1
|
Chen H, Pang B, Liu Z, Li B, Wang Q, Fan B, Han M, Gong J, Zhou C, Chen Y, Li Y, Jiang J. The Diagnostic Value of Plasma Small Extracellular Vesicle-Derived CAIX Protein in Prostate Cancer and Clinically Significant Prostate Cancer: A Study on Predictive Models. Prostate 2025; 85:723-741. [PMID: 40013658 PMCID: PMC12038087 DOI: 10.1002/pros.24879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 01/25/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Current diagnostic tools are inaccurate and not specific to prostate cancer (PCa) diagnosis. Cancer-derived small extracellular vehicles (sEVs) play a key role in intercellular communication. In this study, we examined the diagnostic value of plasma sEV-derived carbonic anhydrase IX (CAIX) protein for PCa and clinically significant prostate cancer (csPCa) diagnosis and avoiding unnecessary biopsies. METHODS Plasma samples (n = 230) were collected from the patients who underwent prostate biopsy with elevated prostate-specific antigen (PSA) levels. sEVs were isolated and characterized, and sEV protein CAIX was measured using an enzyme-linked immunosorbent assay. Independent predictors of csPCa (Gleason score ≥ 7) were identified, and a predictive model was established. A Nomogram for predicting csPCa was developed using data from the training cohort. RESULTS The expression of sEV protein CAIX was significantly higher in both PCa and csPCa compared to benign patients and nonsignificant PCa (nsPCa) (Gleason score < 7, p < 0.001). sEV protein CAIX performed well in distinguishing PCa from benign patients. The predictive model defined by sEV protein CAIX and PSA density (PSAD) demonstrated the highest discriminative ability for csPCa (AUC = 0.895), with diagnostic sensitivity and specificity of 82.5% and 85.8%, respectively. Furthermore, sEV protein CAIX is an effective predictor of 2-year biochemical recurrence (BCR) in PCa patients (p = 0.013), and its high expression is significantly associated with poorer BCR-free survival (p < 0.05). CONCLUSIONS Our findings demonstrate the excellent performance of sEV protein CAIX in PCa and csPCa diagnosis. The Nomogram-based csPCa predictive model incorporating sEV protein CAIX and PSAD exhibits strong predictive value. Additionally, assessing plasma sEV protein CAIX expression levels can further aid in evaluating patient prognosis and provide a basis for making effective treatment decisions.
Collapse
Affiliation(s)
- Haotian Chen
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Bairen Pang
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Zhihan Liu
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Benjie Li
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Qi Wang
- Cancer Care Centre, St George HospitalKogarahNew South WalesAustralia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW SydneyKensingtonNew South WalesAustralia
| | - Baokun Fan
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Meng Han
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Jie Gong
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Cheng Zhou
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Yingzhi Chen
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Yong Li
- Cancer Care Centre, St George HospitalKogarahNew South WalesAustralia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW SydneyKensingtonNew South WalesAustralia
| | - Junhui Jiang
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| |
Collapse
|
2
|
Ngo CHL, Tukova A, Zhang W, Tsao SCH, Wang Y. Sensitive detection of small extracellular vesicles using a gold nanostar-based SERS assay. Analyst 2025; 150:2108-2117. [PMID: 40202797 DOI: 10.1039/d5an00110b] [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: 04/11/2025]
Abstract
Small extracellular vesicles (sEVs) are lipid bilayer-bound vesicles that carry critical biomarkers for disease detection. However, the inherent heterogeneity and complexity of sEV molecular characteristics pose significant challenges for accurate and comprehensive molecular profiling. Traditional analytical methods, including immunoblotting, enzyme-linked immunosorbent assay (ELISA), and flow cytometry, exhibit several limitations, such as being time-consuming, requiring large sample volumes, and demonstrating relatively low sensitivity. Therefore, there is an urgent need to develop a highly sensitive and specific assay for the reliable detection of sEVs. Surface-enhanced Raman scattering (SERS) assays have emerged as a promising approach for sEV detection, offering advantages including high sensitivity and specificity. In the proposed SERS assay, SERS nanotags - comprising nanoparticles coated with Raman-active molecules and conjugated with antibodies - are employed to label surface-bound molecules on sEVs. This approach facilitates the generation of a high-intensity signal from molecules present in low abundance. Recently, anisotropic nanoparticles, such as star-shaped nanostructures, have garnered interest due to their ability to significantly amplify generated SERS signals for ultra-sensitive biomarker detection. In this study, we explore the application of gold nanostars (AuNSs) as SERS nanotags for the detection of sEVs. In principle, AuNS-based SERS nanotags were used to label the EpCAM protein, which can be found on the surface of cancer cell derived sEVs, and then sEV labelled SERS nanotags were captured by CD9-conjugated magnetic beads to form an immunocomplex, which exhibits a SERS signal. Our results demonstrate that the proposed SERS assay utilizing AuNSs provides high specificity and sensitivity, with a detection limit as low as 2.47 × 103 particles per μL. Furthermore, the assay was tested with spiked plasma samples (cancer cell-derived sEVs spiked into healthy plasma), showing that its specificity remains unaffected by the presence of plasma. These findings suggest that the SERS assay incorporating AuNSs holds significant promise as an effective and reliable detection method for potential clinical applications.
Collapse
Affiliation(s)
| | - Anastasiia Tukova
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Macquarie University, Sydney, Australia.
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
3
|
Ngo CHL, Hnit SST, Hassanzadeh-Barforoushi A, Chowdhury F, Zhang W, Inglis DW, Chang-Hao Tsao S, Wang Y. A New Strategy to Modify Glass for Capture and Detection of Small Extracellular Vesicles. J Phys Chem B 2025; 129:4373-4382. [PMID: 40277291 DOI: 10.1021/acs.jpcb.5c00290] [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: 04/26/2025]
Abstract
Small extracellular vesicles (sEVs) are nanoscale lipid bilayer vesicles secreted from all types of cells to the extracellular environment. They inherit membrane proteins from their parent cells, making them one of the key biomaterials or biomarkers for disease diagnosis. Microfluidics is emerging as a promising platform for sEV capture, with many methods relying on the modification to a glass substrate for efficient capture. In this study, we propose a new, one-step surface modification method, using silane─poly(ethylene glycol) (PEG) - 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), named as silane-PEG-DSPE, to capture sEVs on glass. In this design, silane group attaches to hydroxylated glass surface via covalent bond between Si-(OEt)3 and hydroxyl (OH) group. DSPE then binds to sEVs with distearyl chains firmly anchoring to the vesicle lipid membrane. We determined the optimal conditions for silane-PEG-DSPE modification and tested the efficiency of silane-PEG-DSPE in capturing sEVs by fluorescent imaging. Additionally, surface-enhanced Raman scattering (SERS) demonstrated that the EpCAM-positive sEVs were presented on the glass surface. This suggests that the platform is applicable for sEV detection using various methods, including fluorescent imaging and SERS. Furthermore, we have demonstrated that SERS can detect sEVs from breast cancer (BC) patient plasma with high specificity and sensitivity (as low as 1.6 × 107 particles/mL). Additionally, our analysis reveals a significantly higher expression of EpCAM in BC-derived sEVs compared with those obtained from healthy individuals. Thus, we postulate that the proposed method will find broad applications in the future, particularly as an effective tool in cancer diagnosis.
Collapse
Affiliation(s)
- Cao Hoang Long Ngo
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| | - Su Su Thae Hnit
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| | | | - Faiza Chowdhury
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| | - David W Inglis
- School of Engineering, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| |
Collapse
|
4
|
He Q, Qin L, Yao Y, Wang W. Clinical study of the diagnosis of thyroid tumours using Raman spectroscopy. Braz J Otorhinolaryngol 2025; 91:101568. [PMID: 40022834 PMCID: PMC11914986 DOI: 10.1016/j.bjorl.2025.101568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/27/2024] [Accepted: 12/28/2024] [Indexed: 03/04/2025] Open
Abstract
OBJECTIVE The feasibility of the RS for the clinical diagnosis of thyroid tumours was explored. METHODS The tumour specimens from 30 benign patients and 30 malignant patients were collected. The collected specimens were subjected to RS and histopathological analysis. The Raman peak intensities of all the specimens were calculated, and the data were analysed using discriminant analysis. RESULTS (1) The prevalence rate of malignant tumours in females was as high as 76.7%. Central lymph node metastasis of malignant thyroid tumours accounted for 33.3% of cases, and lateral cervical lymph node metastasis accounted for only 6.7%. (2) The spectral intensity of malignant thyroid tumours was significantly greater than benign thyroid tumours at 1309 cm-1, which should be the characteristic peak of thyroid cancer. The accuracy, sensitivity, and specificity of the RS for differentiating benign from malignant thyroid tumours were 95%, 83.3% and 89.2%. CONCLUSION RS is feasible for the diagnosis of thyroid tumours. This study provides experimental and clinical support for the wider application of RS in the evaluation of thyroid tissue. LEVELS OF EVIDENCE Levels 4.
Collapse
Affiliation(s)
- Qingjian He
- The First People's Hospital of Huzhou City, Department of Breast and Thyroid Surgery, Huzhou, China
| | - Lianjin Qin
- The First People's Hospital of Huzhou City, Department of Breast and Thyroid Surgery, Huzhou, China
| | - Yongqiang Yao
- Zhong Shan Hospital of Dalian University, Department of Breast and Thyroid Surgery, Dalian, Liaoning, China.
| | - WenJuan Wang
- First People's Hospital of Huzhou City, Department of Cardiovascular Diagnosis and Treatment Center, Huzhou, China.
| |
Collapse
|
5
|
Davidson CL, Kshirsagar P, Vengoji R, Shonka N. Exosomes in Glioma: Diagnostic and Therapeutic Potentials. ADVANCES IN ONCOLOGY 2025; 5:151-162. [PMID: 40443744 PMCID: PMC12119113 DOI: 10.1016/j.yao.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Affiliation(s)
- Caroline L. Davidson
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE 68198-8437, USA
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198-6840, USA
| |
Collapse
|
6
|
Zheng Y, Bai B, Wei Z, Zhang M, Zhang Q, Li X. Plasma Exosomal-Derived SERPINA1 and GNAI2 Downregulation as Potential Diagnostic Biomarkers of Kawasaki Disease with Coronary Artery Aneurysms. Int J Mol Sci 2025; 26:2668. [PMID: 40141310 PMCID: PMC11942354 DOI: 10.3390/ijms26062668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Kawasaki disease (KD) with coronary artery aneurysms (CAAs) is currently the primary cause of childhood acquired heart disease with an unclear pathogenesis. We established five groups for the discovery of differentially expressed proteins (DEPs): healthy control, febrile control, KD without CAAs, KD with small and medium CAAs, and KD with giant CAAs (n = 8 in each group). The validation of selected DEPs was conducted in another five groups (n = 4 in each group). We conducted comprehensive bioinformatics analyses to elucidate the functional roles of the DEPs in the groups of KD with CAAs and KD without CAAs. A total of 104 DEPs were identified in KD patients, which were primarily associated with complement-related pathways. A trend analysis of these 104 DEPs revealed 54 significantly changed DEPs associated with increased disease severity, which were primarily associated with G-protein-related functions. The alterations in α-1-antitrypsin short peptide (SERPINA1) and guanine nucleotide-binding protein G(i) subunit alpha-2 (GNAI2), which were selected from complement-related and G-protein-related pathways, respectively, were validated by Western blotting, and they were significantly decreased in KD patients with vs. without CAAs. In addition, we conducted an analysis of the DEPs in the groups of KD with CAAs and KD without CAAs, separately. There were 91 DEPs specifically expressed in KD patients with CAAs, associated with the neutrophil extracellular trap and complement pathways, while 16 DEPs were specific to those without CAAs, associated with viral infection and immunity pathways. Additionally, for DEPs among different severities of CAAs, there were 102 DEPs in KD patients with small and medium CAAs, associated with complement pathways and platelet activation pathways, whereas 34 DEPs were specific to giant CAAs, associated with the Rap1 signaling pathway and cell functions. In conclusion, this study provides plasmatic exosomal protein profiles in KD patients with CAAs, suggesting that SERPINA1 and GNIA2 might serve as novel potential diagnostic biomarkers for KD with CAAs.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Peking Union Medical College Graduate School, Beijing 100020, China;
| | - Baoling Bai
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China;
| | - Zhimiao Wei
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Beijing 100020, China; (Z.W.); (M.Z.)
| | - Mingming Zhang
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Beijing 100020, China; (Z.W.); (M.Z.)
| | - Qin Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China;
| | - Xiaohui Li
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Peking Union Medical College Graduate School, Beijing 100020, China;
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Beijing 100020, China; (Z.W.); (M.Z.)
| |
Collapse
|
7
|
Liu YJ, Kyne M, Kang C, Wang C. Raman spectroscopy in extracellular vesicles analysis: Techniques, applications and advancements. Biosens Bioelectron 2025; 270:116970. [PMID: 39603214 DOI: 10.1016/j.bios.2024.116970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Raman spectroscopy provides a robust approach for detailed analysis of the chemical and molecular profiles of extracellular vesicles (EVs). Recent advancements in Raman techniques have significantly enhanced the sensitivity and accuracy of EV characterization, enabling precise detection and profiling of molecular components within EV samples. This review introduces and compares various Raman-based techniques for EV characterization. These include Raman spectroscopy (RS), which provides fundamental molecular information; Raman trapping analysis (RTA), which combines optical trapping with Raman scattering for the manipulation and analysis of individual EVs; surface-enhanced Raman spectroscopy (SERS), which enhances the Raman signal through the use of metallic nanostructures, significantly improving sensitivity; and microfluidic SERS, which integrates SERS with microfluidic platforms to allow high-throughput, label-free analysis of EVs in biological fluids. In addition to comparing various Raman techniques, this review provides a comprehensive analysis that includes comparisons of machine learning methods, EV isolation techniques, and characterization strategies. By integrating these approaches, the review presents a holistic perspective on Raman-based EV analysis, covering profiling, purity, heterogeneity and size analysis as well as imaging. The combined assessment of Raman technologies with advanced computational and experimental methodologies supports the development of more robust diagnostic and therapeutic applications involving EVs.
Collapse
Affiliation(s)
- Ya-Juan Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology, and the NMPA & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Michelle Kyne
- School of Chemistry, National University of Ireland, Galway, Galway, H91 CF50, Ireland
| | - Chao Kang
- School of Chemistry and Chemical Engineering, Guizhou University, Guiyang, 550025, China.
| | - Cheng Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China; Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
8
|
Wang Q, Yu B, Yang B, Zhang X, Yu G, Wang Z, Qin H, Ma Y. Precision Fabrication and Optimization of Nanostructures for Exosome Detection via Surface-Enhanced Raman Spectroscopy. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:266. [PMID: 39997829 PMCID: PMC11858208 DOI: 10.3390/nano15040266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025]
Abstract
Exosome detection is crucial for biomedical research and clinical diagnostics due to their unique characteristics. Surface-enhanced Raman spectroscopy (SERS) based on nanostructure substrates with local field enhancement capability is a promising detection approach. However, the random distribution of nanostructures leads to uneven "hotspots" distribution, which limits their application in SERS detection. Here, we systematically investigated the impact of experimental parameters on nanostructure morphology and analyzed their formation mechanism, achieving controllable nanocone fabrication. Subsequent experiments confirmed the reliability and effectiveness of the fabricated nanocone in exosome SERS detection. This work not only realized flexible control of nanostructures but also expanded their application prospects in the field of exosome analysis.
Collapse
Affiliation(s)
- Qingyi Wang
- School of Mechanical-Electronic and Vehicle Engineering, Beijing University of Civil Engineering and Architecture, Beijing 102616, China; (Q.W.); (Z.W.)
| | - Bowen Yu
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China; (B.Y.); (X.Z.); (G.Y.)
| | - Bingbing Yang
- Department of Laboratory Medicine, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210006, China;
| | - Xuanhe Zhang
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China; (B.Y.); (X.Z.); (G.Y.)
| | - Guoxu Yu
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China; (B.Y.); (X.Z.); (G.Y.)
| | - Zeyu Wang
- School of Mechanical-Electronic and Vehicle Engineering, Beijing University of Civil Engineering and Architecture, Beijing 102616, China; (Q.W.); (Z.W.)
| | - Hua Qin
- School of Mechanical-Electronic and Vehicle Engineering, Beijing University of Civil Engineering and Architecture, Beijing 102616, China; (Q.W.); (Z.W.)
| | - Yuan Ma
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China; (B.Y.); (X.Z.); (G.Y.)
| |
Collapse
|
9
|
Fan R, Chen S, Lan F, Li W, Zhu Y, Zhang L, Zhang Y, Li L. Surface-Enhanced Raman Scattering (SERS)-based biosensors for advanced extracellular vesicle detection: A review. Anal Chim Acta 2025; 1336:343264. [PMID: 39788643 DOI: 10.1016/j.aca.2024.343264] [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: 07/09/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Extracellular Vesicles (EVs), as nano-scale vesicles rich in biological information, hold an indispensable status in the biomedical field. However, due to the intrinsic small size and low abundance of EVs, their effective detection presents significant challenges. Although various EV detection techniques exist, their sensitivity and ease of operation still need enhancement. RESULTS Surface-Enhanced Raman Scattering (SERS) is known for its high sensitivity and specificity. It stands out in tackling the challenges that traditional EV detection methods face. In this review, we focus on the application of SERS-based biosensors in EV detection. It provides a detailed introduction to the recognition and capture of EVs, strategies for mediating signal amplification, and detection of EV biomarkers. Finally, the challenges and prospects of SERS-based biosensors are discussed. SIGNIFICANCE SERS-based biosensor enhances the Raman signal, allowing for the detection of biomarkers at low concentrations. This capability reveals its substantial potential in identifying EVs and analyzing molecular data. It paves the path for advanced EV detection.
Collapse
Affiliation(s)
- Rui Fan
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China; Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Siting Chen
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China; School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Fei Lan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Wenbin Li
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Yitong Zhu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China; School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Lifeng Zhang
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China; Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Ye Zhang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China.
| | - Ling Li
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China; School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
10
|
Jia Y, Li Y, Bai X, Liu L, Shan Y, Wang F, Yu Z, Zheng C. Raman Spectroscopy and Exosome-Based Machine Learning Predicts the Efficacy of Neoadjuvant Therapy for HER2-Positive Breast Cancer. Anal Chem 2025; 97:1374-1385. [PMID: 39780544 DOI: 10.1021/acs.analchem.4c05833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Early prediction of the neoadjuvant therapy efficacy for HER2-positive breast cancer is crucial for personalizing treatment and enhancing patient outcomes. Exosomes, which play a role in tumor development and treatment response, are emerging as potential biomarkers for cancer diagnosis and efficacy prediction. Despite their promise, current exosome detection and isolation methods are cumbersome and time-consuming and often yield limited purity and quantity. In this study, we employed Raman spectroscopy to analyze the molecular changes in exosomes from the sera of HER2-positive breast cancer patients before and after two cycles of neoadjuvant therapy. Utilizing machine learning techniques (PCA, LDA, and SVM), we developed a predictive model with an AUC value exceeding 0.89. Additionally, we introduced an innovative HER2-positive exosome capture and detection system, termed Magnetic beads@HER2-Exos@HER2-SERS detection nanoprobes (HER2-MEDN). This system enabled us to efficiently extract and analyze HER2-positive exosomes, refining our predictive model to achieve an accuracy greater than 0.94. Our study has demonstrated the potential of the HER2-MEDN system in accurately predicting early treatment response, offering novel insights and methodologies for assessing the efficacy of neoadjuvant therapy in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Yining Jia
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| | - Yongqi Li
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| | - Xintong Bai
- School of Mathematics, Shandong University, Jinan, Shandong 250100, China
| | - Liyuan Liu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| | - Ying Shan
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| | - Fei Wang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| | - Chao Zheng
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, Shandong 250033, China
| |
Collapse
|
11
|
Yu Y, Liu X, Zhang W, Sun Z, Zhu Z, Zhang GJ. A PLA-mediated transistor biosensor enables specific identification of tumor-derived exosomal PD-L1. Biosens Bioelectron 2025; 267:116846. [PMID: 39418866 DOI: 10.1016/j.bios.2024.116846] [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: 07/08/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
The expression of programmed death ligand 1 (PD-L1) on tumor-derived exosomes (tExos) forecasts the efficacy of immunotherapy and tumor diagnosis. Due to the heterogeneity of exosomes, current detection methods face challenges in distinguishing between tumor-derived and non-tumor-derived exosome PD-L1. To address this challenge, we introduce a novel field effect transistor (FET) biosensor based on proximity ligation assay (PLA) technology. This approach uses a single probe to simultaneously recognize two biomarkers on exosomes to identify tumor-derived exosome PD-L1 (tExo-PD-L1). This method, for the first time, integrates the PLA strategy with FET technology, allowing for tracking of exosomes that co-express multiple biomarkers. In clinical diagnostics, this strategy not only significantly improves the sensitivity and specificity, but also enhances the precision and accuracy, compared to conventional approaches that identify total Exo-PD-L1 or Exo-EpCAM using a single biomarker. This technology holds promise for enhancing the reliability of using exosomes as biomarkers in clinical diagnostics and further exploring the biological functions of exosomes more effectively.
Collapse
Affiliation(s)
- Yi Yu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Xi Liu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Wenhao Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Zhongyue Sun
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of PhysicalChemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, PR China.
| |
Collapse
|
12
|
Liu X, Zhang J, Chen Z, He X, Yan C, Lv H, Chen Z, Liu Y, Wang L, Song C. Branched hybridization chain reaction and tetrahedral DNA-based trivalent aptamer powered SERS sensor for ultra-highly sensitive detection of cancer-derived exosomes. Biosens Bioelectron 2025; 267:116737. [PMID: 39243449 DOI: 10.1016/j.bios.2024.116737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Exosomes have emerged as a promising noninvasive biomarker for early cancer diagnosis due to their ability to carry specific bioinformation related to cancer cells. However, accurate detection of trace amount of cancer-derived exosomes in complex blood remains a significant challenge. Herein, an ultra-highly sensitive SERS sensor, powered by the branched hybridization chain reaction (bHCR) and tetrahedral DNA-based trivalent aptamer (triApt-TDN), has been proposed for precise detection of cancer-derived exosomes. Taking gastric cancer SGC-7901 cells-derived exosomes as a test model, the triApt-TDNs were constructed by conjugating aptamers specific to mucin 1 (MUC1) protein with tetrahedral DNAs and subsequently immobilized on the surface of silver nanorods (AgNRs) arrays to create SERS-active sensing chips capable of specifically capturing exosomes overexpressing MUC1 proteins. The bHCR was further initiated by the trigger aptamers (tgApts) bound to exosomes, and as a result the SERS tags were assembled into AuNP network structures with abundant SERS hotspots. By optimizing the sensing conditions, the SERS sensor showed good performance in ultra-highly sensitive detection of target exosomes within 60 min detection time, with a broad response ranging of 1.44 to 1.44 × 104 particles·μL-1 and an ultralow limit of detection capable of detecting a single exosome in 2 μL sample. Furthermore, the SERS sensor exhibited good uniformity, repeatability and specificity, and capability to distinguish between gastric cancer (GC) patients and healthy controls (HC) through the detection of exosomes in clinical human serums, indicating its promising clinical potential for early diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Xinyu Liu
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Jingjing Zhang
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Zeyan Chen
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Xiyu He
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Chenlong Yan
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Huiming Lv
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Zhilong Chen
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Ying Liu
- Xuzhou College of Industrial Technology, Xuzhou, 221140, China.
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Chunyuan Song
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| |
Collapse
|
13
|
Lu J, Shi X, Zhou Z, Lu N, Chu G, Jin H, Zhu L, Chen A. Enhancing Fracture Healing with 3D Bioprinted Hif1a-Overexpressing BMSCs Hydrogel: A Novel Approach to Accelerated Bone Repair. Adv Healthc Mater 2025; 14:e2402415. [PMID: 39580668 DOI: 10.1002/adhm.202402415] [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: 07/02/2024] [Revised: 10/17/2024] [Indexed: 11/26/2024]
Abstract
Addressing the urgent need for effective fracture treatments, this study investigates the efficacy of a 3D bioprinted biomimetic hydrogel, enriched with bone marrow mesenchymal stem cells (BMSCs) and targeted hypoxia-inducible factor 1 alpha (Hif1a) gene activation, in enhancing fracture healing. A photocross-linkable bioink, gelatin methacryloyl bone matrix anhydride (GBMA) is developed, and selected its 5% concentration for bioink formulation. Rat BMSCs are isolated and combined with GBMA to create the GBMA@BMSCs bioink. This bioink is then used in 3D bioprinting to fabricate a hydrogel for application in a rat femoral fracture model. Through transcriptome sequencing, WGCNA, and Venn analysis, the hypoxia-inducible factor Hif1a is identified as a critical gene in the fracture healing process. In vitro studies showed that Hif1a promoted BMSC proliferation, chondrogenic differentiation, and cartilage matrix stability. The in vivo application of the GBMA@BMSCs hydrogel with Hif1a overexpression significantly accelerated fracture healing, evidenced by early and enhanced cartilage callus formation. The study demonstrates that 3D bioprinting of GBMA@BMSCs hydrogel, particularly with Hif1a-enhanced BMSCs, offers a promising approach for rapid and effective fracture repair.
Collapse
Affiliation(s)
- Jiajia Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200001, P. R. China
| | - Xiaojian Shi
- Department of Orthopedic Trauma, Haimen People's Hospital of Jiangsu Province, Haimen, 226100, P. R. China
| | - Zhibin Zhou
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang, 110016, P. R. China
| | - Nan Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China
| | - Guangxin Chu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Hai Jin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Lei Zhu
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200001, P. R. China
| | - Aimin Chen
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China
| |
Collapse
|
14
|
Zhao X, Qi X, Liu D, Che X, Wu G. A Novel Approach for Bladder Cancer Treatment: Nanoparticles as a Drug Delivery System. Int J Nanomedicine 2024; 19:13461-13483. [PMID: 39713223 PMCID: PMC11662911 DOI: 10.2147/ijn.s498729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Bladder cancer represents one of the most prevalent malignant neoplasms of the urinary tract. In the Asian context, it represents the eighth most common cancer in males. In 2022, there were approximately 613,791 individuals diagnosed with bladder cancer worldwide. Despite the availability of efficacious treatments for the two principal forms of bladder cancer, namely non-invasive and invasive bladder cancer, the high incidence of recurrence following treatment and the suboptimal outcomes observed in patients with high-grade and advanced disease represent significant concerns in the management of bladder cancer at this juncture. Nanoparticles have gained attention for their excellent properties, including stable physical properties, a porous structure that can be loaded with a variety of substances, and so on. The in-depth research on nanoparticles has led to their emergence as a new class of nanoparticles for combination therapy, due to their advantageous properties. These include the extension of the drug release window, the enhancement of drug bioavailability, the improvement of drug targeting ability, the reduction of local and systemic toxicity, and the simultaneous delivery of multiple drugs for combination therapy. As a result, nanoparticles have become a novel agent of the drug delivery system. The advent of nanoparticles has provided a new impetus for the development of non-surgical treatments for bladder cancer, including chemotherapy, immunotherapy, gene therapy and phototherapy. The unique properties of nanoparticles have facilitated the combination of diverse non-surgical therapeutic modalities, enhancing their overall efficacy. This review examines the recent advancements in the use of nanoparticles in non-surgical bladder cancer treatments, encompassing aspects such as delivery, therapeutic efficacy, and the associated toxicity of nanoparticles, as well as the challenges encountered in clinical applications.
Collapse
Affiliation(s)
- Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| |
Collapse
|
15
|
Lin X, Zhu J, Shen J, Zhang Y, Zhu J. Advances in exosome plasmonic sensing: Device integration strategies and AI-aided diagnosis. Biosens Bioelectron 2024; 266:116718. [PMID: 39216205 DOI: 10.1016/j.bios.2024.116718] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Exosomes, as next-generation biomarkers, has great potential in tracking cancer progression. They face many detection limitations in cancer diagnosis. Plasmonic biosensors have attracted considerable attention at the forefront of exosome detection, due to their label-free, real-time, and high-sensitivity features. Their advantages in multiplex immunoassays of minimal liquid samples establish the leading position in various diagnostic studies. This review delineates the application principles of plasmonic sensing technologies, highlighting the importance of exosomes-based spectrum and image signals in disease diagnostics. It also introduces advancements in miniaturizing plasmonic biosensing platforms of exosomes, which can facilitate point-of-care testing for future healthcare. Nowadays, inspired by the surge of artificial intelligence (AI) for science and technology, more and more AI algorithms are being adopted to process the exosome spectrum and image data from plasmonic detection. Using representative algorithms of machine learning has become a mainstream trend in plasmonic biosensing research for exosome liquid biopsy. Typically, these algorithms process complex exosome datasets efficiently and establish powerful predictive models for precise diagnosis. This review further discusses critical strategies of AI algorithm selection in exosome-based diagnosis. Particularly, we categorize the AI algorithms into the interpretable and uninterpretable groups for exosome plasmonic detection applications. The interpretable AI enhances the transparency and reliability of diagnosis by elucidating the decision-making process, while the uninterpretable AI provides high diagnostic accuracy with robust data processing by a "black-box" working mode. We believe that AI will continue to promote significant progress of exosome plasmonic detection and mobile healthcare in the near future.
Collapse
Affiliation(s)
- Xiangyujie Lin
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaheng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaqing Shen
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Youyu Zhang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
16
|
Wu Y, Wang Y, Mo T, Liu Q. Surface-enhanced Raman scattering-based strategies for tumor markers detection: A review. Talanta 2024; 280:126717. [PMID: 39167940 DOI: 10.1016/j.talanta.2024.126717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
The presence of malignant tumors poses a significant threat to people's life and well-being. As biochemical parameters indicate the occurrence and development of tumors, tumor markers play a pivotal role in early cancer detection, treatment, prognosis, efficient monitoring, and other aspects. Surface-enhanced Raman scattering (SERS) is considered a potent tool for the detection of tumor markers owing to its exceptional advantages encompassing high sensitivity, superior selectivity, rapid analysis speed, and photobleaching resistance nature. This review aims to provide a comprehensive understanding of SERS applications in the detection of tumor markers. Firstly, we introduce the SERS enhancement mechanism, classification of active substrates, and SERS detection techniques. Secondly, the latest research progress of in vitro SERS detection of different types of tumor markers in body fluids and the application of SERS imaging in biomedical imaging are highlighted in sections of the review. Finally, according to the current status of SERS detection of tumor markers, the challenges and problems of SERS in biomedical detection are discussed, and insights into future developments in SERS are offered.
Collapse
Affiliation(s)
- Yafang Wu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yinglin Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Tianlu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China.
| | - Qing Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China.
| |
Collapse
|
17
|
An J, Park H, Ju M, Woo Y, Seo Y, Min J, Lee T. An updated review on the development of a nanomaterial-based field-effect transistor-type biosensors to detect exosomes for cancer diagnosis. Talanta 2024; 279:126604. [PMID: 39068827 DOI: 10.1016/j.talanta.2024.126604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Cancer, a life-threatening genetic disease caused by abnormalities in normal cell growth regulatory functions, poses a significant challenge that current medical technologies cannot fully overcome. The current desired breakthrough is to diagnose cancer as early as possible and increase survival rates through treatments tailored to the prognosis and appropriate follow-up. From a perspective that reflects this contemporary paradigm of cancer diagnostics, exosomes are emerging as promising biomarkers. Exosomes, serving as mobile biological information repositories of cancer cells, have been known to create a microtumor environment in surrounding cells, and significant insight into the clinical significance of cancer diagnosis targeting them has been reported. Therefore, there are growing interests in constructing a system that enables continuous screening with a focus on patient-friendly and flexible diagnosis, aiming to improve cancer screening rates through exosome detection. This review focuses on a proposed exosome-embedded biological information-detecting platform employing a field-effect transistor (FET)-based biosensor that leverages portability, cost-effectiveness, and rapidity to minimize the stages of sacrifice attributable to cancer. The FET-applied biosensing technique, stemming from variations in an electric field, is considered an early detection system, offering high sensitivity and a prompt response frequency for the qualitative and quantitative analysis of biomolecules. Hence, an in-depth discussion was conducted on the understanding of various exosome-based cancer biomarkers and the clinical significance of recent studies on FET-based biosensors applying them.
Collapse
Affiliation(s)
- Jeongyun An
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Hyunjun Park
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Minyoung Ju
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Yeeun Woo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Yoshep Seo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Junhong Min
- School of Integrative Engineering, Chung-Ang University, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea.
| |
Collapse
|
18
|
Feng Y, Yang J, He Z, Liu X, Ma C. CRISPR-Cas-based biosensors for the detection of cancer biomarkers. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6634-6653. [PMID: 39258950 DOI: 10.1039/d4ay01446d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Along with discovering cancer biomarkers, non-invasive detection methods have played a critical role in early cancer diagnosis and prognostic improvement. Some traditional detection methods have been used for detecting cancer biomarkers, but they are time-consuming and involve materials and human costs. With great flexibility, sensitivity and specificity, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated system provides a wide range of application prospects in this field. Herein, we introduce the background of the CRISPR-Cas (CRISPR-associated) system and comprehensively summarize the diagnosis strategies of cancer mediated by the CRISPR-Cas system, including four kinds of biochemical-based markers: nucleic acid, enzyme, tumor-specific protein and exosome. Furthermore, we discuss the challenges in implementing the CRISPR-Cas system in clinical applications.
Collapse
Affiliation(s)
- Yuxin Feng
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jinmeng Yang
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Ziping He
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Xinfa Liu
- School of Life Sciences, Central South University, Changsha 410013, China.
| | - Changbei Ma
- School of Life Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
19
|
Cialla-May D, Bonifacio A, Bocklitz T, Markin A, Markina N, Fornasaro S, Dwivedi A, Dib T, Farnesi E, Liu C, Ghosh A, Popp J. Biomedical SERS - the current state and future trends. Chem Soc Rev 2024; 53:8957-8979. [PMID: 39109571 DOI: 10.1039/d4cs00090k] [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: 09/17/2024]
Abstract
Surface enhanced Raman spectroscopy (SERS) is meeting the requirements in biomedical science being a highly sensitive and specific analytical tool. By employing portable Raman systems in combination with customized sample pre-treatment, point-of-care-testing (POCT) becomes feasible. Powerful SERS-active sensing surfaces with high stability and modification layers if required are available for testing and application in complex biological matrices such as body fluids, cells or tissues. This review summarizes the current state in sample collection and pretreatment in SERS detection protocols, SERS detection schemes, i.e. direct and indirect SERS as well as targeted and non-targeted SERS, and SERS-active sensing surfaces. Moreover, the recent developments and advances of SERS in biomedical application scenarios, such as infectious diseases, cancer diagnostics and therapeutic drug monitoring is given, which enables the readers to identify the sample collection and preparation protocols, SERS substrates and detection strategies that are best-suited for their specific applications in biomedicine.
Collapse
Affiliation(s)
- Dana Cialla-May
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Alois Bonifacio
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio 6, 34127 Trieste (TS), Italy
| | - Thomas Bocklitz
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
- Faculty of Mathematics, Physics and Computer Science, University of Bayreuth (UBT), Nürnberger Straße 38, 95440 Bayreuth, Germany
| | - Alexey Markin
- Institute of Chemistry, Saratov State University, Astrakhanskaya Street 83, 410012 Saratov, Russia
| | - Natalia Markina
- Institute of Chemistry, Saratov State University, Astrakhanskaya Street 83, 410012 Saratov, Russia
| | - Stefano Fornasaro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127 Trieste (TS), Italy
| | - Aradhana Dwivedi
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Tony Dib
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Edoardo Farnesi
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Chen Liu
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Arna Ghosh
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Juergen Popp
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany.
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| |
Collapse
|
20
|
Küçük B, Yilmaz EG, Aslan Y, Erdem Ö, Inci F. Shedding Light on Cellular Secrets: A Review of Advanced Optical Biosensing Techniques for Detecting Extracellular Vesicles with a Special Focus on Cancer Diagnosis. ACS APPLIED BIO MATERIALS 2024; 7:5841-5860. [PMID: 39175406 PMCID: PMC11409220 DOI: 10.1021/acsabm.4c00782] [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: 06/11/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
In the relentless pursuit of innovative diagnostic tools for cancer, this review illuminates the cutting-edge realm of extracellular vesicles (EVs) and their biomolecular cargo detection through advanced optical biosensing techniques with a primary emphasis on their significance in cancer diagnosis. From the sophisticated domain of nanomaterials to the precision of surface plasmon resonance, we herein examine the diverse universe of optical biosensors, emphasizing their specified applications in cancer diagnosis. Exploring and understanding the details of EVs, we present innovative applications of enhancing and blending signals, going beyond the limits to sharpen our ability to sense and distinguish with greater sensitivity and specificity. Our special focus on cancer diagnosis underscores the transformative potential of optical biosensors in early detection and personalized medicine. This review aims to help guide researchers, clinicians, and enthusiasts into the captivating domain where light meets cellular secrets, creating innovative opportunities in cancer diagnostics.
Collapse
Affiliation(s)
- Beyza
Nur Küçük
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Eylul Gulsen Yilmaz
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Yusuf Aslan
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Özgecan Erdem
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
| | - Fatih Inci
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| |
Collapse
|
21
|
Hong Y, Yang J, Liu X, Huang S, Liang T, Bai X. Deciphering extracellular vesicles protein cargo in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189142. [PMID: 38914240 DOI: 10.1016/j.bbcan.2024.189142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a significant therapeutic challenge as it is frequently diagnosed at advanced inoperable stages. Therefore, the development of a reliable screening tool for PDAC is crucial for effective prevention and treatment. Extracellular vesicles (EVs), characterized by their cup-shaped lipid bilayer structure and ubiquitous release from various cell types, offer notable advantages as an emerging liquid biopsy technique that is rapid, minimally invasive, easily sampled, and cost-effective. While EVs play a substantial role in cancer progression, EV proteins serve as direct mediators of diverse cellular behaviors and have immense potential as biomarkers for PDAC diagnosis and prognostication. This review provides an overview of EV proteins regarding PDAC diagnosis and prognostic implications as well as disease progression.
Collapse
Affiliation(s)
- Yifan Hong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Jiaqi Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Sicong Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
22
|
Verdin A, Malherbe C, Eppe G. Designing SERS nanotags for profiling overexpressed surface markers on single cancer cells: A review. Talanta 2024; 276:126225. [PMID: 38749157 DOI: 10.1016/j.talanta.2024.126225] [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: 03/13/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/14/2024]
Abstract
This review focuses on the chemical design and the use of Surface-Enhanced Raman Scattering (SERS)-active nanotags for measuring surface markers that can be overexpressed at the surface of single cancer cells. Indeed, providing analytical tools with true single-cell measurements capabilities is capital, especially since cancer research is increasingly leaning toward single-cell analysis, either to guide treatment decisions or to understand complex tumor behaviour including the single-cell heterogeneity and the appearance of treatment resistance. Over the past two decades, SERS nanotags have triggered significant interest in the scientific community owing their advantages over fluorescent tags, mainly because SERS nanotags resist photobleaching and exhibit sharper signal bands, which reduces possible spectral overlap and enables the discrimination between the SERS signals and the autofluorescence background from the sample itself. The extensive efforts invested in harnessing SERS nanotags for biomedical purposes, particularly in cancer research, highlight their potential as the next generation of optical labels for single-cell studies. The review unfolds in two main parts. The first part focuses on the structure of SERS nanotags, detailing their chemical composition and the role of each building block of the tags. The second part explores applications in measuring overexpressed surface markers on single-cells. The latter encompasses studies using single nanotags, multiplexed measurements, quantitative information extraction, monitoring treatment responses, and integrating phenotype measurements with SERS nanotags on single cells isolated from complex biological matrices. This comprehensive review anticipates SERS nanotags to persist as a pivotal technology in advancing single-cell analytical methods, particularly in the context of cancer research and personalized medicine.
Collapse
Affiliation(s)
- Alexandre Verdin
- Mass Spectrometry Laboratory, MolSys Research Unit, University of Liège, Belgium.
| | - Cedric Malherbe
- Mass Spectrometry Laboratory, MolSys Research Unit, University of Liège, Belgium
| | - Gauthier Eppe
- Mass Spectrometry Laboratory, MolSys Research Unit, University of Liège, Belgium
| |
Collapse
|
23
|
Lei H, Wang H, Wang X, Xiao Z, Tian T, Cui K. Surface-enhanced Raman scattering-based identification of breast cancer progression using extracellular vesicles-derived integrin α6β4. Talanta 2024; 275:126092. [PMID: 38615459 DOI: 10.1016/j.talanta.2024.126092] [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: 02/08/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Detection of progression is of great importance to breast cancer treatment and can benefit patients. Limited by current detection technologies and biomarkers, early breast cancer progression diagnosis remains challenging. Researchers have found blood extracellular vesicles (EVs)-derived integrin α6β4 directly facilitate progression in breast cancer, enabling cancer detection. However, EVs size and heterogeneity hinder protein detection, masked by abundant background EVs. Hence, novel tools for efficient detection of EVs with high selectivity and low interference are significantly desired. Here, a new silver-coated gold nanorods SERS probe, termed as Au@Ag@IDA-B/4MSTP, based on DNA aptamer was established for the detection of integrin α6β4 derived from EVs. Validation of the Au@Ag@IDA-B/4MSTP probes using cell-culture-derived EVs revealed a LOD of 23 particles/μL for EVs detection. This tool was further confirmed to mimic the real state of cancer with subcutaneous tumor model and lung metastasis model in mice. With 10 μL of blood plasma and simple Raman analysis process, the test achieved 85.7 % sensitivity and 83.3 % specificity. Moreover, our method achieves a simplified approach that expedites the detection process. These results demonstrate the good detection performance of Au@Ag@IDA-B/4MSTP probes for EVs integrin α6β4, and suggest that this non-invasive approach could be a promising tool for early detection of breast cancer progression.
Collapse
Affiliation(s)
- Haozhi Lei
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Shanghai, 200127, China
| | - Haoze Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China; College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200233, China
| | - Xiqiu Wang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zeyu Xiao
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Tian Tian
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Kai Cui
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
24
|
Wang Z, Zhou X, Kong Q, He H, Sun J, Qiu W, Zhang L, Yang M. Extracellular Vesicle Preparation and Analysis: A State-of-the-Art Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401069. [PMID: 38874129 PMCID: PMC11321646 DOI: 10.1002/advs.202401069] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/11/2024] [Indexed: 06/15/2024]
Abstract
In recent decades, research on Extracellular Vesicles (EVs) has gained prominence in the life sciences due to their critical roles in both health and disease states, offering promising applications in disease diagnosis, drug delivery, and therapy. However, their inherent heterogeneity and complex origins pose significant challenges to their preparation, analysis, and subsequent clinical application. This review is structured to provide an overview of the biogenesis, composition, and various sources of EVs, thereby laying the groundwork for a detailed discussion of contemporary techniques for their preparation and analysis. Particular focus is given to state-of-the-art technologies that employ both microfluidic and non-microfluidic platforms for EV processing. Furthermore, this discourse extends into innovative approaches that incorporate artificial intelligence and cutting-edge electrochemical sensors, with a particular emphasis on single EV analysis. This review proposes current challenges and outlines prospective avenues for future research. The objective is to motivate researchers to innovate and expand methods for the preparation and analysis of EVs, fully unlocking their biomedical potential.
Collapse
Affiliation(s)
- Zesheng Wang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Qinglong Kong
- The Second Department of Thoracic SurgeryDalian Municipal Central HospitalDalian116033P. R. China
| | - Huimin He
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Wenting Qiu
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Liang Zhang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| |
Collapse
|
25
|
Yuan W, Liu W, Huang H, Chen X, Zhang R, Lyu H, Xiao S, Guo D, Zhang Q, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Screening and identification of miRNAs negatively regulating FAM83A/Wnt/β-catenin signaling pathway in non-small cell lung cancer. Sci Rep 2024; 14:17394. [PMID: 39075121 PMCID: PMC11286843 DOI: 10.1038/s41598-024-67686-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
The prevalence of non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancers, with the Wnt/β-catenin signaling pathway exhibiting robust activation in this particular subtype. The expression of FAM83A (family with sequence similarity 83, member A) has been found to be significantly upregulated in lung cancer, leading to the stabilization of β-catenin and activation of the Wnt signaling pathway. In this study, we conducted a screening of down-regulated miRNAs in lung cancer with FAM83A as the target. Ultimately, we identified miR-1 as a negative regulator of FAM83A and confirmed that FAM83A is a direct target gene of miR-1 through dual luciferase reporter assays. The overexpression of miR-1 significantly attenuated the expression level of FAM83A and suppressed the Wnt signaling pathway, leading to a reduction in the expression levels of downstream target genes AXIN2, CyclinD1, and C-MYC. Additionally, it decreased the nuclear translocation of β-catenin. In addition, overexpression of miR-1 accelerated the degradation of β-catenin by inhibiting FAM83A, promoted the assembly of β-catenin degradation complex, and inhibited the proliferation, migration and invasion of NSCLC cells. In summary, miR-1 may be a potential candidate miRNA for the treatment of NSCLC.
Collapse
Affiliation(s)
- Wenbin Yuan
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Wei Liu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Huili Huang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Xingyu Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Rui Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Hao Lyu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Shuai Xiao
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Dong Guo
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Qi Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China.
| | - Jingfeng Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China.
| |
Collapse
|
26
|
Yuan W, Liu W, Huang H, Chen X, Zhang R, Lyu H, Xiao S, Guo D, Zhang Q, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Screening and identification of miRNAs negatively regulating FAM83A/Wnt/β-catenin signaling pathway in non-small cell lung cancer. Sci Rep 2024; 14:17394. [PMID: 39075121 DOI: 10.1038/s41598-024-67686-3.pmid:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 10/14/2024] Open
Abstract
The prevalence of non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancers, with the Wnt/β-catenin signaling pathway exhibiting robust activation in this particular subtype. The expression of FAM83A (family with sequence similarity 83, member A) has been found to be significantly upregulated in lung cancer, leading to the stabilization of β-catenin and activation of the Wnt signaling pathway. In this study, we conducted a screening of down-regulated miRNAs in lung cancer with FAM83A as the target. Ultimately, we identified miR-1 as a negative regulator of FAM83A and confirmed that FAM83A is a direct target gene of miR-1 through dual luciferase reporter assays. The overexpression of miR-1 significantly attenuated the expression level of FAM83A and suppressed the Wnt signaling pathway, leading to a reduction in the expression levels of downstream target genes AXIN2, CyclinD1, and C-MYC. Additionally, it decreased the nuclear translocation of β-catenin. In addition, overexpression of miR-1 accelerated the degradation of β-catenin by inhibiting FAM83A, promoted the assembly of β-catenin degradation complex, and inhibited the proliferation, migration and invasion of NSCLC cells. In summary, miR-1 may be a potential candidate miRNA for the treatment of NSCLC.
Collapse
Affiliation(s)
- Wenbin Yuan
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Wei Liu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Huili Huang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Xingyu Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Rui Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Hao Lyu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Shuai Xiao
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Dong Guo
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Qi Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China.
| | - Jingfeng Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education and Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, People's Republic of China.
| |
Collapse
|
27
|
Chang YJ, Yang WT, Lei CH. Identification and Quantification of Extracellular Vesicles: Comparison of SDS-PAGE Analysis and Biosensor Analysis with QCM and IDT Chips. BIOSENSORS 2024; 14:366. [PMID: 39194595 DOI: 10.3390/bios14080366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
This study presents and compares two methods for identifying the types of extracellular vesicles (EVs) from different cell lines. Through SDS-PAGE analysis, we discovered that the ratio of CD63 to CD81 in different EVs is consistent and distinct, making it a reliable characteristic for recognizing EVs secreted by cancer cells. However, the electrophoresis and imaging processes may introduce errors in the concentration values, especially at lower concentrations, rendering this method potentially less effective. An alternative approach involves the use of quartz crystal microbalance (QCM) and electroanalytical interdigitated electrode (IDT) biosensors for EV type identification and quantification. The QCM frequency shift caused by EVs is directly proportional to their concentration, while electroanalysis relies on measuring the curvature of the I-V curve as a distinguishing feature, which is also proportional to EV concentration. Linear regression lines for the QCM frequency shift and the electroanalysis curvature of various EV types are plotted separately, enabling the estimation of the corresponding concentration for an unknown EV type on the graphs. By intersecting the results from both biosensors, the unknown EV type can be identified. The biosensor analysis method proves to be an effective means of analyzing both the type and concentration of EVs from different cell lines.
Collapse
Affiliation(s)
- Yaw-Jen Chang
- Department of Mechanical Engineering, Chung Yuan Christian University, Chung Li District, Taoyuan City 320314, Taiwan
| | - Wen-Tung Yang
- Department of Mechanical Engineering, Chung Yuan Christian University, Chung Li District, Taoyuan City 320314, Taiwan
| | - Cheng-Hsuan Lei
- Department of Mechanical Engineering, Chung Yuan Christian University, Chung Li District, Taoyuan City 320314, Taiwan
| |
Collapse
|
28
|
Kazemzadeh M, Martinez-Calderon M, Otupiri R, Artuyants A, Lowe M, Ning X, Reategui E, Schultz ZD, Xu W, Blenkiron C, Chamley LW, Broderick NGR, Hisey CL. Deep autoencoder as an interpretable tool for Raman spectroscopy investigation of chemical and extracellular vesicle mixtures. BIOMEDICAL OPTICS EXPRESS 2024; 15:4220-4236. [PMID: 39022543 PMCID: PMC11249694 DOI: 10.1364/boe.522376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 07/20/2024]
Abstract
Surface-enhanced Raman spectroscopy (SERS) is a powerful tool that provides valuable insight into the molecular contents of chemical and biological samples. However, interpreting Raman spectra from complex or dynamic datasets remains challenging, particularly for highly heterogeneous biological samples like extracellular vesicles (EVs). To overcome this, we developed a tunable and interpretable deep autoencoder for the analysis of several challenging Raman spectroscopy applications, including synthetic datasets, chemical mixtures, a chemical milling reaction, and mixtures of EVs. We compared the results with classical methods (PCA and UMAP) to demonstrate the superior performance of the proposed technique. Our method can handle small datasets, provide a high degree of generalization such that it can fill unknown gaps within spectral datasets, and even quantify relative ratios of cell line-derived EVs to fetal bovine serum-derived EVs within mixtures. This simple yet robust approach will greatly improve the analysis capabilities for many other Raman spectroscopy applications.
Collapse
Affiliation(s)
- Mohammadrahim Kazemzadeh
- Department of Mechanical and Mechatronics Engineering, University of Auckland, Auckland 1010, New Zealand
- Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9016, New Zealand
| | | | - Robert Otupiri
- Photon Factory, University of Auckland, Auckland 1010, New Zealand
| | - Anastasiia Artuyants
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
| | - MoiMoi Lowe
- Photon Factory, University of Auckland, Auckland 1010, New Zealand
| | - Xia Ning
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Reategui
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Zachary D Schultz
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Weiliang Xu
- Department of Mechanical and Mechatronics Engineering, University of Auckland, Auckland 1010, New Zealand
- Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9016, New Zealand
| | - Cherie Blenkiron
- Auckland Cancer Society Research Centre, Auckland 1023, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
| | - Neil G R Broderick
- Dodd-Walls Centre for Photonic and Quantum Technologies, Dunedin 9016, New Zealand
- Photon Factory, University of Auckland, Auckland 1010, New Zealand
| | - Colin L Hisey
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
29
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
30
|
Stawarska A, Bamburowicz-Klimkowska M, Runden-Pran E, Dusinska M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Extracellular Vesicles as Next-Generation Diagnostics and Advanced Therapy Medicinal Products. Int J Mol Sci 2024; 25:6533. [PMID: 38928240 PMCID: PMC11204223 DOI: 10.3390/ijms25126533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Extracellular vesicles (EVs) hold great promise for clinical application as new diagnostic and therapeutic modalities. This paper describes major GMP-based upstream and downstream manufacturing processes for EV large-scale production, also focusing on post-processing technologies such as surface bioengineering and uploading studies to yield novel EV-based diagnostics and advanced therapy medicinal products. This paper also focuses on the quality, safety, and efficacy issues of the bioengineered EV drug candidates before first-in-human studies. Because clinical trials involving extracellular vesicles are on the global rise, this paper encompasses different clinical studies registered on clinical-trial register platforms, with varying levels of advancement, highlighting the growing interest in EV-related clinical programs. Navigating the regulatory affairs of EVs poses real challenges, and obtaining marketing authorization for EV-based medicines remains complex due to the lack of specific regulatory guidelines for such novel products. This paper discusses the state-of-the-art regulatory knowledge to date on EV-based diagnostics and medicinal products, highlighting further research and global regulatory needs for the safe and reliable implementation of bioengineered EVs as diagnostic and therapeutic tools in clinical settings. Post-marketing pharmacovigilance for EV-based medicinal products is also presented, mainly addressing such topics as risk assessment and risk management.
Collapse
Affiliation(s)
- Agnieszka Stawarska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, 02-097 Warsaw, Poland; (M.B.-K.); (I.P.G.)
| | - Magdalena Bamburowicz-Klimkowska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, 02-097 Warsaw, Poland; (M.B.-K.); (I.P.G.)
| | - Elise Runden-Pran
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (M.D.)
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (M.D.)
| | - Mihaela Roxana Cimpan
- Biomaterials—Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien Str. 19, 5009 Bergen, Norway; (M.R.C.); (I.R.-M.)
| | - Ivan Rios-Mondragon
- Biomaterials—Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien Str. 19, 5009 Bergen, Norway; (M.R.C.); (I.R.-M.)
| | - Ireneusz P. Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, 02-097 Warsaw, Poland; (M.B.-K.); (I.P.G.)
| |
Collapse
|
31
|
Davis EHS, Jones C, Coward K. Rethinking the application of nanoparticles in women's reproductive health and assisted reproduction. Nanomedicine (Lond) 2024; 19:1231-1251. [PMID: 38686941 PMCID: PMC11285225 DOI: 10.2217/nnm-2023-0346] [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: 11/29/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
Nanoparticles and nanotechnology may present opportunities to revolutionize the prevention, treatment and diagnosis of a range of reproductive health conditions in women. These technologies are also used to improve outcomes of assisted reproductive technology. We highlight a range of these potential clinical uses of nanoparticles for polycystic ovary syndrome, endometriosis, uterine fibroids and sexually transmitted infections, considering in vitro and in vivo studies along with clinical trials. In addition, we discuss applications of nanoparticles in assisted reproductive technology, including sperm loading, gamete and embryo preservation and preventing preterm birth. Finally, we present some of the concerns associated with the medical use of nanoparticles, identifying routes for further exploration before nanoparticles can be applied to women's reproductive health in the clinic.
Collapse
Affiliation(s)
- Emily HS Davis
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Celine Jones
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Kevin Coward
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
32
|
Al-Hawary SIS, Almajidi YQ, Bansal P, Ahmad I, Kaur H, Hjazi A, Deorari M, Zwamel AH, Hamzah HF, Mohammed BA. Dendritic cell-derived exosome (DEX) therapy for digestive system cancers: Recent advances and future prospect. Pathol Res Pract 2024; 257:155288. [PMID: 38653088 DOI: 10.1016/j.prp.2024.155288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/10/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
Tumor-mediated immunosuppression is a fundamental obstacle to the development of dendritic cell (DC)-based cancer vaccines, which despite their ability to stimulate host anti-tumor CD8 T cell immunity, have not been able to generate meaningful therapeutic responses. Exosomes are inactive membrane vesicles that are nanoscale in size and are produced by the endocytic pathway. They are essential for intercellular communication. Additionally, DC-derived exosomes (DEXs) contained MHC class I/II (MHCI/II), which is frequently complexed with antigens and co-stimulatory molecules and is therefore able to prime CD4 and CD8 T cells that are specific to particular antigens. Indeed, vaccines with DEXs have been shown to exhibit better anti-tumor efficacy in eradicating tumors compared to DC vaccines in pre-clinical models of digestive system tumors. Also, there is room for improvement in the tumor antigenic peptide (TAA) selection process. DCs release highly targeted exosomes when the right antigenic peptide is chosen, which could aid in the creation of DEX-based antitumor vaccines that elicit more targeted immune responses. Coupled with their resistance to tumor immunosuppression, DEXs-based cancer vaccines have been heralded as the superior alternative cell-free therapeutic vaccines over DC vaccines to treat digestive system tumors. In this review, current studies of DEXs cancer vaccines as well as potential future directions will be deliberated.
Collapse
Affiliation(s)
| | - Yasir Qasim Almajidi
- Department of pharmacy (pharmaceutics), Baghdad College of Medical Sciences, Baghdad, Iraq.
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University Al-Kharj 11942, Saudi Arabia
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ahmed Hussein Zwamel
- Department of Medical Laboratory Technology, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | |
Collapse
|
33
|
Shen J, Ma Z, Xu J, Xue T, Lv X, Zhu G, Huang B. Exosome Isolation and Detection: From Microfluidic Chips to Nanoplasmonic Biosensor. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38676635 DOI: 10.1021/acsami.3c19396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Exosomes are becoming more widely acknowledged as significant circulating indicators for the prognosis and diagnosis of cancer. Circulating exosomes are essential to the development and spread of cancer, according to a growing body of research. Using existing technology, characterizing exosomes is quite difficult. Therefore, a direct, sensitive, and targeted approach to exosome detection will aid in illness diagnosis and prognosis. The review discusses the new strategies for exosome isolation and detection technologies from microfluidic chips to nanoplasmonic biosensors, analyzing the advantages and limitations of these new technologies. This review serves researchers to better understand exosome isolation and detection methods and to help develop better exosome isolating and detecting devices for clinical applications.
Collapse
Affiliation(s)
- Jianing Shen
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Zhengtai Ma
- Key Laboratory of Optoelectronic Materials and Devices, Chinese Academy of Sciences, Beijing 100083, China
- College of Materials Science and Optoelectronic Technology, University of Chinese, Academy of Sciences, Beijing 100049, China
| | - Jiaqi Xu
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Tianhao Xue
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Xiaoqing Lv
- Key Laboratory of Optoelectronic Materials and Devices, Chinese Academy of Sciences, Beijing 100083, China
| | - Guixian Zhu
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Beiju Huang
- Key Laboratory of Optoelectronic Materials and Devices, Chinese Academy of Sciences, Beijing 100083, China
- College of Materials Science and Optoelectronic Technology, University of Chinese, Academy of Sciences, Beijing 100049, China
| |
Collapse
|
34
|
Agnihotram R, Dhar R, Dhar D, Purushothaman K, Narasimhan AK, Devi A. Fusion of Exosomes and Nanotechnology: Cutting-Edge Cancer Theranostics. ACS APPLIED NANO MATERIALS 2024; 7:8489-8506. [DOI: 10.1021/acsanm.4c01033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Rohan Agnihotram
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Debolina Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Kaavya Purushothaman
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Ashwin Kumar Narasimhan
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| |
Collapse
|
35
|
Saleh RO, Hjazi A, Bansal P, Ahmad I, Kaur H, Ali SHJ, Deorari M, Abosaoda MK, Hamzah HF, Mohammed BA. Mysterious interactions between macrophage-derived exosomes and tumors; what do we know? Pathol Res Pract 2024; 256:155261. [PMID: 38518733 DOI: 10.1016/j.prp.2024.155261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/24/2024]
Abstract
Through their ability to modify the tumor microenvironment and cancer cells, macrophages play a crucial role in the promotion of tumorigenesis, development of tumors and metastasis, and chemotherapy resistance. A growing body of research has indicated that exosomes may be essential for coordinating the communication between cancer cells and macrophages. One type of extracellular vehicle called an exosome is utilized for delivering a variety of molecules, such as proteins, lipids, and nucleic acids, to specific cells in order to produce pleiotropic effects. Exosomes derived from macrophages exhibit heterogeneity across various cancer types and function paradoxically, suppressing tumor growth while stimulating it, primarily through post-transcriptional control and protein phosphorylation regulation in the receiving cells. Exosomes released by various macrophage phenotypes offer a variety of therapeutic alternatives in the interim. We outlined the most recent developments in this article, including our understanding of the roles that mechanisms and macrophage-derived exosomal biogenesis play in mediating the progression of cancer and their possible therapeutic uses.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India.
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India.
| | - Saad Hayif Jasim Ali
- Department of medical laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq.
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| | - Munther Kadhim Abosaoda
- College of pharmacy, the Islamic University, Najaf, Iraq; College of pharmacy, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of pharmacy, the Islamic University of Babylon, Al Diwaniyah, Iraq.
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq.
| | | |
Collapse
|
36
|
Cao Y, Xiong J, Du Y, Tang Y, Yin L. Raman spectroscopy combined with multivariate statistical algorithms for the simultaneous screening of cervical and breast cancers. Lasers Med Sci 2024; 39:68. [PMID: 38374512 DOI: 10.1007/s10103-024-04019-w] [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: 11/24/2023] [Accepted: 02/11/2024] [Indexed: 02/21/2024]
Abstract
Breast and cervical cancers are becoming the leading causes of death among women worldwide, but current diagnostic methods have many drawbacks, such as being time-consuming and high cost. Raman spectroscopy, as a rapid, reliable, and non-destructive spectroscopic detection technique, has achieved many breakthrough results in the screening and prognosis of various cancer tumors. Therefore, in this study, Raman spectroscopy technology was used to diagnose breast cancer and cervical cancer. A total of 225 spectra were recorded from 87 patients with cervical cancer, 60 patients with breast cancer, and 78 healthy individuals. The obvious difference in Raman spectrum between the three groups was mainly shown at 809 cm-1 (tyrosine), 958 cm-1 (carotenoid), 1004 cm-1 (phenylalanine), 1154 cm-1 (β-carotene), 1267 cm-1 (Amide III), 1445 cm-1 (phospholipids), 1515 cm-1 (β-carotene), and 1585 cm-1 (C = C olefinic stretch). We used one-way analysis of variance for these peaks and demonstrated that they were significantly different. Then, we combined the detected Raman spectra with multivariate statistical calculations using the principal component analysis-linear discrimination algorithm (PCA-LDA) to discriminate between the three groups of collected serum samples. The diagnostic results showed that the model's accuracy, precision, recall, and F1 score of the model were 92.90%, 92.62%, 92.10%, and 92.36%, respectively. These results suggest that Raman spectroscopy can achieve ultra-sensitive detection of serum, and the developed diagnostic models have great potential for the prognosis and simultaneous screening of cervical and breast cancers.
Collapse
Affiliation(s)
- Yue Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Jiaran Xiong
- School of Electronic Engineering, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Yu Du
- School of Electronic Engineering, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Yishu Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
| | - Longfei Yin
- School of Electronic Engineering, Beijing University of Posts and Telecommunications, Beijing, 100876, China.
| |
Collapse
|
37
|
Li X, Jing Z, Li X, Liu L, Xiao X, Zhong Y, Ren Z. The role of exosomes in cancer-related programmed cell death. Immunol Rev 2024; 321:169-180. [PMID: 37950410 DOI: 10.1111/imr.13286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Cancer arises from the growth and division of uncontrolled erroneous cells. Programmed cell death (PCD), or regulated cell death (RCD), includes natural processes that eliminate damaged or abnormal cells. Dysregulation of PCD is a hallmark of cancer, as cancer cells often evade cell death and continue to proliferate. Exosomes nanoscale extracellular vesicles secreted by different types of cells carrying a variety of molecules, including nucleic acids, proteins, and lipids, to have indispensable role in the communication between cells, and can influence various cellular processes, including PCD. Exosomes have been shown to modulate PCD in cancer cells by transferring pro- or antideath molecules to neighboring cells. Additionally, exosomes can facilitate the spread of PCD to surrounding cancer cells, making them promising in the treatment of various cancers. The exosomes' diagnostic potential in cancer is also an active area of research. Exosomes can be isolated from a wide range of bodily fluids and tissues, such as blood and urine, and can provide a noninvasive way to monitor cancer progression and treatment response. Furthermore, exosomes have also been employed as a delivery system for therapeutic agents. By engineering exosomes to carry drugs or other therapeutic molecules, they can be targeted specifically to cancer cells, reducing toxicity to healthy tissues. Here, we discussed exosomes in the diagnosis and prevention of cancers, tumor immunotherapy, and drug delivery, as well as in different types of PCD.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Zuoqian Jing
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Xuejie Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Lei Liu
- Department of Ophthalmology, Guangdong provincial People's hospital, Guangzhou, China
| | - Xiang Xiao
- Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Yifan Zhong
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Zihan Ren
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
38
|
Liu X, Cheng H, Zhao Y, Wang Y, Ge L, Huang Y, Li F. Immobilization-free dual-aptamer-based photoelectrochemical platform for ultrasensitive exosome assay. Talanta 2024; 266:125001. [PMID: 37517342 DOI: 10.1016/j.talanta.2023.125001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
Exosomes, involved in cancer-specific biological processes, are promising noninvasive biomarkers for early diagnosis of cancer. Herein, an immobilization-free dual-aptamer-based photoelectrochemical (PEC) biosensor was proposed for the enrichment and quantification of cancer exosome based on photoactive bismuch oxyiodide/gold/cadmium sulfide (BiOI/Au/CdS) composites, nucleic acid-based recognition and signal amplification. In this biosensor, the recognition of exosome by two aptamers would trigger the deoxyribonucleotidyl transferase (TdT) enzyme-aided polymerization, leading to the enrichment of alkaline phosphatase (ALP) on Fe3O4 surface. After magnetic separation, ALP could catalyze the generation of ascorbic acid (AA) as electron donor and initiate the following redox cycle reaction for further signal amplification. Furthermore, all the above processes were performed in solution, the recognition and signal amplification efficiency would be superior than the heterogeneous strategy owing to the avoidance of steric hindrance effect. As a result, the proposed PEC biosensor was capable of enriching and detecting of cancer exosomes with high sensitivity and selectivity. The linear range of the biosensor was from 1.0 × 102 particles·μL-1 to 1.0 × 106 particles·μL-1 and the detection limit was estimated to be 21 particles·μL-1. Therefore, the proposed PEC biosensor holds great promise in quantifying tumor exosome for nondestructive early clinical cancer diagnosis and various other bioassay applications.
Collapse
Affiliation(s)
- Xiaojuan Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Hao Cheng
- College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yuecan Zhao
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yue Wang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Lei Ge
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yiping Huang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Feng Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.
| |
Collapse
|
39
|
Bamodu OA, Chung CC, Pisanic TR. Harnessing liquid biopsies: Exosomes and ctDNA as minimally invasive biomarkers for precision cancer medicine. THE JOURNAL OF LIQUID BIOPSY 2023; 2:100126. [PMID: 40028482 PMCID: PMC11863985 DOI: 10.1016/j.jlb.2023.100126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 03/05/2025]
Abstract
Liquid biopsies have emerged as groundbreaking tools for minimally invasive monitoring of cancer, encompassing the analysis of Cell-Free DNA (cfDNA), circulating tumor DNA (ctDNA) and exosomes. This paradigm shift offers an emerging approach for understanding tumor dynamics, treatment responses, and disease progression. Leveraging advancements in molecular biology and technology, liquid biopsies enable clinicians to gain intricate insights from peripheral blood, thereby transforming the landscape of cancer care. This review describes the clinical impact, technological innovations, and recent evidence surrounding the integration of ctDNA and exosome analysis in cancer monitoring. Through early detection, real-time treatment response assessment, and the tracking of minimal residual disease, liquid biopsies have redefined the standards of precision oncology. Key advancements in ctDNA analysis, such as high-throughput sequencing and digital PCR, empower the detection of actionable mutations with high sensitivity. Concurrently, the characterization of exosomal cargo, facilitated by next-generation sequencing and mass spectrometry, unveils the molecular nuances of tumors. Recent studies underscore the utility of these approaches, demonstrating their efficacy in predicting relapse, guiding therapeutic decisions, and ultimately improving patient outcomes. As the field continues to evolve, liquid biopsies hold promise not only as diagnostic tools but also as agents of personalized medicine, enabling precise navigation of the intricate landscape of cancer with minimally invasiveness.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Directorate of Postgraduate Studies, School of Medicine, Muhimbili University of Health and Allied Sciences, Ilala District, Dar es Salaam, Tanzania
- Ocean Road Cancer Institute, Ilala District, Dar es Salaam, Tanzania
| | - Chen-Chih Chung
- Department of Neurology, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, 110, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Oncology - Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
40
|
Zhang W, He Y, Zhang Y. CircRNA in ocular neovascular diseases: Fundamental mechanism and clinical potential. Pharmacol Res 2023; 197:106946. [PMID: 37797661 DOI: 10.1016/j.phrs.2023.106946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
Ocular neovascular disease (OND), characterized by the aberrant formation of immature blood vessels, is the leading cause of vision impairment and blindness. It is important to find effective ways to diagnose and treat these diseases. Circular RNA (circRNA) is a group of endogenous non-coding RNA that play a crucial role in regulating different biological processes. Due to their close association with ocular disease and angiogenesis, circRNAs have become a hotspot in OND research. In this review, we intensively investigate the possibility of using circRNAs in the management of ONDs. In general, angiogenesis is divided into five phases. On the basis of these five steps, we describe the potential of using circRNAs by introducing how they regulate angiogenesis. Subsequently, the interactions between circRNAs and ONDs, including pterygium, corneal neovascularization, age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity, are analyzed in detail. We also introduce the potential use of circRNAs as OND diagnostic biomarkers. Finally, we summarize the prospects of using circRNAs as a potential strategy in OND management. The gaps in recent research are also pointed out with the purpose of promoting the introduction of circRNAs into clinical applications.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Ophthalmology, 2nd Hospital of Jilin University, Changchun 130041, Jilin, China
| | - Yuxi He
- Department of Ophthalmology, 2nd Hospital of Jilin University, Changchun 130041, Jilin, China
| | - Yan Zhang
- Department of Ophthalmology, 2nd Hospital of Jilin University, Changchun 130041, Jilin, China; Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
41
|
Yang X, Xie X, Liu S, Ma W, Zheng Z, Wei H, Yu CY. Engineered Exosomes as Theranostic Platforms for Cancer Treatment. ACS Biomater Sci Eng 2023; 9:5479-5503. [PMID: 37695590 DOI: 10.1021/acsbiomaterials.3c00745] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Tremendous progress in nanotechnology and nanomedicine has made a significant positive effect on cancer treatment by integrating multicomponents into a single multifunctional nanosized delivery system for combinatorial therapies. Although numerous nanocarriers developed so far have achieved excellent therapeutic performance in mouse models via elegant integration of chemotherapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, their synthetic origin may still cause systemic toxicity, immunogenicity, and preferential detection or elimination by the immune system. Exosomes, endogenous nanosized particles secreted by multiple biological cells, could be absorbed by recipient cells to facilitate intercellular communication and content delivery. Therefore, exosomes have emerged as novel cargo delivery tools and attracted considerable attention for cancer diagnosis and treatment due to their innate stability, biological compatibility, and biomembrane penetration capacity. Exosome-related properties and functions have been well-documented; however, there are few reviews, to our knowledge, with a focus on the combination of exosomes and nanotechnology for the development of exosome-based theranostic platforms. To make a timely review on this hot subject of research, we summarize the basic information, isolation and functionalization methodologies, diagnostic and therapeutic potential of exosomes in various cancers with an emphasis on the description of exosome-related nanomedicine for cancer theranostics. The existing appealing challenges and outlook in exosome clinical translation are finally introduced. Advanced biotechnology and nanotechnology will definitely not only promote the integration of intrinsic advantages of natural nanosized exosomes with traditional synthetic nanomaterials for modulated precise cancer treatment but also contribute to the clinical translations of exosome-based nanomedicine as theranostic nanoplatforms.
Collapse
Affiliation(s)
- Xu Yang
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Xiangyu Xie
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Songbin Liu
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Wei Ma
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Zhi Zheng
- Postdoctoral Research Station of Basic Medicine, Hengyang Medical College, College of Chemistry and Chemical Engineering, Hunan Province Cooperative, Hengyang, Hunan 421001, China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Hua Wei
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| | - Cui-Yun Yu
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
42
|
Sridharan G, Atchudan R, Magesh V, Arya S, Ganapathy D, Nallaswamy D, Sundramoorthy AK. Advanced electrocatalytic materials based biosensors for cancer cell detection – A review. ELECTROANAL 2023; 35. [DOI: 10.1002/elan.202300093] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/22/2023] [Indexed: 01/03/2025]
Abstract
AbstractHerein, we have highlighted the latest developments on biosensors for cancer cell detection. Electrochemical (EC) biosensors offer several advantages such as high sensitivity, selectivity, rapid analysis, portability, low‐cost, etc. Generally, biosensors could be classified into other basic categories such as immunosensors, aptasensors, cytosensors, electrochemiluminescence (ECL), and photo‐electrochemical (PEC) sensors. The significance of the EC biosensors is that they could detect several biomolecules in human body including cholesterol, glucose, lactate, uric acid, DNA, blood ketones, hemoglobin, and others. Recently, various EC biosensors have been developed by using electrocatalytic materials such as silver sulfide (Ag2S), black phosphene (BPene), hexagonal carbon nitrogen tube (HCNT), carbon dots (CDs)/cobalt oxy‐hydroxide (CoOOH), cuprous oxide (Cu2O), polymer dots (PDs), manganese oxide (MnO2), graphene derivatives, and gold nanoparticles (Au‐NPs). In some cases, these newly developed biosensors could be able to detect cancer cells with a limit of detection (LOD) of 1 cell/mL. In addition, many remaining challenges have to be addressed and validated by testing more real samples and confirm that these EC biosensors are more accurate and reliable to measure cancer cells in the blood and salivary samples.
Collapse
Affiliation(s)
- Gokul Sridharan
- Centre for Nano-Biosensors Department of Prosthodontics Saveetha Dental College and Hospitals Saveetha Institute of Medical and Technical Sciences Poonamallee High Road Velappanchavadi, Chennai 600077, Tamil Nadu India
| | - Raji Atchudan
- School of Chemical Engineering Yeungnam University Gyeongsan 38541 Korea
| | - Vasanth Magesh
- Centre for Nano-Biosensors Department of Prosthodontics Saveetha Dental College and Hospitals Saveetha Institute of Medical and Technical Sciences Poonamallee High Road Velappanchavadi, Chennai 600077, Tamil Nadu India
| | - Sandeep Arya
- Department of Physics University of Jammu Jammu, And Kashmir 180006 Jammu India
| | - Dhanraj Ganapathy
- Centre for Nano-Biosensors Department of Prosthodontics Saveetha Dental College and Hospitals Saveetha Institute of Medical and Technical Sciences Poonamallee High Road Velappanchavadi, Chennai 600077, Tamil Nadu India
| | - Deepak Nallaswamy
- Centre for Nano-Biosensors Department of Prosthodontics Saveetha Dental College and Hospitals Saveetha Institute of Medical and Technical Sciences Poonamallee High Road Velappanchavadi, Chennai 600077, Tamil Nadu India
| | - Ashok K. Sundramoorthy
- Centre for Nano-Biosensors Department of Prosthodontics Saveetha Dental College and Hospitals Saveetha Institute of Medical and Technical Sciences Poonamallee High Road Velappanchavadi, Chennai 600077, Tamil Nadu India
| |
Collapse
|
43
|
Diao L, Liu M. Rethinking Antigen Source: Cancer Vaccines Based on Whole Tumor Cell/tissue Lysate or Whole Tumor Cell. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300121. [PMID: 37254712 PMCID: PMC10401146 DOI: 10.1002/advs.202300121] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/29/2023] [Indexed: 06/01/2023]
Abstract
Cancer immunotherapies have improved human health, and one among the important technologies for cancer immunotherapy is cancer vaccine. Antigens are the most important components in cancer vaccines. Generally, antigens in cancer vaccines can be divided into two categories: pre-defined antigens and unidentified antigens. Although, cancer vaccines loaded with predefined antigens are commonly used, cancer vaccine loaded with mixed unidentified antigens, especially whole cancer cells or cancer cell lysates, is a very promising approach, and such vaccine can obviate some limitations in cancer vaccines. Their advantages include, but are not limited to, the inclusion of pan-spectra (all or most kinds of) antigens, inducing pan-clones specific T cells, and overcoming the heterogeneity of cancer cells. In this review, the recent advances in cancer vaccines based on whole-tumor antigens, either based on whole cancer cells or whole cancer cell lysates, are summarized. In terms of whole cancer cell lysates, the focus is on applying whole water-soluble cell lysates as antigens. Recently, utilizing the whole cancer cell lysates as antigens in cancer vaccines has become feasible. Considering that pre-determined antigen-based cancer vaccines (mainly peptide-based or mRNA-based) have various limitations, developing cancer vaccines based on whole-tumor antigens is a promising alternative.
Collapse
Affiliation(s)
- Lu Diao
- Department of PharmaceuticsCollege of Pharmaceutical Sciences, Soochow University199 of Ren ai RoadSuzhouJiangsu215123P. R. China
- Kunshan Hospital of Traditional Chinese MedicineKunshanJiangsu215300P. R. China
- Suzhou Ersheng Biopharmaceutical Co., Ltd.Suzhou215123P. R. China
| | - Mi Liu
- Department of PharmaceuticsCollege of Pharmaceutical Sciences, Soochow University199 of Ren ai RoadSuzhouJiangsu215123P. R. China
- Kunshan Hospital of Traditional Chinese MedicineKunshanJiangsu215300P. R. China
- Suzhou Ersheng Biopharmaceutical Co., Ltd.Suzhou215123P. R. China
| |
Collapse
|
44
|
Oshima Y, Haruki T, Koizumi K, Yonezawa S, Taketani A, Kadowaki M, Saito S. Practices, Potential, and Perspectives for Detecting Predisease Using Raman Spectroscopy. Int J Mol Sci 2023; 24:12170. [PMID: 37569541 PMCID: PMC10418989 DOI: 10.3390/ijms241512170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Raman spectroscopy shows great potential for practical clinical applications. By analyzing the structure and composition of molecules through real-time, non-destructive measurements of the scattered light from living cells and tissues, it offers valuable insights. The Raman spectral data directly link to the molecular composition of the cells and tissues and provides a "molecular fingerprint" for various disease states. This review focuses on the practical and clinical applications of Raman spectroscopy, especially in the early detection of human diseases. Identifying predisease, which marks the transition from a healthy to a disease state, is crucial for effective interventions to prevent disease onset. Raman spectroscopy can reveal biological processes occurring during the transition states and may eventually detect the molecular dynamics in predisease conditions.
Collapse
Affiliation(s)
- Yusuke Oshima
- Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
- Faculty of Medicine, Oita University, Yufu 879-5593, Japan
| | - Takayuki Haruki
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
- Faculty of Sustainable Design, University of Toyama, Toyama 930-8555, Japan
| | - Keiichi Koizumi
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-8555, Japan
| | - Shota Yonezawa
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
| | - Akinori Taketani
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
| | - Makoto Kadowaki
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
| | - Shigeru Saito
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan
| |
Collapse
|
45
|
Zheng J, Hu X, Zeng Y, Zhang B, Sun Z, Liu X, Zheng W, Chai Y. Review of the advances in lipid anchors-based biosensors for the isolation and detection of exosomes. Anal Chim Acta 2023; 1263:341319. [PMID: 37225343 DOI: 10.1016/j.aca.2023.341319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/26/2023]
Abstract
Exosomes are nanoparticles with a bilayer lipid structure that carry cargo from their cells of origin. These vesicles are vital to disease diagnosis and therapeutics; however, conventional isolation and detection techniques are generally complicated, time-consuming, and costly, thus hampering the clinical applications of exosomes. Meanwhile, sandwich-structured immunoassays for exosome isolation and detection rely on the specific binding of membrane surface biomarkers, which may be limited by the type and amount of target protein present. Recently, lipid anchors inserted into the membranes of vesicles through hydrophobic interactions have been adopted as a new strategy for extracellular vesicle manipulation. By combining nonspecific and specific binding, the performance of biosensors can be improved variously. This review presents the reaction mechanisms and properties of lipid anchors/probes, as well as advances in the development of biosensors. The combination of signal amplification methods with lipid anchors is discussed in detail to provide insights into the design of convenient and sensitive detection techniques. Finally, the advantages, challenges, and future directions of lipid anchor-based exosome isolation and detection methods are highlighted from the perspectives of research, clinical use, and commercialization.
Collapse
Affiliation(s)
- Junyuan Zheng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Xiaoxiang Hu
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Yuping Zeng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Binmao Zhang
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Zhonghao Sun
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Xiaowei Liu
- Department of Management, Shenzhen University, Shenzhen, 518055, China.
| | - Weidong Zheng
- Department of Laboratory Medicine, Shenzhen University General Hospital, Shenzhen, 518055, China.
| | - Yujuan Chai
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
46
|
Skouras P, Gargalionis AN, Piperi C. Exosomes as Novel Diagnostic Biomarkers and Therapeutic Tools in Gliomas. Int J Mol Sci 2023; 24:10162. [PMID: 37373314 DOI: 10.3390/ijms241210162] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Exosomes constitute small extracellular vesicles that contain lipids, proteins, nucleic acids, and glycoconjugates from the secreted cells and are capable of transmitting signals between cells and coordinating cellular communication. By this means, they are ultimately involved in physiology and disease, including development, homeostasis, and immune system regulation, as well as contributing to tumor progression and neurodegenerative diseases pathology. Recent studies have shown that gliomas secrete a panel of exosomes which have been associated with cell invasion and migration, tumor immune tolerance, potential for malignant transformation, neovascularization, and resistance to treatment. Exosomes have therefore emerged as intercellular communicators, which mediate the tumor-microenvironment interactions and exosome-regulated glioma cell stemness and angiogenesis. They may induce tumor proliferation and malignancy in normal cells by carrying pro-migratory modulators from cancer cells as well as many different molecular cancer modifiers, such as oncogenic transcripts, miRNAs, mutant oncoproteins, etc., which promote the communication of cancer cells with the surrounding stromal cells and provide valuable information on the molecular profile of the existing tumor. Moreover, engineered exosomes can provide an alternative system for drug delivery and enable efficient treatment. In the present review, we discuss the latest findings regarding the role of exosomes in glioma pathogenesis, their utility in non-invasive diagnosis, and potential applications to treatment.
Collapse
Affiliation(s)
- Panagiotis Skouras
- Department of Neurosurgery, 'Evangelismos' Hospital, Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios N Gargalionis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Biopathology, 'Eginition' Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
47
|
Back SJ, Kim W, Kim DY, Kim SJ, Hwang SR, Jung GB. Rapid and simple isolation and detection of exosomes using CaTiO 3:Eu 3+@Fe 3O 4 multifunctional nanocomposites. Anal Biochem 2023; 673:115161. [PMID: 37201773 DOI: 10.1016/j.ab.2023.115161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/27/2023] [Accepted: 04/18/2023] [Indexed: 05/20/2023]
Abstract
Exosomes are potential biomarkers for disease diagnosis and treatment, as well as drug carriers. However, as their isolation and detection remain critical issues, convenient, rapid, low-cost, and effective methods are necessary. In this study, we present a rapid and simple method for directly capturing and analyzing exosomes from complex cell culture media using CaTiO3:Eu3+@Fe3O4 multifunctional nanocomposites. The CaTiO3:Eu3+@Fe3O4 nanocomposites were prepared by high-energy ball-milling and used to isolate exosomes by binding CaTiO3:Eu3+@Fe3O4 nanocomposites and the hydrophilic phosphate head of the exosome phospholipids. Notably, the developed CaTiO3:Eu3+@Fe3O4 multifunctional nanocomposites achieved results comparable with those of commercially available TiO2 and were separated using a magnet within 10 min. Moreover, we report a surface-enhanced Raman scattering (SERS)-based immunoassay for detecting the exosome biomarker CD81. Gold nanorods (Au NRs) were modified with detection antibodies, and antibody-conjugated Au NRs were labeled with 3, 3, diethylthiatricarbocyanine iodide (DTTC) as the SERS tags. A method combining magnetic separation and SERS was developed to detect exosomal biomarker CD81. The results of this study demonstrate the feasibility of this new technique as a useful tool for exosome isolation and detection.
Collapse
Affiliation(s)
- Sung Jin Back
- Department of Physics Education, Chosun University, Gwangju, 61452, Republic of Korea
| | - Woong Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
| | - Da Young Kim
- Department of Physics Education, Chosun University, Gwangju, 61452, Republic of Korea
| | - Seok-Jun Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Gyeong Bok Jung
- Department of Physics Education, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
48
|
Sun S, Zhang Y, Li Y, Wei L. Crosstalk between colorectal cancer cells and cancer-associated fibroblasts in the tumor microenvironment mediated by exosomal noncoding RNAs. Front Immunol 2023; 14:1161628. [PMID: 37234178 PMCID: PMC10206140 DOI: 10.3389/fimmu.2023.1161628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023] Open
Abstract
Colorectal cancer (CRC) is a common malignant tumor of the digestive system, and its morbidity rates are increasing worldwide. Cancer-associated fibroblasts (CAFs), as part of the tumor microenvironment (TME), are not only closely linked to normal fibroblasts, but also can secrete a variety of substances (including exosomes) to participate in the regulation of the TME. Exosomes can play a key role in intercellular communication by delivering intracellular signaling substances (e.g., proteins, nucleic acids, non-coding RNAs), and an increasing number of studies have shown that non-coding RNAs of exosomal origin from CAFs are not only closely associated with the formation of the CRC microenvironment, but also increase the ability of CRC to grow in metastasis, mediate tumor immunosuppression, and are involved in the mechanism of drug resistance in CRC patients receiving. It is also involved in the mechanism of drug resistance after radiotherapy in CRC patients. In this paper, we review the current status and progress of research on CAFs-derived exosomal non-coding RNAs in CRC.
Collapse
Affiliation(s)
| | | | | | - Linlin Wei
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
49
|
Ding Z, Wei Y, Liu X, Han F, Xu Z. Substantial dimerized G-quadruplex signal units engineered by cutting-mediated exponential rolling circle amplification for ultrasensitive and label-free detection of exosomes. Anal Chim Acta 2023; 1253:341098. [PMID: 36965991 DOI: 10.1016/j.aca.2023.341098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Sensitive and accurate determination of tumor-derived exosomes from complicated biofluids is an important prerequisite for early tumor diagnosis through exosome-based liquid biopsy. Herein, a label-free fluorescence immunoassay protocol for ultrasensitive detection of exosomes was developed by engineering substantial dimerized guanine-quadruplex (Dimer-G4) signal units via in situ cutting-mediated exponential rolling circle amplification (CM-ERCA). First, exosomes were captured and enriched via immunomagnetic separation. Then, molecular recognition was built by the formation of antibody-aptamer sandwich immunocomplex through the specific binding of the designed aptamer-primers with the targeted exosomes. The accuracy of exosome detection was significantly improved by the specific recognition of two typical exosomal protein markers simultaneously. Eventually, in situ CM-ERCA was triggered by a perfect match between the multifunctional circular DNA template and the aptamer-primer on exosomal surface. Amplicons of CM-ERCA loaded with Dimer-G4 were exponentially accumulated during continuous cyclic amplification, dramatically lighting up the thioflavin T (ThT) and generating substantial Dimer-G4 signal units. As a result, ultrasensitive detection of exosomes with the detection limit down to 2.4 × 102 particles/mL was achieved due to the fluorescence enhancement of substantial Dimer-G4 signal units, which is ahead of most of available fluorescence-based methods reported currently. In addition, the intense fluorescence emission and favorable anti-interference of the proposed immunoassay supports identification of exosomes direct in human serums, overcoming the limitations of conventional G4/ThT in serum analysis and revealing its potential for exosome-based liquid biopsy.
Collapse
Affiliation(s)
- Ziling Ding
- Research Center for Analytical Sciences, Northeastern University, 110819, Shenyang, China
| | - Yunyun Wei
- Research Center for Analytical Sciences, Northeastern University, 110819, Shenyang, China
| | - Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, 110819, Shenyang, China
| | - Fei Han
- Research Center for Analytical Sciences, Northeastern University, 110819, Shenyang, China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, 110819, Shenyang, China.
| |
Collapse
|
50
|
Zou X, Huang Z, Guan C, Shi W, Gao J, Wang J, Cui Y, Wang M, Xu Y, Zhong X. Exosomal miRNAs in the microenvironment of pancreatic cancer. Clin Chim Acta 2023; 544:117360. [PMID: 37086943 DOI: 10.1016/j.cca.2023.117360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
Pancreatic cancer (PC) is highly aggressive having an extremely poor prognosis. The tumor microenvironment (TME) of PC is complex and heterogeneous. Various cellular components in the microenvironment are capable of secreting different active substances that are involved in promoting tumor development. Their release may occur via exosomes, the most abundant extracellular vesicles (EVs), that can carry numerous factors as well as act as a mean of intercellular communication. Emerging evidence suggests that miRNAs are involved in the regulation and control of many pathological and physiological processes. They can also be transported by exosomes from donor cells to recipient cells, thereby regulating the TME. Exosomal miRNAs show promise for use as future targets for PC diagnosis and prognosis, which may reveal new treatment strategies for PC. In this paper, we review the important role of exosomal miRNAs in mediating cellular communication in the TME of PC as well as their potential use in clinical applications.
Collapse
Affiliation(s)
- Xinlei Zou
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ziyue Huang
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Canghai Guan
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wujiang Shi
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jianjun Gao
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jiangang Wang
- Central hospital of Baoji, Baoji, Shaanxi 721000, China
| | - Yunfu Cui
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Mei Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
| | - Yi Xu
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China; Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou 310000, China; State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Xiangyu Zhong
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|