1
|
Jia M, Wang C, Chen M, Dong W, Zhang H, Ou J, Wei Y. TiO 2-coated honeycomb-like super-macroporous silica for high-purity extraction of exosomes from human plasma. Talanta 2025; 293:128098. [PMID: 40215722 DOI: 10.1016/j.talanta.2025.128098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/20/2025] [Accepted: 04/05/2025] [Indexed: 05/14/2025]
Abstract
Screening biomarkers from exosomes has emerged as a new strategy for non-invasive early diagnosis of diseases. Nevertheless, the present screening efficiency and accuracy of biomarkers are limited by the low extraction efficiency and purity of exosomes. To address this issue, a highly selective adsorbent, which integrates size-exclusion and chemisorption, was created by coating TiO2 on honeycomb-like super-macroporous silica. Cell culture medium and human plasma were employed to investigate the enrichment performance, and the results indicate that the super-macropores ranging from 63.5 to 147.5 nm together with thin pore walls allow exosomes to enter and be adsorbed by TiO2 in the pores, enhancing the available surface area for exosomes meanwhile physically excluding the large-sized cell debris and vesicles. Taking advantages of these properties, the prepared adsorbent achieves a higher extraction efficiency, recovery and purity of exosomes compared with the normal adsorbents and ultracentrifugation (UC) method. Combining this method with proteomic analysis, a total of 392 proteins were identified in exosomes from healthy human plasma, which is significantly higher than the number obtained by UC (200 proteins). For clinical samples, 59 upregulated and 124 downregulated proteins were identified in the plasma from colorectal cancer (CRC) patients, of which 44 upregulated proteins and 69 downregulated proteins are strongly associated with the progression of CRC. These findings suggest that this adsorbent possesses considerable potential in the extraction of exosomes for screening biomarkers and diagnosing tumor progress.
Collapse
Affiliation(s)
- Mengqian Jia
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Chenyang Wang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Wenzhuo Dong
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Junjie Ou
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Yinmao Wei
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
2
|
Wu Y, Qiao Y, Yang C, Chen Y, Shen X, Deng C, Yao Q, Sun N. Accelerated Exosomal Metabolic Profiling Enabled by Robust On-Target Array Sintering with Metal-Organic Frameworks. SMALL METHODS 2025; 9:e2401238. [PMID: 39263996 DOI: 10.1002/smtd.202401238] [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: 08/07/2024] [Revised: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Pancreatic cancer is highly lethal, and survival chances improve only with early detection at a precancerous stage. However, there remains a significant gap in developing tools for large-scale, rapid screening. To this end, a high-throughput On-Target Array Extraction Platform (OTAEP) by direct sintering of a series of metal-organic frameworks (MOFs) for dual in situ extraction, encompassing both exosomes and their metabolic profiles, is developed. Based on the principle of geometry-dependent photothermal conversion efficiency and standard testing, the appropriate MOF functional unit is identified. This unit enables exosome enrichment within 10 min and metabolic fingerprint extraction in under 1 s of laser irradiation, with over five reuse. To further accelerate and enhance the quality of metabolic profile analysis, the application of Surrogate Variable Analysis to eliminate hidden confounding factors within the profiles is proposed, and five biomarkers demonstrated by MS/MS experiments are identified. These biomarkers enable early diagnosis, risk stratification, and staging of pancreatic cancer simultaneously, with sensitivity of 94.1%, specificity of 98.8%, and precision of 94.9%. This work represents a breakthrough for overcoming throughput challenges in large-scale testing and for addressing confounding factors in big data analysis.
Collapse
Affiliation(s)
- Yun Wu
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yiming Qiao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chenyu Yang
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yueying Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chunhui Deng
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, P. R. China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
3
|
Cao Y, Qin Y, Cheng Q, Zhong J, Han B, Li Y. Bifunctional nanomaterial enabled high-specific isolation of urinary exosomes for cervical cancer metabolomics analysis and biomarker discovery. Talanta 2025; 285:127280. [PMID: 39613490 DOI: 10.1016/j.talanta.2024.127280] [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: 10/18/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
Cervical cancer (CC) remains a critical public health issue, highlighting the importance of early detection. However, current methods such as cytological and HPV testing face challenges of invasiveness and low patient compliance. Exosomes, emerging as crucial in cancer diagnosis, offer promise due to their noninvasive, highly specificity, and abundant biomarkers. However, isolating exosomes efficiently remains challenging. In this study, we designed and synthesized a bifunctional affinity nanomaterial Fe3O4 @CD63-CLIKKPF, based on the synergistic interaction between its modified aptamer CD63 and peptide CLIKKPF, and CD63 protein and PS of exosomes which can achieve high specificity and high yield separation of urinary exosomes. Notably, the co-modified aptamer CD63 and peptide CLIKKPF not only enable efficient exosome isolation by leveraging dual-affinity mechanisms through a synergistic "AND" logic analysis, but also could be achieved on the Fe3O4 in one-step reaction at room temperature via Fe-S bonding. Combined with LC-MS/MS, we conducted exosome metabolomics analysis in healthy individuals and CC patients across various stages, and machine learning models demonstrated accurate classification (accuracy >0.822) and prediction capabilities for CC. Furthermore, six key metabolites indicative of CC progression were identified and validated in additional patient samples, highlighting their potential as biomarkers. Overall, this study establishes a novel method for exosome metabolomics in CC, offering insights for non-invasive early diagnosis and progression prediction on a large scale.
Collapse
Affiliation(s)
- Yiqing Cao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yulin Qin
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Qunxian Cheng
- Department of Gynecology and Obstetrics, Minhang Hospital, Fudan University, Shanghai, China
| | - Jialiang Zhong
- National Key Laboratory of Lead Druggability Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China.
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201100, China.
| | - Yan Li
- Department of Pharmaceutical Analysis, School of Pharmacy, Fudan University, Shanghai, 201203, China; Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, 201203, China; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
4
|
Zhan J, Wang S, Li X, Zhang J. Molecular engineering of functional DNA molecules toward point-of-care diagnostic devices. Chem Commun (Camb) 2025; 61:4316-4338. [PMID: 39998439 DOI: 10.1039/d5cc00338e] [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: 02/26/2025]
Abstract
The pursuit of rapid, sensitive, and specific diagnostic methodologies is imperative across diverse applications, including the detection of pathogens and disease biomarkers, food safety testing and environmental monitoring. Point-of-care testing (POCT) is characterized by its portability, ease of use, rapidity, and affordability, emerging as an attractive alternative for traditional diagnostics. Over recent years, the incorporation of functional DNA (fDNA) into POC diagnostic devices has emerged as a groundbreaking advancement, significantly enhancing sensitivity, specificity, and user-friendliness. In this review, we explore the innovative applications of fDNA in POC devices, highlighting its potential to revolutionize diagnostics by providing rapid, portable, and precise solutions. We discuss the unique advantages of fDNA, including its stability in complex biological matrices and its ability to recognize a wide range of targets. Furthermore, we explore the potential synergy between fDNA and cutting-edge technologies, such as nanotechnology and artificial intelligence (AI), to forge a path toward more personalized and accessible healthcare solutions. Despite significant progress, challenges remain in translating these innovations from the bench to the clinic. This review aims to provide a comprehensive overview of the current status of fDNA-based POCT devices and future directions for their development, emphasizing their critical role in meeting the global demand for accessible, efficient, and precise diagnostic solutions.
Collapse
Affiliation(s)
- Jiayin Zhan
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun 130022, China.
| | - Siyuan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China.
| | - Xiang Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China.
| | - Jingjing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China.
| |
Collapse
|
5
|
Chen Y, Zhang M, Qi Y, Lin Y, Liu S, Deng C, Jiang S, Sun N. Efficient extraction via titanium organic frameworks facilitates in-depth profiling of urinary exosome metabolite fingerprints. Anal Bioanal Chem 2025; 417:1543-1555. [PMID: 39853354 DOI: 10.1007/s00216-025-05741-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Urinary exosome metabolite analysis has demonstrated notable advantages in uncovering disease status, yet its potential in decoding the intricacies of clear cell renal cell carcinoma (ccRCC) remains untapped. To address this, a core-shell magnetic titanium organic framework was designed to capture urinary exosomes and assist laser desorption/ionization mass spectrometry (LDI MS) to decipher the exosomal metabolic profile of ccRCC, with high sensitivity, throughput, and speed. A total of 492 urinary exosome metabolite fingerprints (UEMFs) from 176 samples were extracted for exploring the differences between ccRCC and healthy individuals. Leveraging machine learning algorithms, the exosomal metabolic profile was disclosed, achieving accurate differentiation and prediction of ccRCC patients versus healthy individuals, with an accuracy exceeding 97.3%. Furthermore, an optimized algorithm panel comprising five key features demonstrated consistent and high diagnosing accuracy rates of over 94.0% both in the training and blind test sets for ccRCC, underscoring the remarkable effectiveness and superiority of this strategy in ccRCC detection. This study not only refines the LDI MS method for metabolite analysis in urinary exosomes but also introduces a promising technical approach for unraveling the mysteries of ccRCC.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Man Zhang
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China
| | - Yiwen Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shasha Liu
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China.
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China.
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Jiang X, Tao L, Cao S, Xu Z, Zheng S, Zhang H, Xu X, Qu X, Liu X, Yu J, Chen X, Wu J, Liang X. Porous Silicon Particle-Assisted Mass Spectrometry Technology Unlocks Serum Metabolic Fingerprints in the Progression From Chronic Hepatitis B to Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5893-5908. [PMID: 39812132 DOI: 10.1021/acsami.4c17563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy and generally develops from liver cirrhosis (LC), which is primarily caused by the chronic hepatitis B (CHB) virus. Reliable liquid biopsy methods for HCC screening in high-risk populations are urgently needed. Here, we establish a porous silicon-assisted laser desorption ionization mass spectrometry (PSALDI-MS) technology to profile metabolite information hidden in human serum in a high throughput manner. Serum metabolites can be captured in the pore channel of APTES-modified porous silicon (pSi) particles and well-preserved during storage or transportation. Furthermore, serum metabolites captured in the APTES-pSi particles can be directly detected on the LDI-MS without the addition of an organic matrix, thus greatly accelerating the acquisition of metabolic fingerprints of serum samples. The PSALDI-MS displays the capability of high throughput (5 min per 96 samples), high reproducibility (coefficient of variation <15%), high sensitivity (LOD ∼ 1 pmol), and high tolerance to background salt and proteins. In a multicenter cohort study, 1433 subjects including healthy controls (HC), CHB, LC, and HCC volunteers were enrolled and nontargeted serum metabolomic analysis was performed on the PSALDI-MS platform. After the selection of feature metabolites, a stepwise diagnostic model for the classification of different liver disease stages was constructed by the machine learning algorithm. In external testing, the accuracy of 91.2% for HC, 71.4% for CHB, 70.0% for LC, and 95.3% for HCC was achieved by chemometrics. Preliminary studies indicated that the diagnostic model constructed from serum metabolic fingerprint also displays good predictive performance in a prospective observation. We believe that the combination of PSALDI-MS technology and machine learning may serve as an efficient tool in clinical practice.
Collapse
Affiliation(s)
- Xinrong Jiang
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Liye Tao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Shuo Cao
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhengao Xu
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Shuang Zheng
- Taizhou First People's Hospital, Taizhou, Zhejiang 318020, China
| | - Huafang Zhang
- Wuyi First People's Hospital, Jinhua, Zhejiang 321200, China
| | - Xinran Xu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xuetong Qu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xingyue Liu
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiekai Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Xiaoming Chen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Well-healthcare Technologies Co., Hangzhou, Zhejiang 310051, China
| | - Jianmin Wu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Institution of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Xiao Liang
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- School of medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
7
|
Xu K, Feng H, Zhao R, Huang Y. Targeting Tetraspanins at Cell Interfaces: Functional Modulation and Exosome-Based Drug Delivery for Precise Disease Treatment. ChemMedChem 2025; 20:e202400664. [PMID: 39415492 DOI: 10.1002/cmdc.202400664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/18/2024]
Abstract
Tetraspanins are key players in various physiological and pathological processes, including malignancy, immune response, fertilization, and infectious disease. Affinity ligands targeting the interactions between tetraspanins and partner proteins are promising for modulating downstream signaling pathways, thus emerging as attractive candidates for interfering related biological functions. Due to the involvement in vesicle biogenesis and cargo trafficking, tetraspanins are also regarded as exosome markers, and become molecular targets for drug loading and delivery. Given the rapid development in these areas, this minireview focuses on recent advances in design and engineering of affinity binders toward tetraspanins including CD63, CD81, and CD9. Their mechanism of actions in modulating protein interactions at cell interfaces and treatment of malignant diseases are discussed. Strategies for constructing exosome-based drug delivery platforms are also reviewed, with emphasis on the important roles of tetraspanins and the affinity ligands. Finally, challenges and future development of tetraspanin-targeting therapy and exosomal drug delivery platforms are also discussed.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
8
|
Yang C, Chen H, Wu Y, Shen X, Liu H, Liu T, Shen X, Xue R, Sun N, Deng C. Deep Learning-Enabled Rapid Metabolic Decoding of Small Extracellular Vesicles via Dual-Use Mass Spectroscopy Chip Array. Anal Chem 2025; 97:271-280. [PMID: 39711466 DOI: 10.1021/acs.analchem.4c04106] [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: 12/24/2024]
Abstract
The increasing focus of small extracellular vesicles (sEVs) in liquid biopsy has created a significant demand for streamlined improvements in sEV isolation methods, efficient collection of high-quality sEV data, and powerful rapid analysis of large data sets. Herein, we develop a high-throughput dual-use mass spectroscopic chip array (DUMSCA) for the rapid isolation and detection of plasma sEVs. The DUMSCA realizes more than a 50% increase in speed compared to traditional method and confirms proficiency in robust storage, reuse, high-efficiency desorption/ionization, and metabolite quantification. With the collected metabolic data matrix of sEVs, a deep learning model achieves high-performance diagnosis of Crohn's disease. Furthermore, discovered biomarkers by feature sparsification and tandem mass spectrometry experiments also exhibited remarkable performance in diagnosis. This work demonstrates the rapidity and validity of DUMSCA for disease diagnosis, enabling the diagnosis of diseases without the necessity for prior knowledge and providing a high-throughput technology for sEV-based liquid biopsy that will empower its vigorous development.
Collapse
Affiliation(s)
- Chenyu Yang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Department of Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - He Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yun Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Department of Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Xiangguo Shen
- Department of Gastroenterology and Hepatology, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch Fudan University), Shanghai 200940, China
| | - Hongchun Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology and Hepatology, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch Fudan University), Shanghai 200940, China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Department of Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
9
|
Yan H, Abdulla A, Wang A, Ding S, Zhang M, Zhang Y, Zhuang TY, Wu L, Wang Y, Ren R, Jiang L, Ding X. Time-Lapse Acquisition of Both Freely Secreted Proteome and Exosome Encapsulated Proteome in Live Organoids' Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406509. [PMID: 39573935 PMCID: PMC11727246 DOI: 10.1002/advs.202406509] [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: 06/12/2024] [Revised: 10/24/2024] [Indexed: 01/14/2025]
Abstract
Proteomic communications in neighboring microenvironments during early organ development is a dynamic process that continuously reshapes human embryonic stem cells (hESCs) developmental fate. Such dynamic proteomic alteration in the microenvironment consists of both freely secreted proteome and exosome-encapsulated proteome. Simultaneous monitoring of the time-lapse shift of both proteomes with live organoids remains technically challenging. Here, a continuous organoid secretion/encapsulation proteome tandem LC-MS/MS (COSEP-LCM) is introduced, which permits time-lapse monitoring of proteomic alterations both in free secretion form and in exosome encapsulated form at live organoids' microenvironment. Continuous growth of human cerebral organoids (COs) and free-secretion/exosome-encapsulation proteomics acquisition with COSEP-LCM for 60 days is demonstrated. SERPINF1, F5, and EFNB1 are initially enriched inside exosomes as encapsulated excretion and then gradually enriched outside exosomes as freely secreted excretion, while C3 is initially enriched outside exosomes as freely secreted excretion and gradually enriched inside exosomes as encapsulated excretion. Such dynamic excretion pattern paradigm shift may imply critical developmental strategy evolution during early human cerebral development. COSEP-LCM offers a platform technique for continuous inside/outside exosome proteomics co-analysis in live organoids' microenvironment.
Collapse
Affiliation(s)
- Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Shuyu Ding
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Manlin Zhang
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Yizhi Zhang
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Tsz Yui Zhuang
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Leqi Wu
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Rongrong Ren
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalShanghai Jiaotong University School of MedicineShanghai200092P. R. China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| |
Collapse
|
10
|
Yang H, Wu P, Li B, Huang X, Shi Q, Qiao L, Liu B, Chen X, Fang X. Diagnosis and Biomarker Screening of Endometrial Cancer Enabled by a Versatile Exosome Metabolic Fingerprint Platform. Anal Chem 2024; 96:17679-17688. [PMID: 39440888 DOI: 10.1021/acs.analchem.4c03726] [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: 10/25/2024]
Abstract
Exosomes have emerged as a revolutionary tool for liquid biopsy (LB), as they carry specific cargo from cells. Profiling the metabolites of exosomes is crucial for cancer diagnosis and biomarker discovery. Herein, we propose a versatile platform for exosomal metabolite assay of endometrial cancer (EC). The platform is based on a nanostructured composite material comprising gold nanoparticle-coated magnetic COF with aptamer modification (Fe3O4@COF@Au-Apt). The unique design and novel synthesis strategy of Fe3O4@COF@Au-Apt provide the material with a large specific surface area, enabling the efficient and specific isolation of exosomes. The exosomes captured Fe3O4@COF@Au-Apt can be directly used as the laser desorption/ionization mass spectrometry (LDI-MS) matrix for rapid exosomal metabolic patterns. By integrating these functionalities into a single platform, the analytical process is simplified, eliminating the need for additional elution steps and minimizing potential sample loss, resulting in large-scale exosomal metabolic fingerprints. Combining with machine learning algorithms on the metabolic patterns, accurate discrimination between endometrial patients (EGs) and benign controls (CGs) was achieved, and the area under the receiver operating characteristic curve of the blind test cohort was 0.924. Confusion matrix analysis of important metabolic fingerprint features further demonstrates the high accuracy of the proposed approach toward EC diagnosis, with an overall accuracy of 94.1%. Moreover, four metabolites, namely, hydroxychalcone, l-acetylcarnitine, elaidic acid, and glutathione, have been identified as potential biomarkers of EC. These results highlight the great value of the integrated exosome metabolic fingerprint platform in facilitating low-cost and high-throughput characterization of exosomal metabolites for cancer diagnosis and biomarker discovery.
Collapse
Affiliation(s)
- Haonan Yang
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Pengfei Wu
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Binxiao Li
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Qian Shi
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Liang Qiao
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| | - Xiaojun Chen
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
- Shanghai Tenth People's Hospital of Tongji University, Shanghai 200000, China
| | - Xiaoni Fang
- Department of Chemistry, Shanghai Stomatological Hospital, Obstetrics and Gynecology Hospital of Fudan University, and School of Pharmacy, Fudan University, Shanghai 200000, China
| |
Collapse
|
11
|
Seo SB, Lim J, Kim K, Maeng I, Rho HW, Son HY, Kim E, Jang E, Kang T, Jung J, Oh SJ, Huh YM, Lim EK. Nucleic Acid Amplification Circuit-Based Hydrogel (NACH) Assay for One-Step Detection of Metastatic Gastric Cancer-Derived Exosomal miRNA. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407621. [PMID: 39308180 DOI: 10.1002/advs.202407621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/23/2024] [Indexed: 11/22/2024]
Abstract
Gastric cancer (GC) is recognized as the fifth most prevalent malignant tumor worldwide. It is characterized by diverse clinical symptoms, treatment responses, and prognoses. In GC prognosis, the promotion of epithelial-mesenchymal transition (EMT) fosters cancer cell invasion and metastasis, thereby triggering the dissemination of tumor cells. This study proposes a nucleic acid amplification circuit-based hydrogel (NACH) assay for identifying exosomal miRNA derived from metastatic GC. The NACH assay employs the rolling circle amplification method and targets miRNA-21, a tumor-related oncogene, and miRNA-99a, which promotes EMT. Specific amplification probes for each target are immobilized within the hydrogel, enabling a streamlined, one-step amplification reaction. The NACH assay exhibits a detection limit of 1 fm for miRNA-21 and miRNA-99a, thereby enabling rapid and highly sensitive on-site detection. Performance evaluation using exosomal miRNA extracted from cell culture media, mouse plasma, and human plasma revealed fluorescence intensity patterns similar to those obtained in qRT-PCR. Furthermore, deploying a custom-developed portable fluorometer for the NACH assay allows for diagnostic performance assessment and point-of-care testing using clinical samples from GC patients. These findings emphasize the potential of the NACH assay to be used as a robust tool for the genetic diagnosis of GC based on exosome detection.
Collapse
Grants
- 2021M3H4A1A02051048 Ministry of Science and ICT, South Korea
- 2023R1A2C2005185 Ministry of Science and ICT, South Korea
- 2021M3E5E3080844 Ministry of Science and ICT, South Korea
- 2022R1C1C1008815 Ministry of Science and ICT, South Korea
- 2022R1A2C2007490 Ministry of Science and ICT, South Korea
- RS-2024-00348576 Ministry of Science and ICT, South Korea
- RS-2024-00438316 Ministry of Science and ICT, South Korea
- RS-2024-00339077 Ministry of Science and ICT, South Korea
- RS-2022-II221044 Ministry of Science and ICT, South Korea
- RS-2022-00154853 Ministry of Trade, Industry and Energy, South Korea
- RS-2024-00403563 Ministry of Trade, Industry and Energy, South Korea
- RS-2024-00432382 Ministry of Trade, Industry and Energy, South Korea
- CPS23101-100 National Research Council of Science & Technology
- KGM5472413 Korea Research Institute of Bioscience and Biotechnology
- 2021003370003 Korea Environmental Industry and Technology Institute
- Nanomedical Devices Development Program of National Nano Fab Center
Collapse
Affiliation(s)
- Seung Beom Seo
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Cogno-Mechatronics Engineering, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jaewoo Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Medical Device Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Chungcheongbuk-do, 28160, Republic of Korea
| | - Kyujung Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Pusan, 46241, Republic of Korea
| | - Inhee Maeng
- YUHS-KRIBB Medical Convergence Research Institute, Yonsei University, Seoul, 03772, Republic of Korea
| | - Hyun Wook Rho
- Department of Radiology, Yonsei University, Seoul, 03772, Republic of Korea
| | - Hye Young Son
- YUHS-KRIBB Medical Convergence Research Institute, Yonsei University, Seoul, 03772, Republic of Korea
- Department of Radiology, Yonsei University, Seoul, 03772, Republic of Korea
| | - Eunjung Kim
- Department of Bioengineering & Nano-bioengineering, Research Center for Bio Materials and Process Development, Incheon National University, Incheon, 22012, Republic of Korea
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Eunji Jang
- MediBio-Informatics Research Center, Novomics Co., Ltd., Seoul, 07217, Republic of Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Juyeon Jung
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, UST, Daejeon, 34113, Republic of Korea
| | - Seung Jae Oh
- YUHS-KRIBB Medical Convergence Research Institute, Yonsei University, Seoul, 03772, Republic of Korea
| | - Yong-Min Huh
- YUHS-KRIBB Medical Convergence Research Institute, Yonsei University, Seoul, 03772, Republic of Korea
- Department of Radiology, Yonsei University, Seoul, 03772, Republic of Korea
- Department of Biochemistry & Molecular Biology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eun-Kyung Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, UST, Daejeon, 34113, Republic of Korea
| |
Collapse
|
12
|
Wu G, Zhang Y, Jia S, Qi X, Feng X, Ren Y, Lu X, Hu L. Preparation of Dysprosium(III)-Metal Organic Framework Nanofiber for Exosome Capture and Biomarker Discovery toward Liver Disease. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56874-56883. [PMID: 39393007 DOI: 10.1021/acsami.4c14045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
As an emerging source for liquid biopsy, exosomes hold significant promise for clinical diagnosis. However, commonly used exosome isolation methods (e.g., ultracentrifugation) suffer from low throughput for a large number of clinical samples. Herein, a dysprosium-metal organic framework was synthesized and doped with nanofibers by electrospinning for efficient capture of exosomes from body fluid. With the integration of multichannel of pipet or robot automatic workstation, high throughput exosome isolation can be achieved with clinical samples with high reproducibility. To evaluate the clinical value of the developed method, urinary exosomes were enriched from 34 liver disease samples of different stages for the profiling of metabolites by mass spectrometry. The results showed that HCC, cirrhosis, and healthy controls can be significantly differentiated by the Random Forest classification model. The dysprosium-metal organic framework has promising applications in exosome-based liquid biopsy for large-scale clinical disease diagnosis.
Collapse
Affiliation(s)
- Guangyao Wu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yue Zhang
- Alan G. MacDiarmid Institute, College of Chemistry, Jilin University, Changchun 130012, China
| | - Shengnan Jia
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun 130041, China
| | - Xiulei Qi
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Feng
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yujuan Ren
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xiaofeng Lu
- Alan G. MacDiarmid Institute, College of Chemistry, Jilin University, Changchun 130012, China
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
13
|
Tang H, Yu D, Zhang J, Wang M, Fu M, Qian Y, Zhang X, Ji R, Gu J, Zhang X. The new advance of exosome-based liquid biopsy for cancer diagnosis. J Nanobiotechnology 2024; 22:610. [PMID: 39380060 PMCID: PMC11463159 DOI: 10.1186/s12951-024-02863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Liquid biopsy is a minimally invasive method that uses biofluid samples instead of tissue samples for cancer diagnosis. Exosomes are small extracellular vesicles secreted by donor cells and act as mediators of intercellular communication in human health and disease. Due to their important roles, exosomes have been considered as promising biomarkers for liquid biopsy. However, traditional methods for exosome isolation and cargo detection methods are time-consuming and inefficient, limiting their practical application. In the past decades, many new strategies, such as microfluidic chips, nanowire arrays and electrochemical biosensors, have been proposed to achieve rapid, accurate and high-throughput detection and analysis of exosomes. In this review, we discussed about the new advance in exosome-based liquid biopsy technology, including isolation, enrichment, cargo detection and analysis approaches. The comparison of currently available methods is also included. Finally, we summarized the advantages and limitations of the present strategies and further gave a perspective to their future translational use.
Collapse
Affiliation(s)
- Haozhou Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Dan Yu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Min Fu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoxin Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Runbi Ji
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China.
- Affiliated Cancer Hospital of Nantong University, Nantong, 226300, China.
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
14
|
Su Y, Chen M, Xu W, Gu P, Fan X. Advances in Extracellular-Vesicles-Based Diagnostic and Therapeutic Approaches for Ocular Diseases. ACS NANO 2024; 18:22793-22828. [PMID: 39141830 PMCID: PMC11363148 DOI: 10.1021/acsnano.4c08486] [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: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles of various sizes that can be secreted by most cells. EVs contain a diverse array of cargo, including RNAs, lipids, proteins, and other molecules with functions of intercellular communication, immune modulation, and regulation of physiological and pathological processes. The biofluids in the eye, including tears, aqueous humor, and vitreous humor, are important sources for EV-based diagnosis of ocular disease. Because the molecular cargos may reflect the biology of their parental cells, EVs in these biofluids, as well as in the blood, have been recognized as promising candidates as biomarkers for early diagnosis of ocular disease. Moreover, EVs have also been used as therapeutics and targeted drug delivery nanocarriers in many ocular disorders because of their low immunogenicity and superior biocompatibility in nature. In this review, we provide an overview of the recent advances in the field of EV-based studies on the diagnosis and therapeutics of ocular disease. We summarized the origins of EVs applied in ocular disease, assessed different methods for EV isolation from ocular biofluid samples, highlighted bioengineering strategies of EVs as drug delivery systems, introduced the latest applications in the diagnosis and treatment of ocular disease, and presented their potential in the current clinical trials. Finally, we briefly discussed the challenges of EV-based studies in ocular disease and some issues of concern for better focusing on clinical translational studies of EVs in the future.
Collapse
Affiliation(s)
- Yun Su
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Moxin Chen
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Wei Xu
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Xianqun Fan
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
15
|
He Y, Zeng X, Xiong Y, Shen C, Huang K, Chen P. Portable Aptasensor Based on Parallel Rolling Circle Amplification for Tumor-Derived Exosomes Liquid Biopsy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403371. [PMID: 38923850 PMCID: PMC11348067 DOI: 10.1002/advs.202403371] [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: 04/03/2024] [Revised: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Here, a separation-free and label-free portable aptasensor is developed for rapid and sensitive analysis of tumor-derived exosomes (TEXs). It integrated a parallel rolling circle amplification (RCA) reaction, selective binding of metal ions or small molecules to nucleic acid-specific conformations, and a low-cost, highly sensitive handheld fluorometer. Lung cancer, for example, is targeted with two typical biomarkers (mucin 1 and programmed cell death ligand 1 (PD-L1)) on its exosomes. The affinity of aptamers to the targets modulated the amount of RCA products (T-Hg2+-T and cytosine (C)-rich single-stranded DNA), which in turn affected the fluorescence intensity of quantum dots (QDs) and methylene blue (MB). The results revealed that the limit of detection (LOD) of the handheld fluorometer for cell-derived exosomes can be as low as 30 particles mL-1. Moreover, its specificity, sensitivity, and area under the curve (AUC) are 93% (14/15), 92% (23/25), and 0.956, as determined by the analysis of 40 clinical samples. Retesting 16 of these samples with the handheld fluorometer yielded strong concordance between the fluorometer results and those acquired from clinical computed tomography (CT) and pathology.
Collapse
Affiliation(s)
- Yaqin He
- Department of Laboratory MedicineMed+X Center for ManufacturingNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Xianghu Zeng
- Department of Laboratory MedicineMed+X Center for ManufacturingNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Ying Xiong
- Department of Laboratory MedicineMed+X Center for ManufacturingNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Congcong Shen
- Department of Laboratory MedicineMed+X Center for ManufacturingNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Ke Huang
- College of Chemistry and Material ScienceSichuan Normal UniversityChengduSichuan610068China
| | - Piaopiao Chen
- Department of Laboratory MedicineMed+X Center for ManufacturingNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuan610041China
| |
Collapse
|
16
|
Liu X, Jia Y, Zheng C. Recent progress in Surface-Enhanced Raman Spectroscopy detection of biomarkers in liquid biopsy for breast cancer. Front Oncol 2024; 14:1400498. [PMID: 39040452 PMCID: PMC11260621 DOI: 10.3389/fonc.2024.1400498] [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: 03/13/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women globally and a leading cause of cancer-related mortality. However, current detection methods, such as X-rays, ultrasound, CT scans, MRI, and mammography, have their limitations. Recently, with the advancements in precision medicine and technologies like artificial intelligence, liquid biopsy, specifically utilizing Surface-Enhanced Raman Spectroscopy (SERS), has emerged as a promising approach to detect breast cancer. Liquid biopsy, as a minimally invasive technique, can provide a temporal reflection of breast cancer occurrence and progression, along with a spatial representation of overall tumor information. SERS has been extensively employed for biomarker detection, owing to its numerous advantages such as high sensitivity, minimal sample requirements, strong multi-detection ability, and controllable background interference. This paper presents a comprehensive review of the latest research on the application of SERS in the detection of breast cancer biomarkers, including exosomes, circulating tumor cells (CTCs), miRNA, proteins and others. The aim of this review is to provide valuable insights into the potential of SERS technology for early breast cancer diagnosis.
Collapse
Affiliation(s)
- Xiaobei Liu
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yining Jia
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Chao Zheng
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| |
Collapse
|
17
|
Zheng L, Li J, Li Y, Sun W, Ma L, Qu F, Tan W. Empowering Exosomes with Aptamers for Precision Theranostics. SMALL METHODS 2024:e2400551. [PMID: 38967170 DOI: 10.1002/smtd.202400551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/04/2024] [Indexed: 07/06/2024]
Abstract
As information messengers for cell-to-cell communication, exosomes, typically small membrane vesicles (30-150 nm), play an imperative role in the physiological and pathological processes of living systems. Accumulating studies have demonstrated that exosomes are potential biological candidates for theranostics, including liquid biopsy-based diagnosis and drug delivery. However, their clinical applications are hindered by several issues, especially their unspecific detection and insufficient targeting ability. How to upgrade the accuracy of exosome-based theranostics is being widely explored. Aptamers, benefitting from their admirable characteristics, are used as excellent molecular recognition elements to empower exosomes for precision theranostics. With high affinity against targets and easy site-specific modification, aptamers can be incorporated with platforms for the specific detection of exosomes, thus providing opportunities for advancing disease diagnostics. Furthermore, aptamers can be tailored and functionalized on exosomes to enable targeted therapeutics. Herein, this review emphasizes the empowering of exosomes by aptamers for precision theranostics. A brief introduction of exosomes and aptamers is provided, followed by a discussion of recent progress in aptamer-based exosome detection for disease diagnosis, and the emerging applications of aptamer-functionalized exosomes for targeted therapeutics. Finally, current challenges and opportunities in this research field are presented.
Collapse
Affiliation(s)
- Liyan Zheng
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/ Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Jin Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yingying Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/ Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Weidi Sun
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/ Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - LeLe Ma
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Fengli Qu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
| | - Weihong Tan
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
18
|
Qin X, Xiang Y, Mao L, Yang Y, Wei B, Lu H, Li X, Zhang Y, Yang F. Buoyant Metal-Organic Framework Corona-Driven Fast Isolation and Ultrasensitive Profiling of Circulating Extracellular Vesicles. ACS NANO 2024; 18:14569-14582. [PMID: 38781132 DOI: 10.1021/acsnano.4c02339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Accurately assaying tumor-derived circulating extracellular vesicles (EVs) is fundamental in noninvasive cancer diagnosis and therapeutic monitoring but limited by challenges in efficient EV isolation and profiling. Here, we report a bioinspired buoyancy-driven metal-organic framework (MOF) corona that leverages on-bubble coordination and dual-encoded surface-enhanced Raman scattering (SERS) nanotags to streamline rapid isolation and ultrasensitive profiling of plasma EVs in a single assay for cancer diagnostics. This integrated bubble-MOF-SERS EV assay (IBMsv) allows barnacle-like high-density adhesion of MOFs on a self-floating bubble surface to enable fast isolation (2 min, near 90% capture efficiency) of tumor EVs via enhanced EV-MOF binding. Also, IBMsv harnesses four-plexed SERS nanotags to profile the captured EV surface protein markers at a single-particle level. Such a sensitive assay allows multiplexed profiling of EVs across five cancer types, revealing heterogeneous EV surface expression patterns. Furthermore, the IBMsv assay enables cancer diagnosis in a pilot clinical cohort (n = 55) with accuracies >95%, improves discrimination between cancer and noncancer patients via an algorithm, and monitors the surgical treatment response from hepatocellular carcinoma patients. This assay provides a fast, sensitive, streamlined, multiplexed, and portable blood test tool to enable cancer diagnosis and response monitoring in clinical settings.
Collapse
Affiliation(s)
- Xiaojie Qin
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Yuanhang Xiang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Linfeng Mao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Yu Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Binqi Wei
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fan Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
19
|
Zhang Q, Ren T, Cao K, Xu Z. Advances of machine learning-assisted small extracellular vesicles detection strategy. Biosens Bioelectron 2024; 251:116076. [PMID: 38340580 DOI: 10.1016/j.bios.2024.116076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Detection of extracellular vesicles (EVs), particularly small EVs (sEVs), is of great significance in exploring their physiological characteristics and clinical applications. The heterogeneity of sEVs plays a crucial role in distinguishing different types of cells and diseases. Machine learning, with its exceptional data processing capabilities, offers a solution to overcome the limitations of conventional detection methods for accurately classifying sEV subtypes and sources. Principal component analysis, linear discriminant analysis, partial least squares discriminant analysis, XGBoost, support vector machine, k-nearest neighbor, and deep learning, along with some combined methods such as principal component-linear discriminant analysis, have been successfully applied in the detection and identification of sEVs. This review focuses on machine learning-assisted detection strategies for cell identification and disease prediction via sEVs, and summarizes the integration of these strategies with surface-enhanced Raman scattering, electrochemistry, inductively coupled plasma mass spectrometry and fluorescence. The performance of different machine learning-based detection strategies is compared, and the advantages and limitations of various machine learning models are also evaluated. Finally, we discuss the merits and limitations of the current approaches and briefly outline the perspective of potential research directions in the field of sEV analysis based on machine learning.
Collapse
Affiliation(s)
- Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Tingju Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Ke Cao
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
20
|
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
|
21
|
Fan Z, Zhou J, Shu Q, Dong Y, Li Y, Zhang T, Bai G, Yu H, Lu F, Li J, Zhao X. Aptamer-bivalent-cholesterol-mediated proximity entropy-driven exosomal protein reporter for tumor diagnosis. Biosens Bioelectron 2024; 251:116104. [PMID: 38368644 DOI: 10.1016/j.bios.2024.116104] [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: 11/19/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
Exosomal proteins from the parental cells are considered to be promising biomarker sets for precise tumor diagnostics and monitoring. However, the accurate quantitative analysis of low-abundance exosomal proteins remains challenging due to the heterogeneity of clinical samples. Here, we standardized the exosomal concentration with a fluorogenic membrane probe and developed an aptamer-bivalent-cholesterol-mediated Proximity Entropy-driven Exosomal Protein Reporter (PEEPR). The proposed PEEPR enables the in-situ analysis of multiple exosomal proteins by integrating bivalent cholesterol anchor (exosomal lipid bilayer) and aptamer (exosomal proteins) with a proximity entropy-driven circuit. Based on this strategy, we successfully achieved detection limits of 3.9 pg/mL exosomal GPC-3 and 3.4 pg/mL exosomal PD-L1. Notably, the standardization of exosome concentrations is designed to avoid errors due to biological heterogeneity. The results showed that evaluating the levels of exosomal GPC-3 and PD-L1 in clinical samples via this strategy could accurately differentiate healthy individuals, hepatitis B patients, and hepatocellular carcinoma patients. In summary, PEEPR is a promising clinical diagnostic strategy for the quantitative analysis of a variety of tumor-associated exosomal proteins for the precise diagnosis and personalized treatment monitoring of tumors.
Collapse
Affiliation(s)
- Zhichao Fan
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jie Zhou
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Laboratory Medicine, Xingcheng Special Service Sanatorium of Strategic Support Force, Huludao, 125100, China
| | - Qiuxia Shu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yan Dong
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yingxue Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tingrui Zhang
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Gang Bai
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hua Yu
- Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fanghao Lu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jianjun Li
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xiang Zhao
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
22
|
Liu Q, Zheng J, Xie A, Chen M, Gong RY, Sheng Y, Chen HL, Qi CB. Exosome, a Rising Biomarkers in Liquid Biopsy: Advances of Label-Free and Label Strategy for Diagnosis of Cancer. Crit Rev Anal Chem 2024:1-12. [PMID: 38669199 DOI: 10.1080/10408347.2024.2339961] [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: 04/28/2024]
Abstract
Cancer is commonly considered as one of the most severe diseases, posing a significant threat to human health and society due to various serious challenges. These challenges include difficulties in accurate diagnosis and a high propensity to form metastasis. Tissue biopsy remains the gold standard for diagnosing and subtyping cancer. However, concerns arise from its invasive nature and the potential risk of metastasis during these complex diagnostic procedures. Meanwhile, liquid biopsy has recently witnessed the rapid advancements with the emergence of three prominent detection biomarkers: circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and exosomes. Whereas, the very low abundance of CTCs combined with the instability of ctDNA intensify the challenges and decrease the accuracy of these two biomarkers for cancer diagnosis. While exosomes have gained widespread recognition as a promising biomarker in liquid biopsy due to their relatively low-invasive detection method, excellent biostability, rich resources, high abundance, and ability to provide valuable information about cancer. Therefore, it is crucial to systematically summarize recent advancements mainly in exosome-based detection methods for early cancer diagnosis. Specifically, this review will primarily focus on label-based and label-free strategies for detecting cancer using exosomes. We anticipate that this comprehensive analysis will enhance readers' understanding of the significance and value of exosomes in the fields of cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Jing Zheng
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - An Xie
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Min Chen
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Rui-Yue Gong
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yuan Sheng
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Hong-Lei Chen
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Chu-Bo Qi
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
23
|
Zhang M, Liu F, Shi F, Chen H, Hu Y, Sun H, Qi H, Xiong W, Deng C, Sun N. High-throughput detection allied with machine learning for precise monitoring of significant serum metabolic changes in Helicobacter pylori infection. Talanta 2024; 269:125483. [PMID: 38042145 DOI: 10.1016/j.talanta.2023.125483] [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: 10/03/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/04/2023]
Abstract
High-throughput detection of large-scale samples is the foundation for rapidly accessing massive metabolic data in precision medicine. Machine learning is a powerful tool for uncovering valuable information hidden within massive data. In this work, we achieved the extraction of a single fingerprinting of 1 μL serum within 5 s through a high-throughput detection platform based on functionalized nanoparticles. We quickly obtained over a thousand serum metabolic fingerprintings (SMFs) including those of individuals with Helicobacter pylori (HP) infection. Combining four classical machine learning models and enrichment analysis, we attempted to extract and confirm useful information behind these SMFs. Based on all fingerprint signals, all four models achieved area under the curve (AUC) values of 0.983-1. In particular, orthogonal partial least squares discriminant analysis (OPLS-DA) model obtained value of 1 in both the discovery and validation sets. Fortunately, we identified six significant metabolic features, all of which can greatly contribute to the monitoring of HP infection, with AUC values ranging from 0.906 to 0.985. The combination of these six significant metabolic features can enable the precise monitoring of HP infection in serum, with over 95 % of accuracy, specificity and sensitivity. The OPLS-DA model displayed optimal performance and the corresponding scatter plot visualized the clear distinction between HP and HC. Interestingly, they exhibit a consistent reduction trend compared to healthy controls, prompting us to explore the possible metabolic pathways and potential mechanism. This work demonstrates the potential alliance between high-throughput detection and machine learning, advancing their application in precision medicine.
Collapse
Affiliation(s)
- Man Zhang
- Department of Chemistry, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Fenghua Liu
- Department of Gastroenterology, Shibei Hospital of Jing'an District of Shanghai, 4500 Gong He Xin Road, Shanghai, 200435, China
| | - Fangying Shi
- Department of Chemistry, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Haolin Chen
- Department of Chemistry, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Yi Hu
- Department of Emergency Shibei Hospital of Jing'an District of Shanghai, 4500 Gong He Xin Road, Shanghai, 200435, China
| | - Hong Sun
- Medical Examination Section, Shibei Hospital of Jing'an District of Shanghai, 4500 Gong He Xin Road, Shanghai, 200435, China
| | - Hongxia Qi
- Department of Gastroenterology, Shibei Hospital of Jing'an District of Shanghai, 4500 Gong He Xin Road, Shanghai, 200435, China
| | - Wenjian Xiong
- Department of Gastroenterology, Shibei Hospital of Jing'an District of Shanghai, 4500 Gong He Xin Road, Shanghai, 200435, China.
| | - Chunhui Deng
- Department of Chemistry, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200433, China; School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China.
| | - Nianrong Sun
- Department of Chemistry, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
24
|
Yang C, Chen H, Deng C, Sun N. Serological Exosome Metabolic Biopsy of Hepatocellular Carcinoma via Designed Core-Shell Nanoparticles. Anal Chem 2024. [PMID: 38323920 DOI: 10.1021/acs.analchem.3c02068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Exosome metabolite-based liquid biopsy is a promising strategy for large-scale application in practical clinics toward precise medicine. Given the current challenges in successive isolation and analysis of exosomes and their metabolites in this field, we established a low-cost, high-throughput, and rapid platform for serological exosome metabolic biopsy of hepatocellular carcinoma (HCC) via designed core-shell nanoparticles. It starts with the efficient extraction of high-quality serum exosomes and exosome metabolic features, based on which significantly obvious sample clusters are observed by unsupervised cluster analysis. The following integration of feature selection and supervised machine learning enables the identification of six key metabolites and achieves high-performance prediction between HCC, liver cirrhosis, and healthy controls. Specifically, both sensitivity and accuracy achieve 100% among any pairwise intergroup discrimination in a blind test. The quality and reliability of six key metabolites are further evaluated and validated by using different machine learning algorithms and pathway exploration. Our platform contributes to the future growth of new liquid biopsy technologies for precision diagnosis and real-time monitoring of HCC, among other conditions.
Collapse
Affiliation(s)
- Chenyu Yang
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Haolin Chen
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Chunhui Deng
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
25
|
You Q, Liang F, Wu G, Cao F, Liu J, He Z, Wang C, Zhu L, Chen X, Yang Y. The Landscape of Biomimetic Nanovesicles in Brain Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306583. [PMID: 37713652 DOI: 10.1002/adma.202306583] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Brain diseases, such as brain tumors, neurodegenerative diseases, cerebrovascular diseases, and brain injuries, are caused by various pathophysiological changes, which pose a serious health threat. Brain disorders are often difficult to treat due to the presence of the blood-brain barrier (BBB). Biomimetic nanovesicles (BNVs), including endogenous extracellular vesicles (EVs) derived from various cells and artificial nanovesicles, possess the ability to penetrate the BBB and thus can be utilized for drug delivery to the brain. BNVs, especially endogenous EVs, are widely distributed in body fluids and usually carry various disease-related signal molecules such as proteins, RNA, and DNA, and may also be analyzed to understand the etiology and pathogenesis of brain diseases. This review covers the exhaustive classification and characterization of BNVs and pathophysiological roles involved in various brain diseases, and emphatically focuses on nanotechnology-integrated BNVs for brain disease theranostics, including various diagnosis strategies and precise therapeutic regulations (e.g., immunity regulation, disordered protein clearance, anti-neuroinflammation, neuroregeneration, angiogenesis, and the gut-brain axis regulation). The remaining challenges and future perspectives regarding the nanotechnology-integrated BNVs for the diagnosis and treatment of brain diseases are also discussed and outlined.
Collapse
Affiliation(s)
- Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
26
|
Wang P, Liang Z, Li Z, Wang D, Ma Q. Plasmonic nanocavity-modulated electrochemiluminescence sensor for gastric cancer exosomal miRNA detection. Biosens Bioelectron 2024; 245:115847. [PMID: 37995625 DOI: 10.1016/j.bios.2023.115847] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/03/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Plasmonic nanocavity possessing highly light field confinement and electromagnetic field enhancement can concentrate and enhance the luminescence signal. The plasmonic nanocavity has the great potential value in biosensing research and improve analytical sensitivity. In this work, we constructed a plasmonic nanocavity between circular Au nanoplate-film and spherical Au nanoparticle with tetrahedral DNA nanostructures. The nanocavity structure can regulate the local density of optical states and provide the field restriction to enhance the spontaneous ECL radiation of PEDOT-S dots. Additionally, Au nanoparticle acted as nanoantenna which localized and modulated ECL to directional emission. Because the plasmonic nanocavity effectively collected and redistributed ECL signal, the emission was enhanced by 5.9 times with polarized characteristics. The proposed plasmonic nanocavity-based ECL sensor was further used to detect exosomal miRNA-223-3p in ascites. The detection results indicated the novel sensing strategy can assist early diagnosis of peritoneal metastasis of gastric cancer.
Collapse
Affiliation(s)
- Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Zihui Liang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Zhenrun Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Dongyu Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China.
| |
Collapse
|
27
|
Wu G, Lu F, Zhao J, Feng X, Ren Y, Hu S, Yu W, Dong B, Hu L. Investigation of rare earth-based magnetic nanocomposites for specific enrichment of exosomes from human plasma. J Chromatogr A 2024; 1714:464543. [PMID: 38065027 DOI: 10.1016/j.chroma.2023.464543] [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: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
Exosomes, also known as small extracellular vesicles, are widely present in a variety of body fluids (e.g., blood, urine, and saliva). Exosomes are becoming an alternative promising source of diagnostic markers for disease rich in cargo of metabolites, proteins, and nucleic acids. However, due to the low abundance and structure similarity with protein complex, the efficient isolation of exosomes is one of the most important issues for biomedical applications. With a higher order of f-orbitals in rare earth element, it will have strong adsorption toward the phosphate group on the surface of the phospholipid bilayer of exosomes. In this study, we systematically investigated the ability of various rare earths interacting with phosphate-containing molecules and plasma exosomes. One of the best binding europium was selected and used to synthesize core-shell magnetic nanomaterials (Fe3O4@SiO2@Eu2O3) for the enrichment of exosomes from human plasma. The developed nanomaterials exhibited higher enrichment capacity, less time consumption and more convenient handling compared to commonly used ultracentrifugation method. The nanomaterials were applied to separate exosomes from the plasma of patients with hepatocellular carcinoma and healthy controls for metabolomics study with high-resolution mass spectrometry, where 70 differentially expressed metabolites were identified, involving amino acid and lipid metabolic pathway. We anticipated the rare earth-based materials to be an alternative approach on exosome isolation for disease diagnosis or postoperative clinical monitoring.
Collapse
Affiliation(s)
- Guangyao Wu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Feng Lu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jiali Zhao
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Feng
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yujuan Ren
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Songtao Hu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Wenjing Yu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
28
|
Zhang Y, Fang X, Huang W, Li Q, Hu F, Liu H. Record Resolution of Nanometal Surface Energy Transfer Optical Nanoruler Projects 3D Spatial Configuration of Aptamers on a Living Cell Membrane. NANO LETTERS 2023; 23:11968-11974. [PMID: 38059895 DOI: 10.1021/acs.nanolett.3c04322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Decrypting the in situ three-dimensional spatial configuration of an aptamer is of considerable significance; however, suitable nanoscale resolution tools are lacking. Herein, we show that a new nanometal surface energy transfer (NSET) optical nanoruler has a record resolution, down to single-nucleobase levels. We labeled fluorophores on different T bases of XQ-2d, including 5', 3', 6T, 22T, 38T, and 52T positions. The NSET nanoruler in situ decrypted the base sequence-dependent distance projection on the nanogold surface, demonstrating that 5', 3', stem, and loop structures are symmetrical in three-dimensional spatial configuration. The orientation of the 5' and 3' stem was toward the antiCD71-binding site, whereas the loop was in the opposite direction at a considerable distance. Molecular docking simulation was performed to list all of the possible conformations; however, all base distance parameters projecting on the nanogold surface determined a single conformation of XQ-2d. The specific binding sites of XQ-2d were Lys477, Ser691, and Arg698 on the CD71 receptor.
Collapse
Affiliation(s)
- Yu Zhang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Xingru Fang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Wenwen Huang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Qi Li
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Fan Hu
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Honglin Liu
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| |
Collapse
|
29
|
Ouyang D, Wang C, Zhong C, Lin J, Xu G, Wang G, Lin Z. Organic metal chalcogenide-assisted metabolic molecular diagnosis of central precocious puberty. Chem Sci 2023; 15:278-284. [PMID: 38131069 PMCID: PMC10732007 DOI: 10.1039/d3sc05633c] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/25/2023] [Indexed: 12/23/2023] Open
Abstract
Metabolic analysis in biofluids based on laser desorption/ionization mass spectrometry (LDI-MS), featuring rapidity, simplicity, small sample volume and high throughput, is expected to be a powerful diagnostic tool. Nevertheless, the signals of most metabolic biomarkers obtained by matrix-assisted LDI-MS are too limited to achieve a highly accurate diagnosis due to serious background interference. To address this issue, nanomaterials have been frequently adopted in LDI-MS as substrates. However, the "trial and error" approach still dominates the development of new substrates. Therefore, rational design of novel LDI-MS substrates showing high desorption/ionization efficiency and no background interference is extremely desired. Herein, four few-layered organic metal chalcogenides (OMCs) were precisely designed and for the first time investigated as substrates in LDI-MS, which allowed a favorable internal energy and charge transfer by changing the functional groups of organic ligands and metal nodes. As a result, the optimized OMC-assisted platform satisfyingly enhanced the mass signal by ≈10 000 fold in detecting typical metabolites and successfully detected different saccharides. In addition, a high accuracy diagnosis of central precocious puberty (CPP) with potential biomarkers of 12 metabolites was realized. This work is not only expected to provide a universal detection tool for large-scale clinical diagnosis, but also provides an idea for the design and selection of LDI-MS substrates.
Collapse
Affiliation(s)
- Dan Ouyang
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University Fuzhou Fujian 350108 China
| | - Chuanzhe Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences (CAS) Fuzhou Fujian 350002 China
| | - Chao Zhong
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University Fuzhou Fujian 350108 China
| | - Juan Lin
- Department of Cardiology, Fujian Provincial Governmental Hospital Fuzhou 350003 China
| | - Gang Xu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences (CAS) Fuzhou Fujian 350002 China
| | - Guane Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences (CAS) Fuzhou Fujian 350002 China
| | - Zian Lin
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University Fuzhou Fujian 350108 China
| |
Collapse
|
30
|
Wei YN, Yan CY, Zhao ML, Zhao XH. The role and application of vesicles in triple-negative breast cancer: Opportunities and challenges. Mol Ther Oncolytics 2023; 31:100752. [PMID: 38130701 PMCID: PMC10733704 DOI: 10.1016/j.omto.2023.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Extracellular vesicles (EVs) carry DNA, RNA, protein, and other substances involved in intercellular crosstalk and can be used for the targeted delivery of drugs. Triple-negative breast cancer (TNBC) is rich in recurrent and metastatic disease and lacks therapeutic targets. Studies have proved the role of EVs in the different stages of the genesis and development of TNBC. Cancer cells actively secrete various biomolecules, which play a significant part establishing the tumor microenvironment via EVs. In this article, we describe the roles of EVs in the tumor immune microenvironment, metabolic microenvironment, and vascular remodeling, and summarize the application of EVs for objective delivery of chemotherapeutic drugs, immune antigens, and cancer vaccine adjuvants. EVs-based therapy may represent the next-generation tool for targeted drug delivery for the cure of a variety of diseases lacking effective drug treatment.
Collapse
Affiliation(s)
- Ya-Nan Wei
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Chun-Yan Yan
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Meng-Lu Zhao
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Xi-He Zhao
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| |
Collapse
|
31
|
Chen H, Qi Y, Yang C, Tai Q, Zhang M, Shen XZ, Deng C, Guo J, Jiang S, Sun N. Heterogeneous MXene Hybrid-Oriented Exosome Isolation and Metabolic Profiling for Early Screening, Subtyping and Follow-up Evaluation of Bladder Cancer. ACS NANO 2023; 17:23924-23935. [PMID: 38039354 DOI: 10.1021/acsnano.3c08391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
Exosome metabolite-based noninvasive liquid biopsy is an emerging research hotspot that tends to substitute current means in clinics. Nanostructure-based mass spectrometry enables continuous exosome isolation and metabolic profiling with superior analysis speed and high efficiency. Herein, we construct a heterogeneous MXene hybrid that possesses ternary binding sites for exosome capture and outstanding matrix performance for metabolite analysis. Upon optimizing experimental conditions, the average extraction of exosomes and their metabolic patterns from a 60 mL urine sample is completed within 45 s (40 samples per batch for 30 min). According to the exosomal metabolic patterns and the subsequently established biomarker panel, we distinguish early bladder cancer (BCa) from healthy controls with an area under the curve (AUC) value greater than 0.995 in model training and validation sets. As well, we realize subtype classification of BCa in the blind test on metabolic patterns, with an AUC value of 0.867. We also explore the significant biomarkers that are sensitive to follow-up patients, which indeed present reverse change levels compared with pathological progression. This study has the potential to guide the development of the liquid biopsy approach.
Collapse
Affiliation(s)
- Haolin Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200032, P. R. China
| | - Chenyu Yang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Qunfei Tai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Man Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200032, P. R. China
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200032, P. R. China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
32
|
Xu Z, Huang Y, Hu C, Du L, Du YA, Zhang Y, Qin J, Liu W, Wang R, Yang S, Wu J, Cao J, Zhang J, Chen GP, Lv H, Zhao P, He W, Wang X, Xu M, Wang P, Hong C, Yang LT, Xu J, Chen J, Wei Q, Zhang R, Yuan L, Qian K, Cheng X. Efficient plasma metabolic fingerprinting as a novel tool for diagnosis and prognosis of gastric cancer: a large-scale, multicentre study. Gut 2023; 72:2051-2067. [PMID: 37460165 PMCID: PMC11883865 DOI: 10.1136/gutjnl-2023-330045] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/26/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVE Metabolic biomarkers are expected to decode the phenotype of gastric cancer (GC) and lead to high-performance blood tests towards GC diagnosis and prognosis. We attempted to develop diagnostic and prognostic models for GC based on plasma metabolic information. DESIGN We conducted a large-scale, multicentre study comprising 1944 participants from 7 centres in retrospective cohort and 264 participants in prospective cohort. Discovery and verification phases of diagnostic and prognostic models were conducted in retrospective cohort through machine learning and Cox regression of plasma metabolic fingerprints (PMFs) obtained by nanoparticle-enhanced laser desorption/ionisation-mass spectrometry (NPELDI-MS). Furthermore, the developed diagnostic model was validated in prospective cohort by both NPELDI-MS and ultra-performance liquid chromatography-MS (UPLC-MS). RESULTS We demonstrated the high throughput, desirable reproducibility and limited centre-specific effects of PMFs obtained through NPELDI-MS. In retrospective cohort, we achieved diagnostic performance with areas under curves (AUCs) of 0.862-0.988 in the discovery (n=1157 from 5 centres) and independent external verification dataset (n=787 from another 2 centres), through 5 different machine learning of PMFs, including neural network, ridge regression, lasso regression, support vector machine and random forest. Further, a metabolic panel consisting of 21 metabolites was constructed and identified for GC diagnosis with AUCs of 0.921-0.971 and 0.907-0.940 in the discovery and verification dataset, respectively. In the prospective study (n=264 from lead centre), both NPELDI-MS and UPLC-MS were applied to detect and validate the metabolic panel, and the diagnostic AUCs were 0.855-0.918 and 0.856-0.916, respectively. Moreover, we constructed a prognosis scoring system for GC in retrospective cohort, which can effectively predict the survival of GC patients. CONCLUSION We developed and validated diagnostic and prognostic models for GC, which also contribute to advanced metabolic analysis towards diseases, including but not limited to GC.
Collapse
Affiliation(s)
- Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
| | - Yida Huang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
| | - Lingbin Du
- Office of Cancer Center, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yi-An Du
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jiangjiang Qin
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wanshan Liu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruimin Wang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Wu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Cao
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juxiang Zhang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gui-Ping Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hang Lv
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Zhao
- Department of Gastrointestinal Surgery, Sichuan Cancer Hospital, Chengdu, China
| | - Weiyang He
- Department of Gastrointestinal Surgery, Sichuan Cancer Hospital, Chengdu, China
| | - Xiaoliang Wang
- Department of General Surgery, Fenghua People's Hospital, Ningbo, China
| | - Min Xu
- Department of Gastroenterology, Tiantai People's Hospital, Taizhou, China
| | - Pingfang Wang
- Department of Gastroenterology, Xinchang People's Hospital, Shaoxing, China
| | - Chuanshen Hong
- Department of General Surgery, Daishan People's Hospital, Zhoushan, China
| | - Li-Tao Yang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jingli Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jiahui Chen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Qing Wei
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ruolan Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Li Yuan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Office of Cancer Center, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Yan S, Huang Z, Chen X, Chen H, Yang X, Gao M, Zhang X. Metabolic profiling of urinary exosomes for systemic lupus erythematosus discrimination based on HPL-SEC/MALDI-TOF MS. Anal Bioanal Chem 2023; 415:6411-6420. [PMID: 37644324 DOI: 10.1007/s00216-023-04916-z] [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: 06/27/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease which leads to the formation of immune complex deposits in multiple organs and has heterogeneous clinical manifestations. Currently, exosomes for liquid biopsy have been applied in diagnosis and monitoring of diseases, whereas SLE discrimination based on exosomes at the metabolic level is rarely reported. Herein, we constructed a protocol for metabolomic study of urinary exosomes from SLE patients and healthy controls (HCs) with high efficiency and throughput. Exosomes were first obtained by high-performance liquid size-exclusion chromatography (HPL-SEC), and then metabolic fingerprints of urinary exosomes were extracted by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) with high throughput and high efficency. With the statistical analysis by orthogonal partial least-squares discriminant analysis (OPLS-DA) model, SLE patients were efficiently distinguished from HCs, the area under the curve (AUC) of the receiver characteristic curve (ROC) was 1.00, and the accuracy of the unsupervised clustering heatmap was 90.32%. In addition, potential biomarkers and related metabolic pathways were analyzed. This method, with the characteristics of high throughput, high efficiency, and high accuracy, will provide the broad prospect of exosome-driven precision medicine and large-scale screening in clinical applications.
Collapse
Affiliation(s)
- Shaohan Yan
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | - Zhongzhou Huang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofei Chen
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | - Haolin Chen
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | - Xue Yang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China.
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| |
Collapse
|
34
|
Zhang Q, Chen M, Xu F, Wu W, Luo X, Wang Y, Li J, Cui X, Tan Y, Li Z, Lin Y, Zhang H, Wang W. One-pot preparation of bi-functional POSS-based hybrid monolith via photo-initiated polymerization for isolation of extracellular vesicles. Anal Chim Acta 2023; 1279:341785. [PMID: 37827681 DOI: 10.1016/j.aca.2023.341785] [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: 04/20/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023]
Abstract
Extracellular vesicles (EVs) are important participants in numerous pathophysiological processes, and could be used as valuable biomarkers to detect and monitor various diseases. However, facile EV isolation methods are the essential and preliminary issue for their downstream analysis and function investigation. In this work, a polyhedral oligomeric silsesquioxanes (POSS) based hybrid monolith combined metal affinity chromatography (MAC) and distearoyl phospholipid ethanolamine (DSPE) function was developed via photo-initiated thiol-ene polymerization. This synthesis process was facile, simple and convenient, and the obtained hybrid monolith could be applied to efficiently isolate EVs from bio-samples by taking advantages of the specific bond of Ti4+ and phosphate groups on the phospholipid membrane of EVs and the synergistic effect of DSPE insertion. Meanwhile, the eluted EVs could maintain their structural integrity and biological activity, suggesting they could be used for downstream application. Furthermore, 75 up-regulated proteins and 56 down-regulated proteins were identified by comparing the urinary EVs of colorectal cancer (CRC) patients and healthy donors, and these proteins might be used as potential biomarkers for early screening of CRC. These results demonstrated that this hybrid monolith could be used as a simple and convenient tool for isolating EVs from bio-samples and for wider applications in biomarker discovery.
Collapse
Affiliation(s)
- Qi Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Fang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Wen Wu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xintong Luo
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ying Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China; Taichang Liuhe People's Hospital, Suzhou, 215431, China
| | - Jiaxi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xuanhao Cui
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yujia Tan
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yujie Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
35
|
Li H, Yao S, Wang C, Bai C, Zhou P. Diverse applications and development of aptamer detection technology. ANAL SCI 2023; 39:1627-1641. [PMID: 37700097 DOI: 10.1007/s44211-023-00409-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/04/2023] [Indexed: 09/14/2023]
Abstract
Aptamers have received extensive attention in recent years because of their advantages of high specificity, high sensitivity and low immunogenicity. Aptamers can perform almost all functions of antibodies through the combination of spatial structure and target, which are called "chemical antibodies". At present, aptamers have been widely used in cell imaging, new drug development, disease treatment, microbial detection and other fields. Due to the diversity of modifications, aptamers can be combined with different detection technologies to construct aptasensors. This review focuses on the diversity of aptamers in the field of detection and the development of aptamer-based detection technology and proposes new challenges for aptamers in this field.
Collapse
Affiliation(s)
- Haozheng Li
- College of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Shibo Yao
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Cui Wang
- College of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Chenjun Bai
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Pingkun Zhou
- College of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| |
Collapse
|
36
|
Zhou Y, Li X, Zhao Y, Yang S, Huang L. Plasmonic alloys for quantitative determination and reaction monitoring of biothiols. J Mater Chem B 2023; 11:8639-8648. [PMID: 37491995 DOI: 10.1039/d3tb01076g] [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: 07/27/2023]
Abstract
Biothiols participate in numerous physiological and pathological processes in an organism. Quantitative determination and reaction monitoring of biothiols have important implications for evaluating human health. Herein, we synthesized plasmonic alloys as the matrix to assist the laser desorption and ionization (LDI) process of biothiols in mass spectrometry (MS). Plasmonic alloys were constructed with mesoporous structures for LDI enhancement and trimetallic (PdPtAu) compositions for noble metal-thiol hybridization, toward enhanced detection sensitivity and selectivity, respectively. Plasmonic alloys enabled direct detection of biothiols from complex biosamples without any enrichment or separation. We introduced internal standards into the quantitative MS system, achieving accurate quantitation of methionine directly from serum samples with a recovery rate of 103.19% ± 6.52%. Moreover, we established a rapid monitoring platform for the oxidation-reduction reaction of glutathione, consuming trace samples down to 200 nL with an interval of seconds. This work contributes to the development of molecular tools based on plasmonic materials for biothiol detection toward real-case applications.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Xvelian Li
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yuewei Zhao
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Shouzhi Yang
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Lin Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| |
Collapse
|
37
|
Li W, Wang W, Luo S, Chen S, Ji T, Li N, Pan W, Zhang X, Wang X, Li K, Zhang Y, Yan X. A sensitive and rapid electrochemical biosensor for sEV-miRNA detection based on domino-type localized catalytic hairpin assembly. J Nanobiotechnology 2023; 21:328. [PMID: 37689652 PMCID: PMC10492399 DOI: 10.1186/s12951-023-02092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/31/2023] [Indexed: 09/11/2023] Open
Abstract
Small extracellular-vesicule-associated microRNA (sEV-miRNA) is an important biomarker for cancer diagnosis. However, rapid and sensitive detection of low-abundance sEV-miRNA in clinical samples is challenging. Herein, a simple electrochemical biosensor that uses a DNA nanowire to localize catalytic hairpin assembly (CHA), also called domino-type localized catalytic hairpin assembly (DT-LCHA), has been proposed for sEV-miRNA1246 detection. The DT-LCHA offers triple amplification, (i). CHA system was localized in DNA nanowire, which shorten the distance between hairpin substrate, inducing the high collision efficiency of H1 and H2 and domino effect. Then, larger numbers of CHAs were triggered, capture probe bind DT-LCHA by exposed c sites. (ii) The DNA nanowire can load large number of electroactive substance RuHex as amplified electrochemical signal tags. (iii) multiple DT-LCHA was carried by the DNA nanowire, only one CHA was triggered, the DNA nanowire was trapped by the capture probe, which greatly improve the detection sensitivity, especially when the target concentration is extremely low. Owing to the triple signal amplification in this strategy, sEV-miRNA at a concentration of as low as 24.55 aM can be detected in 20 min with good specificity. The accuracy of the measurements was also confirmed using reverse transcription quantitative polymerase chain reaction. Furthermore, the platform showed good performance in discriminating healthy donors from patients with early gastric cancer (area under the curve [AUC]: 0.96) and was equally able to discriminate between benign gastric tumors and early cancers (AUC: 0.77). Thus, the platform has substantial potential in biosensing and clinical diagnosis.
Collapse
Affiliation(s)
- Wenbin Li
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wen Wang
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, 518001, People's Republic of China
| | - Shihua Luo
- Center for Clinical Laboratory Diagnosis and Research, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China
| | - Siting Chen
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Tingting Ji
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ningcen Li
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Weilun Pan
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiaohe Zhang
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiaojing Wang
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ke Li
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ye Zhang
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Xiaohui Yan
- Laboratory Medicine Center, Department of Laboratory Medicine, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
38
|
Liu R, Wu S, Liu W, Wang L, Dong M, Niu W. microRNAs delivered by small extracellular vesicles in MSCs as an emerging tool for bone regeneration. Front Bioeng Biotechnol 2023; 11:1249860. [PMID: 37720323 PMCID: PMC10501734 DOI: 10.3389/fbioe.2023.1249860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Bone regeneration is a dynamic process that involves angiogenesis and the balance of osteogenesis and osteoclastogenesis. In bone tissue engineering, the transplantation of mesenchymal stem cells (MSCs) is a promising approach to restore bone homeostasis. MSCs, particularly their small extracellular vesicles (sEVs), exert therapeutic effects due to their paracrine capability. Increasing evidence indicates that microRNAs (miRNAs) delivered by sEVs from MSCs (MSCs-sEVs) can alter gene expression in recipient cells and enhance bone regeneration. As an ideal delivery vehicle of miRNAs, MSCs-sEVs combine the high bioavailability and stability of sEVs with osteogenic ability of miRNAs, which can effectively overcome the challenge of low delivery efficiency in miRNA therapy. In this review, we focus on the recent advancements in the use of miRNAs delivered by MSCs-sEVs for bone regeneration and disorders. Additionally, we summarize the changes in miRNA expression in osteogenic-related MSCs-sEVs under different microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian, China
| |
Collapse
|
39
|
Pei C, Su R, Lu S, Chen X, Ding Y, Li R, Shu W, Zeng Y, Lin Y, Xu L, Mi Y, Wan J. Hollow multishelled heterostructures with enhanced performance for laser desorption/ionization mass spectrometry based metabolic diagnosis. J Mater Chem B 2023; 11:8206-8215. [PMID: 37554072 DOI: 10.1039/d3tb00766a] [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: 08/10/2023]
Abstract
High-performance metabolic diagnosis-based laser desorption/ionization mass spectrometry (LDI-MS) improves the precision diagnosis of diseases and subsequent treatment. Inorganic matrices are promising for the detection of metabolites by LDI-MS, while the structure and component impacts of the matrices on the LDI process are still under investigation. Here, we designed a multiple-shelled ZnMn2O4/(Co, Mn)(Co, Mn)2O4 (ZMO/CMO) as the matrix from calcined MOF-on-MOF for detecting metabolites in LDI-MS and clarified the synergistic impacts of multiple-shells and the heterostructure on LDI efficiency. The ZMO/CMO heterostructure allowed 3-5 fold signal enhancement compared with ZMO and CMO with the same morphology. Furthermore, the ZMO/CMO heterostructure with a triple-shelled hollow structure displayed a 3-fold signal enhancement compared to its nanoparticle counterpart. Taken together, the triple-shelled hollow ZMO/CMO exhibits 102-fold signal enhancement compared to the commercial matrix products (e.g., DHB and DHAP), allowing for sensitive metabolic profiling in bio-detection. We directly extracted metabolic patterns by the optimized triple-shelled hollow ZMO/CMO particle-assisted LDI-MS within 1 s using 100 nL of serum and used machine learning as the readout to distinguish hepatocellular carcinoma from healthy controls with the area under the curve value of 0.984. Our approach guides us in matrix design for LDI-MS metabolic analysis and drives the development of a nanomaterial-based LDI-MS platform toward precision diagnosis.
Collapse
Affiliation(s)
- Congcong Pei
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Rui Su
- Tianjin Second People's Hospital, Tianjin Medical University, Tianjin 300192, China.
- Tianjin Institute of Hepatology, Tianjin 300192, China
| | - Songting Lu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Xiaonan Chen
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Yajie Ding
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Rongxin Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Weikang Shu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Yu Zeng
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Yingying Lin
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| | - Liang Xu
- Tianjin Second People's Hospital, Tianjin Medical University, Tianjin 300192, China.
| | - Yuqiang Mi
- Tianjin Second People's Hospital, Tianjin Medical University, Tianjin 300192, China.
- Tianjin Institute of Hepatology, Tianjin 300192, China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China.
| |
Collapse
|
40
|
Wen F, Guan X, Qu HX, Jiang XJ. Integrated analysis of single-cell and bulk RNA-seq establishes a novel signature for prediction in gastric cancer. World J Gastrointest Oncol 2023; 15:1215-1226. [PMID: 37546563 PMCID: PMC10401466 DOI: 10.4251/wjgo.v15.i7.1215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Single-cell sequencing technology provides the capability to analyze changes in specific cell types during the progression of disease. However, previous single-cell sequencing studies on gastric cancer (GC) have largely focused on immune cells and stromal cells, and further elucidation is required regarding the alterations that occur in gastric epithelial cells during the development of GC.
AIM To create a GC prediction model based on single-cell and bulk RNA sequencing (bulk RNA-seq) data.
METHODS In this study, we conducted a comprehensive analysis by integrating three single-cell RNA sequencing (scRNA-seq) datasets and ten bulk RNA-seq datasets. Our analysis mainly focused on determining cell proportions and identifying differentially expressed genes (DEGs). Specifically, we performed differential expression analysis among epithelial cells in GC tissues and normal gastric tissues (NAGs) and utilized both single-cell and bulk RNA-seq data to establish a prediction model for GC. We further validated the accuracy of the GC prediction model in bulk RNA-seq data. We also used Kaplan–Meier plots to verify the correlation between genes in the prediction model and the prognosis of GC.
RESULTS By analyzing scRNA-seq data from a total of 70707 cells from GC tissue, NAG, and chronic gastric tissue, 10 cell types were identified, and DEGs in GC and normal epithelial cells were screened. After determining the DEGs in GC and normal gastric samples identified by bulk RNA-seq data, a GC predictive classifier was constructed using the Least absolute shrinkage and selection operator (LASSO) and random forest methods. The LASSO classifier showed good performance in both validation and model verification using The Cancer Genome Atlas and Genotype-Tissue Expression (GTEx) datasets [area under the curve (AUC)_min = 0.988, AUC_1se = 0.994], and the random forest model also achieved good results with the validation set (AUC = 0.92). Genes TIMP1, PLOD3, CKS2, TYMP, TNFRSF10B, CPNE1, GDF15, BCAP31, and CLDN7 were identified to have high importance values in multiple GC predictive models, and KM-PLOTTER analysis showed their relevance to GC prognosis, suggesting their potential for use in GC diagnosis and treatment.
CONCLUSION A predictive classifier was established based on the analysis of RNA-seq data, and the genes in it are expected to serve as auxiliary markers in the clinical diagnosis of GC.
Collapse
Affiliation(s)
- Fei Wen
- Qingdao University, Medical College, Qingdao 266000, Shandong Province, China
| | - Xin Guan
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266071, Shandong Province, China
| | - Hai-Xia Qu
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266071, Shandong Province, China
| | - Xiang-Jun Jiang
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266071, Shandong Province, China
| |
Collapse
|
41
|
Guo Y, Nie Y, Wang P, Li Z, Ma Q. MoS 2 QDs-MXene heterostructure-based ECL sensor for the detection of miRNA-135b in gastric cancer exosomes. Talanta 2023; 259:124559. [PMID: 37080077 DOI: 10.1016/j.talanta.2023.124559] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
Exosomes play an important role in the proliferation, adhesion and migration of cancer cells. In this study, we have developed a novel electrochemiluminescence (ECL) sensor based on MoS2 QDs-MXene heterostructure and Au NPs@biomimetic lipid layer to detect exosomal miRNA. MoS2 QDs-MXene heterostructure had been prepared as the luminescence probe. Ti3C2Tx MXene nanosheets possessed the large specific surface area, excellent flexibility and superior conductivity. MoS2 QDs on the MXene nanosheets worked as the radiation center to generate strong ECL signal. Meanwhile, Au NPs with biomimetic lipid layer have been modified on the electrode, which retained the lipid dynamics and excellent antifouling property. When miRNA-135b was recognized on the Au NPs@biomimetic lipid layer, MoS2 QDs-MXene heterostructure was linked on the electrode and further extended the outer Helmholtz plane. As a result, the self-luminous Faraday cage-mode sensing system has been used to detect miRNA-135b from 30 fM to 20 nM with a detection limit of 10 fM. Furthermore, gastric cancer exosomal miRNA in the ascites of clinical patients has been detected successfully. The sensing system can be served as a versatile platform with huge application potential in the field of exosome detection.
Collapse
Affiliation(s)
- Yuchen Guo
- Department of Gastrocolorectal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China.
| | - Yixin Nie
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Zhenrun Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China.
| |
Collapse
|
42
|
Yan S, Zheng H, Zhao J, Gao M, Zhang X. Quantification of GPC1(+) Exosomes Based on MALDI-TOF MS In Situ Signal Amplification for Pancreatic Cancer Discrimination and Evaluation. Anal Chem 2023. [PMID: 37368911 DOI: 10.1021/acs.analchem.3c00193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Pancreatic cancer (PC) has a high mortality, with a fairly low five-year survival rate, because of its delayed diagnosis. Recently, liquid biopsy, especially based on exosomes, has attracted vast attention, thanks to its low invasiveness. Herein, we constructed a protocol for pancreatic cancer related Glypican 1 (GPC1) exosome quantification, based on in situ mass spectrometry signal amplification, by utilizing mass tag molecules on gold nanoparticles (AuNPs). Exosomes were extracted and purified by size-exclusion chromatography (SEC), captured by TiO2 modified magnetic nanoparticles, and then targeted specifically by anti-GPC1 antibody modified on AuNPs. With matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS), the signal of PC biomarker, GPC1, was converted to a mass tag signal and amplified. With addition of a certain amount of internal standard molecules modified on AuNPs, the relative intensity ratio of mass tag to internal standard was proportional to the concentration of GPC1(+) exosomes derived from pancreatic cancer cell lines, PANC-1, with good linearity (R2 = 0.9945) in a wide dynamic range from 7.1 × 10 to 7.1 × 106 particles/μL. This method was further applied to plasma samples from healthy control (HC) and pancreatic cancer patients with different tumor load, and exhibited a great potential in discriminating diagnosed PC patients from HC, and has the monitoring potential in PC progression.
Collapse
Affiliation(s)
- Shaohan Yan
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Haoyang Zheng
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Jiandong Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
43
|
Lv QY, Cui HF, Song X. Aptamer-based technology for gastric cancer theranostics. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:2142-2153. [PMID: 37114324 DOI: 10.1039/d3ay00415e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Gastric cancer is one of the most common causes of cancer death worldwide. This cancer exhibits high molecular and phenotype heterogeneity. The overall survival rate for gastric cancer is very low because it is always diagnosed in the advanced stages. Therefore, early detection and treatment are of great significance. Currently, biomedical studies have tapped the potential clinical applicability of aptamer-based technology for gastric cancer diagnosis and targeted therapy. Herein, we summarize the enrichment and evolution of relevant aptamers, followed by documentation of the recent developments in aptamer-based techniques for early diagnosis and precision therapy for gastric cancers.
Collapse
Affiliation(s)
- Qi-Yan Lv
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| | - Hui-Fang Cui
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| | - Xiaojie Song
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| |
Collapse
|
44
|
Shi F, Qi Y, Jiang S, Sun N, Deng C. Hollow Core-Shell Metal Oxide Heterojunctions for the Urinary Metabolic Fingerprint-Based Noninvasive Diagnostic Strategy. Anal Chem 2023; 95:7312-7319. [PMID: 37121232 DOI: 10.1021/acs.analchem.3c00369] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Urine is a preferred object for noninvasive diagnostic strategies. Urinary metabolic analysis is speculatively regarded as an ideal tool for screening diseases closely related to the genitourinary system in view of the intimate relationship between metabolomics and phenotype. Herein, we propose a urinary metabolic fingerprint-based noninvasive diagnostic strategy by designing hollow core-shell metal oxide heterojunctions (denoted as MOHs). With outstanding light absorption and electron-hole separation ability, MOHs aid in the extraction of high-performance urine metabolic fingerprints. Coupled with optimized machine learning algorithms, we establish a metabolic marker panel for accurate diagnosis of prostate cancer (PCa), which is the most common malignant tumor of the male genitourinary system, achieving accuracies of 84.72 and 83.33% in the discovery and validation sets, respectively. Furthermore, metabolite variations and related pathway analyses confirm the credibility and change correlation of key metabolic features in PCa. This work tends to advance the noninvasive diagnostic strategy toward clinical realities.
Collapse
Affiliation(s)
- Fangying Shi
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Urology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunhui Deng
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| |
Collapse
|
45
|
Meng F, Yu W, Niu M, Tian X, Miao Y, Li X, Zhou Y, Ma L, Zhang X, Qian K, Yu Y, Wang J, Huang L. Ratiometric electrochemical OR gate assay for NSCLC-derived exosomes. J Nanobiotechnology 2023; 21:104. [PMID: 36964516 PMCID: PMC10037838 DOI: 10.1186/s12951-023-01833-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/27/2023] [Indexed: 03/26/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common pathological type of LC and ranks as the leading cause of cancer deaths. Circulating exosomes have emerged as a valuable biomarker for the diagnosis of NSCLC, while the performance of current electrochemical assays for exosome detection is constrained by unsatisfactory sensitivity and specificity. Here we integrated a ratiometric biosensor with an OR logic gate to form an assay for surface protein profiling of exosomes from clinical serum samples. By using the specific aptamers for recognition of clinically validated biomarkers (EpCAM and CEA), the assay enabled ultrasensitive detection of trace levels of NSCLC-derived exosomes in complex serum samples (15.1 particles μL-1 within a linear range of 102-108 particles μL-1). The assay outperformed the analysis of six serum biomarkers for the accurate diagnosis, staging, and prognosis of NSCLC, displaying a diagnostic sensitivity of 93.3% even at an early stage (Stage I). The assay provides an advanced tool for exosome quantification and facilitates exosome-based liquid biopsies for cancer management in clinics.
Collapse
Affiliation(s)
- Fanyu Meng
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Wenjun Yu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Minjia Niu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yayou Miao
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xvelian Li
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yan Zhou
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yongchun Yu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Lin Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
46
|
Su X, Liu X, Xie Y, Chen M, Zheng C, Zhong H, Li M. Integrated SERS-Vertical Flow Biosensor Enabling Multiplexed Quantitative Profiling of Serological Exosomal Proteins in Patients for Accurate Breast Cancer Subtyping. ACS NANO 2023; 17:4077-4088. [PMID: 36758150 DOI: 10.1021/acsnano.3c00449] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Protein profiles of exosomes (EXOs) in clinical samples of cancer patients have become a promising diagnostic and therapeutic biomarker. However, simultaneous quantitative analysis of multiple exosomal proteins of interest remains challenging. To address the unmet need, we develop a paper-based surface-enhanced Raman spectroscopy (SERS)-vertical flow biosensor, named iREX (integrated Raman spectroscopic EXO) biosensor, for multiplexed quantitative profiling of exosomal proteins in clinical serum samples of patients. Utilizing this iREX biosensor, we are able to quantitatively profile MUC1, HER2 and CEA in EXO samples derived from various breast cancer cell subtypes. The results show discriminative expression profiles of the three exosomal proteins in these cell subtypes, which allows for accurate diagnosis and molecular subtyping of breast cancer. We further validate the clinical utility of the iREX biosensor for simultaneous quantitative analysis of MUC1, HER2 and CEA in patient's blood serums, thereby aiding in noninvasive breast cancer subtyping and longitudinal treatment monitoring. Our iREX biosensor integrating the SERS detection in a vertical flow diagnostic device offers great advantages of high sensitivity, molecular specificity, powerful multiplexing capability, and high diagnostic accuracy. We believe that the iREX biosensor could be a promising clinical tool for comprehensive analysis of exosomal proteins in clinical samples for personalized diagnosis and precise management of breast cancer.
Collapse
Affiliation(s)
- Xiaoming Su
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China
| | - Xinyu Liu
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Yangcenzi Xie
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Mingyang Chen
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Chao Zheng
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Hong Zhong
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
47
|
Ma S, Zhou M, Xu Y, Gu X, Zou M, Abudushalamu G, Yao Y, Fan X, Wu G. Clinical application and detection techniques of liquid biopsy in gastric cancer. Mol Cancer 2023; 22:7. [PMID: 36627698 PMCID: PMC9832643 DOI: 10.1186/s12943-023-01715-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/02/2023] [Indexed: 01/12/2023] Open
Abstract
Gastric cancer (GC) is one of the most common tumors worldwide and the leading cause of tumor-related mortality. Endoscopy and serological tumor marker testing are currently the main methods of GC screening, and treatment relies on surgical resection or chemotherapy. However, traditional examination and treatment methods are more harmful to patients and less sensitive and accurate. A minimally invasive method to respond to GC early screening, prognosis monitoring, treatment efficacy, and drug resistance situations is urgently needed. As a result, liquid biopsy techniques have received much attention in the clinical application of GC. The non-invasive liquid biopsy technique requires fewer samples, is reproducible, and can guide individualized patient treatment by monitoring patients' molecular-level changes in real-time. In this review, we introduced the clinical applications of circulating tumor cells, circulating free DNA, circulating tumor DNA, non-coding RNAs, exosomes, and proteins, which are the primary markers in liquid biopsy technology in GC. We also discuss the current limitations and future trends of liquid biopsy technology as applied to early clinical biopsy technology.
Collapse
Affiliation(s)
- Shuo Ma
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China
| | - Meiling Zhou
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China
| | - Yanhua Xu
- grid.452743.30000 0004 1788 4869Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, 225000 Jiangsu China
| | - Xinliang Gu
- grid.440642.00000 0004 0644 5481Department of Laboratory Medicine, Medical School, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001 Jiangsu China
| | - Mingyuan Zou
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China
| | - Gulinaizhaer Abudushalamu
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China
| | - Yuming Yao
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China
| | - Xiaobo Fan
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China
| | - Guoqiu Wu
- grid.452290.80000 0004 1760 6316Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, 210009 Jiangsu China
| |
Collapse
|