They act as important vehicles for intercellular communication between both local and distant cells and can serve as circulating biomarkers for disease diagnosis and prognosis. Exosomes play a key role in tumor metastasis, are abundant in biofluids, and stabilize biomarkers they carry, and thus can improve cancer detection, treatment monitoring, and cancer staging/prognosis. Despite their clinical potential, lack of sensitive/specific biomarkers and sensitive isolation/enrichment and analytical technologies has posed a barrier to clinical translation of exosomes. This review presents a critical overview of technologies now being used to detect tumor-derived exosome (TDE) biomarkers in clinical specimens that have potential for clinical translation.
Collapse
Affiliation(s)
- Lin Li
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Lili Zhang
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
- HCA Florida Healthcare Westside/Northwest Hospital Internal Medicine, Plantation, Florida, USA
| | - Katelynn C. Montgomery
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, USA
| | - Li Jiang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Tony Y. Hu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, USA
| |
Collapse
|
42
|
Wu J, Xu H, Hu F, Jiang Y, Fan B, Khan A, Sun Y, Di K, Gu X, Shen H, Li Z. CRISPR-Cas and catalytic hairpin assembly technology for target-initiated amplification detection of pancreatic cancer specific tsRNAs. Front Bioeng Biotechnol 2023; 11:1169424. [PMID: 37207121 PMCID: PMC10188930 DOI: 10.3389/fbioe.2023.1169424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/19/2023] [Indexed: 05/21/2023] Open
Abstract
Transfer RNA-derived small RNAs (tsRNAs) tRF-LeuCAG-002 (ts3011a RNA) is a novel class of non-coding RNAs biomarker for pancreatic cancer (PC). Reverse transcription polymerase chain reaction (RT-qPCR) has been unfit for community hospitals that are short of specialized equipment or laboratory setups. It has not been reported whether isothermal technology can be used for detection, because the tsRNAs have rich modifications and secondary structures compared with other non-coding RNAs. Herein, we have employed a catalytic hairpin assembly (CHA) circuit and clustered regularly interspaced short palindromic repeats (CRISPR) to develop an isothermal and target-initiated amplification method for detecting ts3011a RNA. In the proposed assay, the presence of target tsRNA triggers the CHA circuit that transforms new DNA duplexes to activate collateral cleavage activity of CRISPR-associated proteins (CRISPR-Cas) 12a, achieving cascade signal amplification. This method showed a low detection limit of 88 aM at 37 °C within 2 h. Moreover, it was demonstrated for the first time that, this method is less likely to produce aerosol contamination than RT-qPCR by simulating aerosol leakage experiments. This method has good consistency with RT-qPCR in the detection of serum samples and showed great potential for PC-specific tsRNAs point-of-care testing (POCT).
Collapse
Affiliation(s)
- Jie Wu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Hongpan Xu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Fenghua Hu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Yiyue Jiang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Boyue Fan
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Adeel Khan
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education (Southeast University), School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yifan Sun
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Kaili Di
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinrui Gu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Han Shen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
- *Correspondence: Zhiyang Li, ; Han Shen,
| | - Zhiyang Li
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
- *Correspondence: Zhiyang Li, ; Han Shen,
| |
Collapse
|
43
|
Yang T, Li J, Zhang D, Cheng X, Li J, Huang X, Ding S, Tang BZ, Cheng W. Pre-Folded G-Quadruplex as a Tunable Reporter to Facilitate CRISPR/Cas12a-Based Visual Nucleic Acid Diagnosis. ACS Sens 2022; 7:3710-3719. [PMID: 36399094 DOI: 10.1021/acssensors.2c01391] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas12a-based detection strategies with a fluorophore quencher-labeled ssDNA reporter or gold nanoparticle ssDNA reporter have been widely used in point-of-care (POC) molecular diagnostics. However, the potential of these CRISPR/Cas12a strategies for POC molecular diagnostics is often compromised due to the complex labeling, high cost, and low signal-to-noise ratio. Herein, we show a pre-folded G-quadruplex (G4) structure with tunable tolerance to CRISPR/Cas12a trans-cleavage and explore its mechanism. Two G4 structures (i.e., Tel22-10 and G16C) sensitive or tolerant to CRISPR/Cas12a trans-cleavage are designed and used as signal elements to fabricate a label-free visible fluorescent strategy or "signal-on" colorimetric strategy, respectively. These two strategies facilitate an ultrasensitive visual nucleic acid determination of Group B Streptococci with a naked-eye limit of detection of 1 aM. The feasibility of the developed G4-assisted CRISPR/Cas12a strategies for real-world applications is demonstrated in clinical vaginal/anal specimens and further verified by a commercial qPCR assay. This work suggests that the proposed G4 structures with tunable tolerance can act as promising signal reporters in the CRISPR/Cas12a system to enable ultrasensitive visible nucleic acid detection.
Collapse
Affiliation(s)
- Tiantian Yang
- The Centre for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing400016, China
| | - Juan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang330047, China
| | - Decai Zhang
- The Centre for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing400016, China
| | - Xiaoxue Cheng
- The Centre for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing400016, China
| | - Jia Li
- The Centre for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing400016, China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang330047, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing400016, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong518172, China
| | - Wei Cheng
- The Centre for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing400016, China
| |
Collapse
|
44
|
Zhang X, Gao Y, Li J, Yan J, Liu P, Fan X, Song W. A novel TAPP-DHTA COF cathodic photoelectrochemical immunosensor based on CRISPR/Cas12a-induced nanozyme catalytic generation of heterojunction. Electrochim Acta 2022. [DOI: 10.1016/j.electacta.2022.141771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
45
|
Cai A, Yang L, Kang X, Liu J, Wang F, Ji H, Wang Q, Wu M, Li G, Zhou X, Qin Y, Wu L. Target Recognition- and HCR Amplification-Induced In Situ Electrochemical Signal Probe Synthesis Strategy for Trace ctDNA Analysis. BIOSENSORS 2022; 12:bios12110989. [PMID: 36354498 PMCID: PMC9688549 DOI: 10.3390/bios12110989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 05/31/2023]
Abstract
An electrochemical-DNA (E-DNA) sensor was constructed by using DNA metallization to produce an electrochemical signal reporter in situ and hybridization chain reaction (HCR) as signal amplification strategy. The cyclic voltammetry (CV) technique was used to characterize the electrochemical solid-state Ag/AgCl process. Moreover, the enzyme cleavage technique was introduced to reduce background signals and further improve recognition accuracy. On the basis of these techniques, the as-prepared E-DNA sensor exhibited superior sensing performance for trace ctDNA analysis with a detection range of 0.5 fM to 10 pM and a detection limit of 7 aM. The proposed E-DNA sensor also displayed excellent selectivity, satisfied repeatability and stability, and had good recovery, all of which supports its potential applications for future clinical sample analysis.
Collapse
Affiliation(s)
- Aiting Cai
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Luxia Yang
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Xiaoxia Kang
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Jinxia Liu
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Feng Wang
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Haiwei Ji
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Qi Wang
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Mingmin Wu
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Guo Li
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Xiaobo Zhou
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Yuling Qin
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| | - Li Wu
- School of Public Health, Nantong University, No.9 Seyuan Road, Nantong 226019, China
| |
Collapse
|
46
|
Qin P, Chen P, Deng N, Tan L, Yin BC, Ye BC. Switching the Activity of CRISPR/Cas12a Using an Allosteric Inhibitory Aptamer for Biosensing. Anal Chem 2022; 94:15908-15914. [DOI: 10.1021/acs.analchem.2c04315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Peipei Qin
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Pinru Chen
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Nan Deng
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Liu Tan
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Bin-Cheng Yin
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Bang-Ce Ye
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
47
|
Cheng X, Li Y, Kou J, Liao D, Zhang W, Yin L, Man S, Ma L. Novel non-nucleic acid targets detection strategies based on CRISPR/Cas toolboxes: A review. Biosens Bioelectron 2022; 215:114559. [DOI: 10.1016/j.bios.2022.114559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/20/2022] [Accepted: 07/07/2022] [Indexed: 12/26/2022]
|
48
|
Huang R, He L, Jin L, Li Z, He N, Miao W. Recent advancements in DNA nanotechnology-enabled extracellular vesicles detection and diagnosis: A mini review. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
49
|
Wang J, Yang X, Wang X, Wang W. Recent Advances in CRISPR/Cas-Based Biosensors for Protein Detection. Bioengineering (Basel) 2022; 9:512. [PMID: 36290480 PMCID: PMC9598526 DOI: 10.3390/bioengineering9100512] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/19/2022] Open
Abstract
CRISPR is an acquired immune system found in prokaryotes that can accurately recognize and cleave foreign nucleic acids, and has been widely explored for gene editing and biosensing. In the past, CRISPR/Cas-based biosensors were mainly applied to detect nucleic acids in the field of biosensing, and their applications for the detection of other types of analytes were usually overlooked such as small molecules and disease-related proteins. The recent work shows that CRISPR/Cas biosensors not only provide a new tool for protein analysis, but also improve the sensitivity and specificity of protein detections. However, it lacks the latest review to summarize CRISPR/Cas-based biosensors for protein detection and elucidate their mechanisms of action, hindering the development of superior biosensors for proteins. In this review, we summarized CRISPR/Cas-based biosensors for protein detection based on their mechanism of action in three aspects: antibody-assisted CRISPR/Cas-based protein detection, aptamer-assisted CRISPR/Cas-based protein detection, and miscellaneous CRISPR/Cas-based methods for protein detection, respectively. Moreover, the prospects and challenges for CRISPR/Cas-based biosensors for protein detection are also discussed.
Collapse
Affiliation(s)
- Jing Wang
- Collaborative Innovation Center of NPU, Shanghai 201100, China
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China
- Northwestern Polytechnical University Chongqing Technology Innovation Center, Chongqing 400000, China
| | - Xifang Yang
- Collaborative Innovation Center of NPU, Shanghai 201100, China
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China
- Northwestern Polytechnical University Chongqing Technology Innovation Center, Chongqing 400000, China
| | - Xueliang Wang
- Collaborative Innovation Center of NPU, Shanghai 201100, China
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China
- Northwestern Polytechnical University Chongqing Technology Innovation Center, Chongqing 400000, China
| | - Wanhe Wang
- Collaborative Innovation Center of NPU, Shanghai 201100, China
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China
- Northwestern Polytechnical University Chongqing Technology Innovation Center, Chongqing 400000, China
| |
Collapse
|
50
|
Jiang S, Li X, Huang L, Xu Z, Lin J. Prognostic value of PD-1, PD-L1 and PD-L2 deserves attention in head and neck cancer. Front Immunol 2022; 13:988416. [PMID: 36119046 PMCID: PMC9478105 DOI: 10.3389/fimmu.2022.988416] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Head and neck cancer has high heterogeneity with poor prognosis, and emerging researches have been focusing on the prognostic markers of head and neck cancer. PD-L1 expression is an important basis for strategies of immunosuppressive treatment, but whether it has prognostic value is still controversial. Although meta-analysis on PD-L1 expression versus head and neck cancer prognosis has been performed, the conclusions are controversial. Since PD-L1 and PD-L2 are two receptors for PD-1, here we summarize and analyze the different prognostic values of PD-1, PD-L1, and PD-L2 in head and neck cancer in the context of different cell types, tissue localization and protein forms. We propose that for head and neck cancer, the risk warning value of PD-1/PD-L1 expression in precancerous lesions is worthy of attention, and the prognostic value of PD-L1 expression at different subcellular levels as well as the judgment convenience of prognostic value of PD-1, PD-L1, PD-L2 should be fully considered. The PD-L1 evaluation systems established based on immune checkpoint inhibitors (ICIs) are not fully suitable for the evaluation of PD-L1 prognosis in head and neck cancer. It is necessary to establish a new PD-L1 evaluation system based on the prognosis for further explorations. The prognostic value of PD-L1, PD-L2 expression in head and neck cancer may be different for early-stage and late-stage samples, and further stratification is required.
Collapse
Affiliation(s)
- Siqing Jiang
- Department of Comprehensive Chemotherapy/Head and Neck Cancer, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xin Li
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Huang
- Center for Experimental Medicine, Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhensheng Xu
- Department of Oncologic Chemotheraphy, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- *Correspondence: Zhensheng Xu, ; Jinguan Lin,
| | - Jinguan Lin
- Department of Comprehensive Chemotherapy/Head and Neck Cancer, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Zhensheng Xu, ; Jinguan Lin,
| |
Collapse
|