1
|
Mehta PK, Soni A, Dahiya B, Sheoran R, Nehra K, Sharma M. Quantification of MPT-64 within pleural fluid extracellular vesicles of tuberculous pleurisy patients by real-time immuno-PCR. Anal Biochem 2025; 702:115829. [PMID: 40058538 DOI: 10.1016/j.ab.2025.115829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Diagnosis of tuberculous (TB) pleurisy is an exigent task owing to atypical clinical presentations and low bacillary content in clinical samples. Hence, there is a crucial need to deliberate a quick and consistent diagnostic test. We recently quantified Mycobacterium tuberculosis (Mtb)-specific MPT-64 (Rv1980c) within pleural fluid extracellular vesicles (pEVs) of TB pleurisy patients by SYBR Green real-time immuno-PCR (RT-I-PCR) assay and compared its diagnostic efficacy with respective ELISA and GeneXpert assay. The size of pEVs of TB pleurisy patients ranged between 47.7 and 170.2 nm as evaluated by Nanoparticle tracking analysis and Transmission electron microscopy. Noticeably, a dynamic range (0.7 pg/mL-9.7 ng/mL) of Mtb MPT-64 was quantitatively detected within pEVs of TB pleurisy individuals by RT-I-PCR, albeit ELISA exhibited a thin range (2.5 ng/mL-11.2 ng/mL). Our RT-I-PCR demonstrated sensitivity of 80 % and 80.9 % in clinically suspected/probable (n = 35) and total (n = 42) TB pleurisy individuals, respectively, with 97.3 % specificity in 38 non-TB controls, against a composite reference standard. Concurrently, MPT-64 detection within pEVs of clinically suspected/probable TB pleurisy cases by ELISA and GeneXpert displayed substantially lower sensitivities (p < 0.05-0.01) than RT-I-PCR. After further improving the sensitivity and authenticating these RT-I-PCR results with a larger sample size, this assay may yield a promising diagnostic kit.
Collapse
Affiliation(s)
- Promod K Mehta
- Department of Life Sciences, Faculty of Allied Health Sciences, Shree Guru Gobind Singh Tricentenary (SGT) University, Gurugram, 122505, India; Centre for Biotechnology, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Aishwarya Soni
- Department of Life Sciences, Faculty of Allied Health Sciences, Shree Guru Gobind Singh Tricentenary (SGT) University, Gurugram, 122505, India; Centre for Biotechnology, Maharshi Dayanand University, Rohtak, 124001, India; Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology (DCRUST), Murthal, Sonipat, 131039, India
| | - Bhawna Dahiya
- Department of Life Sciences, Faculty of Allied Health Sciences, Shree Guru Gobind Singh Tricentenary (SGT) University, Gurugram, 122505, India; Centre for Biotechnology, Maharshi Dayanand University, Rohtak, 124001, India
| | - Reetu Sheoran
- School of Basic Sciences and Research, Sharda University, Greater Noida, 201301, India
| | - Kiran Nehra
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology (DCRUST), Murthal, Sonipat, 131039, India
| | - Mukesh Sharma
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, SGT University, Gurugram, 122505, India
| |
Collapse
|
2
|
Zhou H, Wu W, Zhang Q, Zhang T, Jiang S, Liu H, Ma Y, Chang L, Xie Y, Zhu J, Zhou D, Zhang Y, Xu P. Quantitative Proteome Analysis of Plasma Extracellular Vesicles Identifies Three Proteins with Potential Diagnostic Value for Mycobacterium bovis Infection in Cows. J Proteome Res 2025. [PMID: 40372928 DOI: 10.1021/acs.jproteome.5c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Bovine tuberculosis (bTB) is a zoonotic disease that affects cattle and human health. Although the tuberculin skin test (TST) is the main detection method, there is a need for simpler on-farm tests using fluid samples. This study analyzed plasma extracellular vesicles (EVs) from two cow groups (Cohort A, 15 negative, 22 positive; Cohort B, 28 negative, 40 positive) to explore bTB indicators using proteome profiling. Among the 756 proteins, 217 (Cohort A) and 233 (Cohort B) showed differences between healthy and infected cows, with 47 consistently dysregulated in both groups. These proteins were related to tuberculosis, neutrophil extracellular trap formation, and antigen processing and presentation pathways. Notably, three proteins, HSPA8, B2M, and HRG, were confirmed as bTB indicators using multiple methods, including least absolute shrinkage and selection operator (Lasso) regression selection, western blot (WB), and enzyme-linked immunosorbent assay (ELISA) validation with an independent cohort (Cohort C). This study identifies plasma EV biomarkers for bTB infection, offering insights for bTB detection.
Collapse
Affiliation(s)
- Hangfan Zhou
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Wenhui Wu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Second Clinical Medicine Collage, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qilong Zhang
- Beijing Center for Animal Disease Control and Prevention, Beijing 102629, China
| | - Tao Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Songhao Jiang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hui Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
- Research Unit of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yuan Ma
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Lei Chang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yuping Xie
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jiaqiang Zhu
- Beijing Xinhui Purui Technology Development Co., Ltd, Beijing 102200, China
| | - Degang Zhou
- Beijing Center for Animal Disease Control and Prevention, Beijing 102629, China
| | - Yao Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ping Xu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Second Clinical Medicine Collage, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Research Unit of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
- School of Medicine, Guizhou University, Guiyang 550025, China
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
3
|
Muzanyi G, Bark CM. Severe Tuberculosis. Med Clin North Am 2025; 109:641-650. [PMID: 40185552 DOI: 10.1016/j.mcna.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
The global tuberculosis (TB) pandemic continues despite living in the era of the greatest advances in human medicine. Severe TB in the form of meningitis and miliary TB kills and disables people around the world every day. It disproportionately affects the most vulnerable populations, especially children. Diagnosis and treatment are challenging, and most have poor outcomes, often never receiving treatment at all. Dedicated TB researchers continue to make improvements in the management of severe TB, but the long-term success will require a global commitment to improving health care for poor people around the world.
Collapse
Affiliation(s)
- Grace Muzanyi
- Uganda-Case Western Reserve University Research Collaboration, PO Box 663, Kampala, Uganda
| | - Charles M Bark
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
4
|
Li X, Chen J, Yang Y, Cai H, Ao Z, Xing Y, Li K, Yang K, Guan W, Friend J, Lee LP, Wang N, Guo F. Extracellular vesicle-based point-of-care testing for diagnosis and monitoring of Alzheimer's disease. MICROSYSTEMS & NANOENGINEERING 2025; 11:65. [PMID: 40246821 PMCID: PMC12006457 DOI: 10.1038/s41378-025-00916-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/15/2024] [Accepted: 12/11/2024] [Indexed: 04/19/2025]
Abstract
Extracellular vesicles (EVs) show potential for early diagnosis of Alzheimer's disease (AD) and monitoring of its progression. However, EV-based AD diagnosis faces challenges due to the small size and low abundance of biomarkers. Here, we report a fully integrated organic electrochemical transistor (OECT) sensor for ultrafast, accurate, and convenient point-of-care testing (POCT) of serum EVs from AD patients. By utilizing acoustoelectric enrichment, the EVs can be quickly propelled, significantly enriched, and specifically bound to the OECT detection area, achieving a gain of over 280 times response in 30 s. The integrated POCT sensor can detect serum EVs from AD patients with a limit of detection as low as 500 EV particles/mL and a reduced detection time of just two minutes. Furthermore, the integrated POCT sensors were used to monitor AD progression in an AD mouse model by testing the mouse Aβ EVs at different time courses (up to 18 months) and compared with the Aβ accumulation using high-resolution magnetic resonance imaging (MRI). This innovative technology has the potential for accurate and rapid diagnosis of Alzheimer's and other neurodegenerative diseases, and monitoring of disease progression and treatment response.
Collapse
Affiliation(s)
- Xiang Li
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Jie Chen
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yang Yang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Yantao Xing
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Kangle Li
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Kaiyuan Yang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Weihua Guan
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - James Friend
- Department of Mechanical and Aerospace Engineering, and Department of Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Luke P Lee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Bioengineering, and Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA, 94720, USA.
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Korea.
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea.
| | - Nian Wang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
5
|
Fang C, He X, Tang F, Wang Z, Pan C, Zhang Q, Wu J, Wang Q, Liu D, Zhang Y. Where lung cancer and tuberculosis intersect: recent advances. Front Immunol 2025; 16:1561719. [PMID: 40242762 PMCID: PMC11999974 DOI: 10.3389/fimmu.2025.1561719] [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: 01/16/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Lung cancer (LC) and tuberculosis (TB) represent two major global public health issues. Prior evidence has suggested a link between TB infection and an increased risk of LC. As advancements in LC treatment have led to extended survival rates for LC patients, the co-occurrence of TB and LC has grown more prevalent and poses novel clinical challenges. The intricate molecular mechanisms connecting TB and LC are closely intertwined and many issues remain to be addressed. This review focuses on resemblance between the immunosuppression in tumor and granuloma microenvironments, exploring immunometabolism, cell plasticity, inflammatory signaling pathways, microbiomics, and up-to-date information derived from spatial multi-omics between TB and LC. Furthermore, we outline immunization-related molecular mechanisms underlying these two diseases and propose future research directions. By discussing recent advances and potential targets, this review aims to establish a foundation for developing future therapeutic strategies targeting LC with concurrent TB infection.
Collapse
Affiliation(s)
- Chunju Fang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xuanlu He
- School of Clinical Medicine, Zunyi Medical University, Zunyi, China
| | - Fei Tang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zi Wang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Cong Pan
- School of Biological Sciences, Guizhou Education University, Guiyang, China
- Translational Medicine Research Center, eBond Pharmaceutical Technology Co., Ltd., Chengdu, China
| | - Qi Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jing Wu
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Qinglan Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Daishun Liu
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Yu Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
- National Health Commission Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
6
|
Wang J, Xing K, Zhang G, Li Z, Ding X, Leong DT. Surface Components and Biological Interactions of Extracellular Vesicles. ACS NANO 2025; 19:8433-8461. [PMID: 39999425 DOI: 10.1021/acsnano.4c16854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Extracellular vesicles (EVs) are critical mediators of intercellular communication, carrying bioactive cargo and displaying diverse surface components that reflect their cellular origins and functions. The EV surface, composed of proteins, lipids, and glycocalyx elements, plays a pivotal role in targeting recipient cells, mediating biological interactions, and enabling selective cargo delivery. This review comprehensively examined the molecular architecture of EV surfaces, linking their biogenesis to functional diversity, and highlights their therapeutic and diagnostic potential in diseases such as cancer and cardiovascular disorders. Additionally, we explore emerging applications of EVs, including machine-learning-assisted analysis, chemical integration, and cross-system combinations. The review also discusses some key challenges in the clinical translation of EV-related technologies.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585 Singapore
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kuoran Xing
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585 Singapore
| | - Guoying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Zhiyang Li
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province 210008, China
| | - Xianguang Ding
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585 Singapore
| |
Collapse
|
7
|
Zhang Y, Qin X, Xu Z, Liu W, Lu H, Yang Y, Yang J, Li X, Zhang Y, Yang F. Electric Field-Resistant Bubble-Enhanced Wash-Free Profiling of Extracellular Vesicle Surface Markers. ACS NANO 2025; 19:8093-8107. [PMID: 39985473 DOI: 10.1021/acsnano.4c16353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
Efficient profiling of circulating extracellular vesicles (EVs) benefits noninvasive cancer diagnosis and therapeutic monitoring, but is technically hampered by tedious isolation, multistep washing, and poor sensitivity. Here, we report multifunctional bubbles that enable self-separation, wash-free, single-step, and ultrasensitive profiling of EV surface markers in plasma samples for early diagnosis and treatment monitoring of lung cancer. In this assay, the buoyancy-dominated bubble is electric field-resistant, allowing EV-responsive release of electroactive probes for electrohydrodynamic nanoshearing force-enhanced hybridization, self-separation from the electrode interface for minimizing noise in electrochemical measurements, and one-step wash-free EV profiling. This assay achieves sensitivity near a single-EV level, shows high specificity against nontarget EVs, and tracks EV phenotypic changes induced by drugs. We further show that this technology can classify plasma samples (n = 111) between cancer patients and noncancer controls with accuracies >95%, enable accurate early diagnosis via machine learning, and monitor pre/post-surgery efficacy with higher accuracy over routine clinical serum markers. This bubble-driven one-step EV assay provides a promising wash-free quantitative tool to enable clinical precision liquid biopsies.
Collapse
Affiliation(s)
- Yuyuan Zhang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Xiaojie Qin
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Zhanyu Xu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Weifeng Liu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Yu Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jie Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fan Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
8
|
Li M, Shen C, Lv M, Luo Y. Nanobiosensors Enable High-Efficiency Detection of Tuberculosis Nucleic Acid. JACS AU 2025; 5:536-549. [PMID: 40017783 PMCID: PMC11862950 DOI: 10.1021/jacsau.4c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb), with a complex pathogenesis that poses a long-term threat to human health globally. Early and accurate diagnosis of TB provides a critical window for timely and effective treatment. The development of nucleic acid testing (NAT) based on polymerase chain reaction (PCR) has greatly improved the diagnostic efficiency of TB. However, balancing detection accuracy, efficiency, and cost in TB NAT remains challenging. Functionalized nanomaterials-based nanobiosensors have demonstrated exceptional performance in detecting TB nucleic acid by integrating their unique physicochemical properties with diverse biological probes that exploit Mtb characteristics to effectively amplify biological signals. Compared to traditional NAT, nanobiosensors simplify nucleic acid detection, improve accuracy, and reduce reliance on external conditions, thereby contributing to more immediate and accurate TB diagnosis. In this perspective, we provide a comprehensive summary and discussion on current strategies for detecting Mtb biomarkers using nucleic acid along with novel solutions for TB diagnosis. Additionally, we explore the advantages and challenges associated with applying nanotechnology to the clinical management of TB, particularly point-of-care testing (POCT).
Collapse
Affiliation(s)
- Mei Li
- Department
of Laboratory Medicine, Sichuan Medical Laboratory Clinical Medicine
Research Center, West China Hospital, Sichuan
University, Chengdu 610041, China
| | - Chen Shen
- Department
of Laboratory Medicine, Sichuan Medical Laboratory Clinical Medicine
Research Center, West China Hospital, Sichuan
University, Chengdu 610041, China
| | - Min Lv
- College
of Chemistry and Materials Science, Shanghai
Normal University, Shanghai 200000, China
| | - Yao Luo
- Department
of Laboratory Medicine, Sichuan Medical Laboratory Clinical Medicine
Research Center, West China Hospital, Sichuan
University, Chengdu 610041, China
- Department
of Pulmonary and Critical Care Medicine, Zigong First People’s Hospital, Zigong 643000, China
| |
Collapse
|
9
|
Wang Y, Xiong J, Ouyang K, Ling M, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Extracellular vesicles: From large-scale production and engineering to clinical applications. J Tissue Eng 2025; 16:20417314251319474. [PMID: 40322740 PMCID: PMC12048759 DOI: 10.1177/20417314251319474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs) have emerged as a promising strategy for treating a wide spectrum of pathologies, as they can deliver their cargo to recipient cells and regulate the signaling pathway of these cells to modulate their fate. Despite the great potential of EVs in clinical applications, their low yield and the challenges of cargo loading remain significant obstacles, hindering their transition from experimental research to clinical practice. Therefore, promoting EV release and enhancing EV cargo-loading are promising fields with substantial research potential and broad application prospects. In this review, we summarize the clinical applications of EVs, the methods and technologies for their large-scale production, engineering, and modification, as well as the challenges that must be addressed during their development. We also discuss the future perspectives of this exciting field of research to facilitate its transformation from bench to clinical reality.
Collapse
Affiliation(s)
- Yuxuan Wang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiali Xiong
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Kun Ouyang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Mingwang Ling
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Junyi Luo
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiajie Sun
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qianyun Xi
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ting Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yongliang Zhang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 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
|
11
|
Rao L, Yuan Y, Shen X, Yu G, Chen X. Designing nanotheranostics with machine learning. NATURE NANOTECHNOLOGY 2024; 19:1769-1781. [PMID: 39362960 DOI: 10.1038/s41565-024-01753-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/08/2024] [Indexed: 10/05/2024]
Abstract
The inherent limits of traditional diagnoses and therapies have driven the development and application of emerging nanotechnologies for more effective and safer management of diseases, herein referred to as 'nanotheranostics'. Although many important technological successes have been achieved in this field, widespread adoption of nanotheranostics as a new paradigm is hindered by specific obstacles, including time-consuming synthesis of nanoparticles, incomplete understanding of nano-bio interactions, and challenges regarding chemistry, manufacturing and the controls required for clinical translation and commercialization. As a key branch of artificial intelligence, machine learning (ML) provides a set of tools capable of performing time-consuming and result-perception tasks, thus offering unique opportunities for nanotheranostics. This Review summarizes the progress and challenges in this emerging field of ML-aided nanotheranostics, and discusses the opportunities in developing next-generation nanotheranostics with reliable datasets and advanced ML models to offer better clinical benefits to patients.
Collapse
Affiliation(s)
- Lang Rao
- Shenzhen Bay Laboratory, Shenzhen, China.
| | - Yuan Yuan
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Computer Science, Boston College, Chestnut Hill, MA, USA
| | - Xi Shen
- Tencent AI Lab, Shenzhen, China
- Intellindust, Shenzhen, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
12
|
Salgado-Cantú MG, Gutiérrez-González LH, Guzmán-Beltrán S, Herrera MT, Sarabia C, González Y. Impact of Mycobacterium tuberculosis H37Rv Infection on Extracellular Vesicle Cargo in Macrophages: Implications for Host-Pathogen Interaction. Microorganisms 2024; 12:2405. [PMID: 39770608 PMCID: PMC11678565 DOI: 10.3390/microorganisms12122405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) is one of the most common respiratory infections worldwide, and it is caused by Mycobacterium tuberculosis (Mtb). Mtb employs immune evasion mechanisms that allow the disease to become chronic. Despite extensive research, the host-pathogen interaction remains incompletely understood. Extracellular vesicles (EVs) are small membrane particles that play a regulatory role in infectious diseases. Host-derived EVs have been identified as carriers of proteins, messenger RNA, and lipids from both the host cells and the pathogens. In this study, we assessed the cargo of EVs in human macrophages infected with the virulent strain H37Rv of Mtb at 1 and 24 h post-infection (hpi). The results showed that 1 hpi, infected macrophages secreted EVs containing Mtb proteins (15 to 37 kDa) and Ag85 kDa, as well as RNA transcripts (ESAT-6, 5KST, Ag85, IS6110, 30 kDa, 19 kDa, and MPT64). However, these decreased at 24 hpi. The infection of macrophages with Mtb was observed to result in the release of EVs containing Ag85 protein and RNA transcripts of Mtb; this process appeared to diminish after 24 hpi, suggesting the existence of an evasion mechanism. Both Ag85 and the RNA transcripts could be potential biomarkers for the diagnosis of TB patients.
Collapse
Affiliation(s)
- Manuel G. Salgado-Cantú
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (M.G.S.-C.); (S.G.-B.); (M.T.H.); (C.S.)
| | - Luis Horacio Gutiérrez-González
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Silvia Guzmán-Beltrán
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (M.G.S.-C.); (S.G.-B.); (M.T.H.); (C.S.)
| | - María Teresa Herrera
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (M.G.S.-C.); (S.G.-B.); (M.T.H.); (C.S.)
| | - Carmen Sarabia
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (M.G.S.-C.); (S.G.-B.); (M.T.H.); (C.S.)
| | - Yolanda González
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (M.G.S.-C.); (S.G.-B.); (M.T.H.); (C.S.)
| |
Collapse
|
13
|
Yao X, He D, Wei P, Niu Z, Chen H, Li L, Fu P, Wang Y, Lou S, Qian S, Zheng J, Zuo G, Wang K. DNA Nanomaterial-Empowered Surface Engineering of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306852. [PMID: 38041689 DOI: 10.1002/adma.202306852] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/30/2023] [Indexed: 12/03/2023]
Abstract
Extracellular vesicles (EVs) are cell-secreted biological nanoparticles that are critical mediators of intercellular communication. They contain diverse bioactive components, which are promising diagnostic biomarkers and therapeutic agents. Their nanosized membrane-bound structures and innate ability to transport functional cargo across major biological barriers make them promising candidates as drug delivery vehicles. However, the complex biology and heterogeneity of EVs pose significant challenges for their controlled and actionable applications in diagnostics and therapeutics. Recently, DNA molecules with high biocompatibility emerge as excellent functional blocks for surface engineering of EVs. The robust Watson-Crick base pairing of DNA molecules and the resulting programmable DNA nanomaterials provide the EV surface with precise structural customization and adjustable physical and chemical properties, creating unprecedented opportunities for EV biomedical applications. This review focuses on the recent advances in the utilization of programmable DNA to engineer EV surfaces. The biology, function, and biomedical applications of EVs are summarized and the state-of-the-art achievements in EV isolation, analysis, and delivery based on DNA nanomaterials are introduced. Finally, the challenges and new frontiers in EV engineering are discussed.
Collapse
Affiliation(s)
- Xuxiang Yao
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Dongdong He
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Pengyao Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Zitong Niu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Hao Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lin Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Yiting Wang
- College of Chemistry, Jilin Normal University, Siping, 136000, P. R. China
| | - Saiyun Lou
- Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
- Ningbo Second Hospital, Ningbo, 315010, P. R. China
| | - Sihua Qian
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Jianping Zheng
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Guokun Zuo
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| |
Collapse
|
14
|
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: 0] [Impact Index Per Article: 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
|
15
|
Ya N, Zhang D, Wang Y, Zheng Y, Yang M, Wu H, Oudeng G. Recent advances of biocompatible optical nanobiosensors in liquid biopsy: towards early non-invasive diagnosis. NANOSCALE 2024; 16:13784-13801. [PMID: 38979555 DOI: 10.1039/d4nr01719f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Liquid biopsy is a non-invasive diagnostic method that can reduce the risk of complications and offers exceptional benefits in the dynamic monitoring and acquisition of heterogeneous cell population information. Optical nanomaterials with excellent light absorption, luminescence, and photoelectrochemical properties have accelerated the development of liquid biopsy technologies. Owing to the unique size effect of optical nanomaterials, their improved optical properties enable them to exhibit good sensitivity and specificity for mitigating signal interference from various molecules in body fluids. Nanomaterials with biocompatible and optical sensing properties play a crucial role in advancing the maturity and diversification of liquid biopsy technologies. This article offers a comprehensive review of recent advanced liquid biopsy technologies that utilize novel biocompatible optical nanomaterials, including fluorescence, colorimetric, photoelectrochemical, and Raman broad-spectrum-based biosensors. We focused on liquid biopsy for the most significant early biomarkers in clinical medicine, and specifically reviewed reports on the effectiveness of optical nanosensing technology in the detection of real patient samples, which may provide basic evidence for the transition of optical nanosensing technology from engineering design to clinical practice. Furthermore, we introduced the integration of optical nanosensing-based liquid biopsy with modern devices, such as smartphones, to demonstrate the potential of the technology in portable clinical diagnosis.
Collapse
Affiliation(s)
- Na Ya
- Pediatric Research Institute, Shenzhen Children's Hospital, Shenzhen, Guangdong, P.R. China
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P.R. China
| | - Dangui Zhang
- Pediatric Research Institute, Shenzhen Children's Hospital, Shenzhen, Guangdong, P.R. China
- Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Yan Wang
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P.R. China
| | - Yi Zheng
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P.R. China
| | - Mo Yang
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P.R. China
| | - Hao Wu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
| | - Gerile Oudeng
- Pediatric Research Institute, Shenzhen Children's Hospital, Shenzhen, Guangdong, P.R. China
| |
Collapse
|
16
|
Mahmoudi S, García MJ, Drain PK. Current approaches for diagnosis of subclinical pulmonary tuberculosis, clinical implications and future perspectives: a scoping review. Expert Rev Clin Immunol 2024; 20:715-726. [PMID: 38879875 DOI: 10.1080/1744666x.2024.2326032] [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: 11/12/2023] [Accepted: 02/28/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Subclinical tuberculosis (TB) is the presence of TB disease among people who are either asymptomatic or have minimal symptoms. AREAS COVERED Currently, there are no accurate diagnostic tools and clear treatment approaches for subclinical TB. In this study, a comprehensive literature search was conducted across major databases. This review aimed to uncover the latest advancements in diagnostic approaches, explore their clinical implications, and outline potential future perspectives. While innovative technologies are in development to enable sputum-free TB tests, there remains a critical need for precise diagnostic tools tailored to the unique characteristics of subclinical TB. Given the complexity of subclinical TB, a multidisciplinary approach involving clinicians, microbiologists, epidemiologists, and public health experts is essential. Further research is needed to establish standardized diagnostic criteria and treatment guidelines specifically tailored for subclinical TB, acknowledging the unique challenges posed by this elusive stage of the disease. EXPERT OPINION Efforts are needed for the detection, diagnosis, and treatment of subclinical TB. In this review, we describe the importance of subclinical TB, both from a clinical and public health perspective and highlight the diagnostic and treatment gaps of this stage.
Collapse
Affiliation(s)
- Shima Mahmoudi
- Biotechnology Centre, Silesian University of Technology, Gliwice, Poland
| | - Maria J García
- Department of Preventive Medicine and Public Health and Microbiology, Autonoma University of Madrid, Madrid, Spain
| | - Paul K Drain
- International Clinical Research Center, Department of Global Health, Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Dasgupta S. Thinking Beyond Disease Silos: Dysregulated Genes Common in Tuberculosis and Lung Cancer as Identified by Systems Biology and Machine Learning. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:347-356. [PMID: 38856681 DOI: 10.1089/omi.2024.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The traditional way of thinking about human diseases across clinical and narrow phenomics silos often masks the underlying shared molecular substrates across human diseases. One Health and planetary health fields particularly address such complexities and invite us to think across the conventional disease nosologies. For example, tuberculosis (TB) and lung cancer (LC) are major pulmonary diseases with significant planetary health implications. Despite distinct etiologies, they can coexist in a given community or patient. This is both a challenge and an opportunity for preventive medicine, diagnostics, and therapeutics innovation. This study reports a bioinformatics analysis of publicly available gene expression data, identifying overlapping dysregulated genes, downstream regulators, and pathways in TB and LC. Analysis of NCBI-GEO datasets (GSE83456 and GSE103888) unveiled differential expression of CEACAM6, MUC1, ADM, DYSF, PLOD2, and GAS6 genes in both diseases, with pathway analysis indicating association with lysine degradation pathway. Random forest, a machine-learning-based classification, achieved accuracies of 84% for distinguishing TB from controls and 83% for discriminating LC from controls using these specific genes. Additionally, potential drug targets were identified, with molecular docking confirming the binding affinity of warfarin to GAS6. Taken together, the present study speaks of the pressing need to rethink clinical diagnostic categories of human diseases and that TB and LC might potentially share molecular substrates. Going forward, planetary health and One Health scholarship are poised to cultivate new ways of thinking about diseases not only across medicine and ecology but also across traditional diagnostic conventions.
Collapse
|
18
|
Li Q, Zhan S, Yang X, Zhang Z, Sun N, Wang X, Kang J, Du R, Hong X, Yue M, Li X, Tang Y, Liu G, Liu Y, Liu D. Choline Phosphate-Grafted Nanozymes as Universal Extracellular Vesicle Probes for Bladder Cancer Detection. ACS NANO 2024; 18:16113-16125. [PMID: 38857428 DOI: 10.1021/acsnano.4c00280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Urinary extracellular vesicles (uEVs) are regarded as highly promising liquid-biopsy biomarkers for the early diagnosis and prognosis of bladder cancer (BC). However, detection of uEVs remains technically challenging owing to their huge heterogeneity and ultralow abundance in real samples. We herein present a choline phosphate-grafted platinum nanozyme (Pt@CP) that acts as a universal EV probe for the construction of a high-throughput and high-sensitivity immunoassay, which allowed multiplex profiling of uEV protein markers for BC detection. With the Pt@CP-based immunoassays, three uEV protein markers (MUC-1, CCDC25, and GLUT1) were identified for BC, by which the BC cases (n = 48), cystitis patients (n = 27), and healthy donors (n = 24) were discriminated with high clinical sensitivity and specificity (area under curve = 98.3%). For the BC cases (n = 9) after surgery, the Pt@CP-based immunoassay could report the postoperative residual tumor that cannot be observed by cystoscopy, which is clinically significant for assessing BC recurrence. This work provides generally high sensitivity for EV detection, facilitating the discovery and clinical use of EV-based biomarkers.
Collapse
Affiliation(s)
- Qiang Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Saisong Zhan
- Key Laboratory of Inorganic-Organic Hybrid Functional Materials Chemistry, Ministry of Education, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, China
| | - Xiaoqing Yang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhaowei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Ning Sun
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jingjing Kang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Rui Du
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoqin Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Minghao Yue
- Department of Urology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xiaomin Li
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yujing Tang
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Guangming Liu
- Department of Urology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Yue Liu
- Key Laboratory of Inorganic-Organic Hybrid Functional Materials Chemistry, Ministry of Education, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
19
|
Qin X, Xiang Y, Mao L, Yang Y, Wei B, Lu H, Li X, Zhang Y, Yang F. Buoyant Metal-Organic Framework Corona-Driven Fast Isolation and Ultrasensitive Profiling of Circulating Extracellular Vesicles. ACS NANO 2024; 18:14569-14582. [PMID: 38781132 DOI: 10.1021/acsnano.4c02339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Accurately assaying tumor-derived circulating extracellular vesicles (EVs) is fundamental in noninvasive cancer diagnosis and therapeutic monitoring but limited by challenges in efficient EV isolation and profiling. Here, we report a bioinspired buoyancy-driven metal-organic framework (MOF) corona that leverages on-bubble coordination and dual-encoded surface-enhanced Raman scattering (SERS) nanotags to streamline rapid isolation and ultrasensitive profiling of plasma EVs in a single assay for cancer diagnostics. This integrated bubble-MOF-SERS EV assay (IBMsv) allows barnacle-like high-density adhesion of MOFs on a self-floating bubble surface to enable fast isolation (2 min, near 90% capture efficiency) of tumor EVs via enhanced EV-MOF binding. Also, IBMsv harnesses four-plexed SERS nanotags to profile the captured EV surface protein markers at a single-particle level. Such a sensitive assay allows multiplexed profiling of EVs across five cancer types, revealing heterogeneous EV surface expression patterns. Furthermore, the IBMsv assay enables cancer diagnosis in a pilot clinical cohort (n = 55) with accuracies >95%, improves discrimination between cancer and noncancer patients via an algorithm, and monitors the surgical treatment response from hepatocellular carcinoma patients. This assay provides a fast, sensitive, streamlined, multiplexed, and portable blood test tool to enable cancer diagnosis and response monitoring in clinical settings.
Collapse
Affiliation(s)
- Xiaojie Qin
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Yuanhang Xiang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Linfeng Mao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Yu Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Binqi Wei
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fan Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
20
|
Salgueiro VC, Passemar C, Vázquez-Iniesta L, Lerma L, Floto A, Prados-Rosales R. Extracellular vesicles in mycobacteria: new findings in biogenesis, host-pathogen interactions, and diagnostics. mBio 2024; 15:e0255223. [PMID: 38567992 PMCID: PMC11077946 DOI: 10.1128/mbio.02552-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024] Open
Abstract
Since the discovery of extracellular vesicles (EVs) in mycobacterial species 15 years back, we have learned that this phenomenon is conserved in the Mycobacterium genus and has critical roles in bacterial physiology and host-pathogen interactions. Mycobacterium tuberculosis (Mtb), the tuberculosis (TB) causative agent, produces EVs both in vitro and in vivo including a diverse set of biomolecules with demonstrated immunomodulatory effects. Moreover, Mtb EVs (MEVs) have been shown to possess vaccine properties and carry biomarkers with diagnostic capacity. Although information on MEV biogenesis relative to other bacterial species is scarce, recent studies have shed light on how MEVs originate and are released to the extracellular space. In this minireview, we discuss past and new information about the vesiculogenesis phenomenon in Mtb, including biogenesis, MEV cargo, aspects in the context of host-pathogen interactions, and applications that could help to develop effective tools to tackle the disease.
Collapse
Affiliation(s)
- Vivian C. Salgueiro
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Charlotte Passemar
- Cambridge Center for Lung Infection, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | - Lucía Vázquez-Iniesta
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Lerma
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Andrés Floto
- Cambridge Center for Lung Infection, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | - Rafael Prados-Rosales
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
21
|
Das S, Lyon CJ, Hu T. A Panorama of Extracellular Vesicle Applications: From Biomarker Detection to Therapeutics. ACS NANO 2024; 18:9784-9797. [PMID: 38471757 PMCID: PMC11008359 DOI: 10.1021/acsnano.4c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Extracellular vesicles (EVs) secreted by all cell types are involved in the cell-to-cell transfer of regulatory factors that influence cell and tissue phenotypes in normal and diseased tissues. EVs are thus a rich source of biomarker targets for assays that analyze blood and urinary EVs for disease diagnosis. Sensitive biomarker detection in EVs derived from specific cell populations is a key major hurdle when analyzing complex biological samples, but innovative approaches surveyed in this Perspective can streamline EV isolation and enhance the sensitivity of EV detection procedures required for clinical application of EV-based diagnostics and therapeutics, including nanotechnology and microfluidics, to achieve EV characterizations. Finally, this Perspective also outlines opportunities and challenges remaining for clinical translation of EV-based assays.
Collapse
Affiliation(s)
- Sumita Das
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
22
|
Dai C, Xiong H, He R, Zhu C, Li P, Guo M, Gou J, Mei M, Kong D, Li Q, Wee ATS, Fang X, Kong J, Liu Y, Wei D. Electro-Optical Multiclassification Platform for Minimizing Occasional Inaccuracy in Point-of-Care Biomarker Detection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312540. [PMID: 38288781 DOI: 10.1002/adma.202312540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/13/2024] [Indexed: 02/06/2024]
Abstract
On-site diagnostic tests that accurately identify disease biomarkers lay the foundation for self-healthcare applications. However, these tests routinely rely on single-mode signals and suffer from insufficient accuracy, especially for multiplexed point-of-care tests (POCTs) within a few minutes. Here, this work develops a dual-mode multiclassification diagnostic platform that integrates an electrochemiluminescence sensor and a field-effect transistor sensor in a microfluidic chip. The microfluidic channel guides the testing samples to flow across electro-optical sensor units, which produce dual-mode readouts by detecting infectious biomarkers of tuberculosis (TB), human rhinovirus (HRV), and group B streptococcus (GBS). Then, machine-learning classifiers generate three-dimensional (3D) hyperplanes to diagnose different diseases. Dual-mode readouts derived from distinct mechanisms enhance the anti-interference ability physically, and machine-learning-aided diagnosis in high-dimensional space reduces the occasional inaccuracy mathematically. Clinical validation studies with 501 unprocessed samples indicate that the platform has an accuracy approaching 99%, higher than the 77%-93% accuracy of rapid point-of-care testing technologies at 100% statistical power (>150 clinical tests). Moreover, the diagnosis time is 5 min without a trade-off of accuracy. This work solves the occasional inaccuracy issue of rapid on-site diagnosis, endowing POCT systems with the same accuracy as laboratory tests and holding unique prospects for complicated scenes of personalized healthcare.
Collapse
Affiliation(s)
- Changhao Dai
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai, 200433, China
| | - Huiwen Xiong
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Rui He
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou, 73000, China
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Chenxin Zhu
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Pintao Li
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Mingquan Guo
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jian Gou
- Department of Physics, National University of Singapore, Singapore, 117542, Singapore
| | - Miaomiao Mei
- Yizheng Hospital of Traditional Chinese Medicine, Yangzhou, 211400, China
| | - Derong Kong
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai, 200433, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Andrew Thye Shen Wee
- Department of Physics, National University of Singapore, Singapore, 117542, Singapore
| | - Xueen Fang
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Jilie Kong
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Yunqi Liu
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai, 200433, China
| | - Dacheng Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai, 200433, China
| |
Collapse
|
23
|
Savage N. Tracking down tuberculosis. Nature 2024:10.1038/d41586-024-00087-8. [PMID: 38273057 DOI: 10.1038/d41586-024-00087-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
|
24
|
Ghoshal A, Verma A, Bhaskar A, Dwivedi VP. The uncharted territory of host-pathogen interaction in tuberculosis. Front Immunol 2024; 15:1339467. [PMID: 38312835 PMCID: PMC10834760 DOI: 10.3389/fimmu.2024.1339467] [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: 11/17/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
Mycobacterium tuberculosis (M.tb) effectively manipulates the host processes to establish the deadly respiratory disease, Tuberculosis (TB). M.tb has developed key mechanisms to disrupt the host cell health to combat immune responses and replicate efficaciously. M.tb antigens such as ESAT-6, 19kDa lipoprotein, Hip1, and Hsp70 destroy the integrity of cell organelles (Mitochondria, Endoplasmic Reticulum, Nucleus, Phagosomes) or delay innate/adaptive cell responses. This is followed by the induction of cellular stress responses in the host. Such cells can either undergo various cell death processes such as apoptosis or necrosis, or mount effective immune responses to clear the invading pathogen. Further, to combat the infection progression, the host secretes extracellular vesicles such as exosomes to initiate immune signaling. The exosomes can contain M.tb as well as host cell-derived peptides that can act as a double-edged sword in the immune signaling event. The host-symbiont microbiota produces various metabolites that are beneficial for maintaining healthy tissue microenvironment. In juxtaposition to the above-mentioned mechanisms, M.tb dysregulates the gut and respiratory microbiome to support its replication and dissemination process. The above-mentioned interconnected host cellular processes of Immunometabolism, Cellular stress, Host Microbiome, and Extracellular vesicles are less explored in the realm of exploration of novel Host-directed therapies for TB. Therefore, this review highlights the intertwined host cellular processes to control M.tb survival and showcases the important factors that can be targeted for designing efficacious therapy.
Collapse
Affiliation(s)
| | | | | | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
25
|
Li L, Lyon CJ, LaCourse SM, Zheng W, Stern J, Escudero JN, Murithi WB, Njagi L, John-Stewart G, Hawn TR, Nduba V, Abdelgaliel W, Tombler T, Horne D, Jiang L, Hu TY. Sensitive Blood-Based Detection of HIV-1 and Mycobacterium tuberculosis Peptides for Disease Diagnosis by Immuno-Affinity Liquid Chromatography-Tandem Mass Spectrometry: A Method Development and Proof-of-Concept Study. Clin Chem 2023; 69:1409-1419. [PMID: 37956323 PMCID: PMC10965313 DOI: 10.1093/clinchem/hvad173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/09/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Novel approaches that allow early diagnosis and treatment monitoring of both human immunodeficiency virus-1 (HIV-1) and tuberculosis disease (TB) are essential to improve patient outcomes. METHODS We developed and validated an immuno-affinity liquid chromatography-tandem mass spectrometry (ILM) assay that simultaneously quantifies single peptides derived from HIV-1 p24 and Mycobacterium tuberculosis (Mtb) 10-kDa culture filtrate protein (CFP10) in trypsin-digested serum derived from cryopreserved serum archives of cohorts of adults and children with/without HIV and TB. RESULTS ILM p24 and CFP10 results demonstrated good intra-laboratory precision and accuracy, with recovery values of 96.7% to 104.6% and 88.2% to 111.0%, total within-laboratory precision (CV) values of 5.68% to 13.25% and 10.36% to 14.92%, and good linearity (r2 > 0.99) from 1.0 to 256.0 pmol/L and 0.016 to 16.000 pmol/L, respectively. In cohorts of adults (n = 34) and children (n = 17) with HIV and/or TB, ILM detected p24 and CFP10 demonstrated 85.7% to 88.9% and 88.9% to 100.0% diagnostic sensitivity for HIV-1 and TB, with 100% specificity for both, and detected HIV-1 infection earlier than 3 commercial p24 antigen/antibody immunoassays. Finally, p24 and CFP10 values measured in longitudinal serum samples from children with HIV-1 and TB distinguished individuals who responded to TB treatment from those who failed to respond or were untreated, and who developed TB immune reconstitution inflammatory syndrome. CONCLUSIONS Simultaneous ILM evaluation of p24 and CFP10 results may allow for early TB and HIV detection and provide valuable information on treatment response to facilitate integration of TB and HIV diagnosis and management.
Collapse
Affiliation(s)
- Lin Li
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Sylvia M. LaCourse
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Wenshu Zheng
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Joshua Stern
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Jaclyn N. Escudero
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Wilfred Bundi Murithi
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Lilian Njagi
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Grace John-Stewart
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Thomas R. Hawn
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Videlis Nduba
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | | | - David Horne
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Li Jiang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
| | - Tony Y. Hu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
26
|
Morrow E, Liu Q, Kiguli S, Swarbrick G, Nsereko M, Null MD, Cansler M, Mayanja-Kizza H, Boom WH, Chheng P, Nyendak MR, Lewinsohn DM, Lewinsohn DA, Lancioni CL. Production of Proinflammatory Cytokines by CD4+ and CD8+ T Cells in Response to Mycobacterial Antigens among Children and Adults with Tuberculosis. Pathogens 2023; 12:1353. [PMID: 38003817 PMCID: PMC10675744 DOI: 10.3390/pathogens12111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/24/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a leading cause of pediatric morbidity and mortality. Young children are at high risk of TB following Mtb exposure, and this vulnerability is secondary to insufficient host immunity during early life. Our primary objective was to compare CD4+ and CD8+ T-cell production of proinflammatory cytokines IFN-gamma, IL-2, and TNF-alpha in response to six mycobacterial antigens and superantigen staphylococcal enterotoxin B (SEB) between Ugandan adults with confirmed TB (n = 41) and young Ugandan children with confirmed (n = 12) and unconfirmed TB (n = 41), as well as non-TB lower respiratory tract infection (n = 39). Flow cytometry was utilized to identify and quantify CD4+ and CD8+ T-cell cytokine production in response to each mycobacterial antigen and SEB. We found that the frequency of CD4+ and CD8+ T-cell production of cytokines in response to SEB was reduced in all pediatric cohorts when compared to adults. However, T-cell responses to Mtb-specific antigens ESAT6 and CFP10 were equivalent between children and adults with confirmed TB. In contrast, cytokine production in response to ESAT6 and CFP10 was limited in children with unconfirmed TB and absent in children with non-TB lower respiratory tract infection. Of the five additional mycobacterial antigens tested, PE3 and PPE15 were broadly recognized regardless of TB disease classification and age. Children with confirmed TB exhibited robust proinflammatory CD4+ and CD8+ T-cell responses to Mtb-specific antigens prior to the initiation of TB treatment. Our findings suggest that adaptive proinflammatory immune responses to Mtb, characterized by T-cell production of IFN-gamma, IL-2, and TNF-alpha, are not impaired during early life.
Collapse
Affiliation(s)
- Erin Morrow
- School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Qijia Liu
- School of Public Health, Oregon Health and Science University, Portland, OR 97239, USA
| | - Sarah Kiguli
- Department of Pediatrics, Makerere University, Mulago Hill Road, Kampala P.O. Box 7072, Uganda
| | - Gwendolyn Swarbrick
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mary Nsereko
- Uganda-Case Western Research Collaboration, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Megan D. Null
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Meghan Cansler
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Harriet Mayanja-Kizza
- Uganda-Case Western Research Collaboration, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Medicine, Makerere University, Mulago Hill Road, Kampala P.O. Box 7072, Uganda
| | - W. Henry Boom
- Uganda-Case Western Research Collaboration, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Phalkun Chheng
- Uganda-Case Western Research Collaboration, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Melissa R. Nyendak
- Department of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - David M. Lewinsohn
- Department of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Pulmonary and Critical Care Medicine, Portland VA Medical Center, Portland, OR 97239, USA
| | - Deborah A. Lewinsohn
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Christina L. Lancioni
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
27
|
Lei H, Li Q, Pei Z, Liu L, Yang N, Cheng L. Nonferrous Ferroptosis Inducer Manganese Molybdate Nanoparticles to Enhance Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303438. [PMID: 37420331 DOI: 10.1002/smll.202303438] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/27/2023] [Indexed: 07/09/2023]
Abstract
Tumor immunotherapy is an important tool in oncology treatment. However, only a small percentage of patients have an effective immune response to tumor immunotherapy due to the poor infiltration of pro-inflammatory immune cells in immune "cold" tumors and an immunosuppressive network in the tumor microenvironment (TME). Ferroptosis has been widely used as a novel strategy to enhance tumor immunotherapy. Herein, manganese molybdate nanoparticles (MnMoOx NPs) depleted the highly expressed glutathione (GSH) in tumors and inhibited glutathione peroxidase 4 (GPX4) expression, thus triggering ferroptosis, inducing immune cell death (ICD), further releasing damage-associated molecular patterns (DAMPs), and enhancing tumor immunotherapy. Furthermore, MnMoOx NPs can efficiently suppress tumors, promote the maturation of dendritic cells (DCs), infiltrate T cells, and reverse the immunosuppressive microenvironment, making the tumor an immune "hot" tumor. Combination with an immune checkpoint inhibitor (ICI) (α-PD-L1) further enhanced the anti-tumor effect and inhibited metastases as well. The work provides a new idea for the development of nonferrous inducers of ferroptosis to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Quguang Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Lin Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Suzhou University, Suzhou, 215000, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| |
Collapse
|
28
|
Yi L, Hou B, Liu X. Optical Integration in Wearable, Implantable and Swallowable Healthcare Devices. ACS NANO 2023; 17:19491-19501. [PMID: 37807286 DOI: 10.1021/acsnano.3c04284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Recent advances in materials and semiconductor technologies have led to extensive research on optical integration in wearable, implantable, and swallowable health devices. These optical systems utilize the properties of light─intensity, wavelength, polarization, and phase─to monitor and potentially intervene in various biological events. The potential of these devices is greatly enhanced through the use of multifunctional optical materials, adaptable integration processes, advanced optical sensing principles, and optimized artificial intelligence algorithms. This synergy creates many possibilities for clinical applications. This Perspective discusses key opportunities, challenges, and future directions, particularly with respect to sensing modalities, multifunctionality, and the integration of miniaturized optoelectronic devices. We present fundamental insights and illustrative examples of such devices in wearable, implantable, and swallowable forms. The constant pursuit of innovation and the dedicated approach to critical challenges are poised to influence diverse fields.
Collapse
Affiliation(s)
- Luying Yi
- Department of Chemistry, National University of Singapore, 117543, Singapore
| | - Bo Hou
- Department of Chemistry, National University of Singapore, 117543, Singapore
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, 117543, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
- Center for Functional Materials, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| |
Collapse
|
29
|
Zhang J, Wu J, Wang G, He L, Zheng Z, Wu M, Zhang Y. Extracellular Vesicles: Techniques and Biomedical Applications Related to Single Vesicle Analysis. ACS NANO 2023; 17:17668-17698. [PMID: 37695614 DOI: 10.1021/acsnano.3c03172] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Extracellular vesicles (EVs) are extensively dispersed lipid bilayer membrane vesicles involved in the delivery and transportation of molecular payloads to certain cell types to facilitate intercellular interactions. Their significant roles in physiological and pathological processes make EVs outstanding biomarkers for disease diagnosis and treatment monitoring as well as ideal candidates for drug delivery. Nevertheless, differences in the biogenesis processes among EV subpopulations have led to a diversity of biophysical characteristics and molecular cargos. Additionally, the prevalent heterogeneity of EVs has been found to substantially hamper the sensitivity and accuracy of disease diagnosis and therapeutic monitoring, thus impeding the advancement of clinical applications. In recent years, the evolution of single EV (SEV) analysis has enabled an in-depth comprehension of the physical properties, molecular composition, and biological roles of EVs at the individual vesicle level. This review examines the sample acquisition tactics prior to SEV analysis, i.e., EV isolation techniques, and outlines the current state-of-the-art label-free and label-based technologies for SEV identification. Furthermore, the challenges and prospects of biomedical applications based on SEV analysis are systematically discussed.
Collapse
Affiliation(s)
- Jie Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Jiacheng Wu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Guanzhao Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Luxuan He
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Ziwei Zheng
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Minhao Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P. R. China
| | - Yuanqing Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
30
|
Chen Y, Zhang S, Lu J, Li D, Wu H, Zhang L, Li X, Gao X, Xu Y, Zeng Z, Zeng L, Ding X, Li X, Ding S. DNA-Guided Extracellular-Vesicle Metallization with High Catalytic Activity for Accurate Diagnosis of Pulmonary Nodules. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208142. [PMID: 37066711 DOI: 10.1002/smll.202208142] [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: 12/25/2022] [Revised: 03/19/2023] [Indexed: 06/19/2023]
Abstract
Sensitive and specific analysis of extracellular vesicles (EVs) offers a promising minimally invasive way to identify malignant pulmonary nodules from benign lesions. However, accurate analysis of EVs is subject to free target proteins in blood samples, which compromises the clinical diagnosis value of EVs. Here a DNA-guided extracellular-vesicle metallization (DEVM) strategy is described for ultrasensitive and specific analysis of EV protein biomarkers and classification of pulmonary nodules. The facile DEVM process mainly includes the incorporation of DNA labeled with cholesterol and thiol groups into EV membranes and subsequent deposition of Au3+ and Pt4+ to allow the DNA-functionalized EVs to be encapsulated with AuPt nanoshells. It is found that the synthesized AuPt-metallized EVs possess extrinsic peroxidase-like activity. Utilizing the feature of the catalytic metal nanoshells just growth on the EV membranes, the DEVM method enables multiparametric recognition of target proteins and EV membranes and can produce an amplified colorimetric signal, avoiding the interference of free proteins. By profiling four surface proteins of EVs from 48 patients with pulmonary nodules, the highest area under the receiver operating characteristic curve (0.9983) is obtained. Therefore, this work provides a feasible EVs analysis tool for accurate pulmonary nodules management.
Collapse
Affiliation(s)
- Yirong Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Songzhi Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jun Lu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Dandan Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haiping Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lu Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xin Gao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuan Xu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zijie Zeng
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Li Zeng
- Department of Laboratory Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, Chongqing, 400016, China
| | - Xiaojuan Ding
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinmin Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
31
|
Tang J, Jia X, Li Q, Cui Z, Liang A, Ke B, Yang D, Yao C. A DNA-based hydrogel for exosome separation and biomedical applications. Proc Natl Acad Sci U S A 2023; 120:e2303822120. [PMID: 37399419 PMCID: PMC10334772 DOI: 10.1073/pnas.2303822120] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023] Open
Abstract
Exosomes (EXOs) have been proven as biomarkers for disease diagnosis and agents for therapeutics. Great challenge remains in the separation of EXOs with high-purity and low-damage from complex biological media, which is critical for the downstream applications. Herein, we report a DNA-based hydrogel to realize the specific and nondestructive separation of EXOs from complex biological media. The separated EXOs were directly utilized in the detection of human breast cancer in clinical samples, as well as applied in the therapeutics of myocardial infarction in rat models. The materials chemistry basis of this strategy involved the synthesis of ultralong DNA chains via an enzymatic amplification, and the formation of DNA hydrogels through complementary base-pairing. These ultralong DNA chains that contained polyvalent aptamers were able to recognize and bind with the receptors on EXOs, and the specific and efficient binding ensured the selective separation of EXOs from media into the further formed networked DNA hydrogel. Based on this DNA hydrogel, rationally designed optical modules were introduced for the detection of exosomal pathogenic microRNA, which achieved the classification of breast cancer patients versus healthy donors with 100% precision. Furthermore, the DNA hydrogel that contained mesenchymal stem cell-derived EXOs was proved with significant therapeutic efficacy in repairing infarcted myocardium of rat models. We envision that this DNA hydrogel-based bioseparation system is promising as a powerful biotechnology, which will promote the development of extracellular vesicles in nanobiomedicine.
Collapse
Affiliation(s)
- Jianpu Tang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Xuemei Jia
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Qian Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Zhen Cui
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Aiqi Liang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Bin Ke
- Tianjin Medical University Cancer Institute and Hospital, Tianjin300060, P. R. China
| | - Dayong Yang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (Ministry of Education), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin300350, P. R. China
| |
Collapse
|
32
|
Zhang P, Zhou X, Wang S. Plasmonic Scattering Microscopy for Label-Free Imaging of Molecular Binding Kinetics: From Single Molecules to Single Cells. CHEMISTRY METHODS : NEW APPROACHES TO SOLVING PROBLEMS IN CHEMISTRY 2023; 3:e202200066. [PMID: 37448471 PMCID: PMC10344632 DOI: 10.1002/cmtd.202200066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Indexed: 07/15/2023]
Abstract
Measuring molecular binding kinetics represents one of the most important tasks in molecular interaction analysis. Surface plasmon resonance (SPR) is a popular tool for analyzing molecular binding. Plasmonic scattering microscopy (PSM) is a newly developed SPR imaging technology, which detects the out-of-plane scattering of surface plasmons by analytes and has pushed the detection limit of label-free SPR imaging down to a single-protein level. In addition, PSM also allows SPR imaging with high spatiotemporal resolution, making it possible to analyze cellular response to the molecular bindings. In this Mini Review, we present PSM as a method of choice for chemical and biological imaging, introduce its theoretical mechanism, present its experimental schemes, summarize its exciting applications, and discuss its challenges as well as the promising future.
Collapse
Affiliation(s)
- Pengfei Zhang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, 85287 (USA)
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190 (P. R. China)
| | - Xinyu Zhou
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, 85287 (USA)
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287 (USA)
| | - Shaopeng Wang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, 85287 (USA)
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287 (USA)
| |
Collapse
|
33
|
Shami-Shah A, Norman M, Walt DR. Ultrasensitive protein detection technologies for extracellular vesicle measurements. Mol Cell Proteomics 2023; 22:100557. [PMID: 37088150 DOI: 10.1016/j.mcpro.2023.100557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023] Open
Abstract
Extracellular Vesicles (EVs) are nanoscopic, heterogenous, lipid-rich particles that carry a multitude of cargo biomolecules including proteins, nucleic acids, and metabolites. Although historically, EVs were regarded as cellular debris with no intrinsic value, growing understanding of EV biogenesis has led to the realization that EVs facilitate intercellular communication and are sources of liquid biomarkers. EVs can be isolated and analyzed from a wide variety of accessible biofluids for biomarker discovery and diagnostic applications. There is a diversity of EVs from different biological compartments (e.g., cells, tissues) and some of these EVs are present at extremely low concentrations. Consequently, a challenge in the field is to find appropriate markers that enable selective isolation of these rare EVs. Many conventional protein detection technologies have limited sensitivity to detect low abundance biomarkers in EVs, limiting their use in EV research. Advances in ultrasensitive detection technologies are needed to harness the potential of EVs for clinical application. This Perspective highlights current EV research focusing on ultrasensitive detection technologies, their limitations, and areas of potential growth in the future.
Collapse
Affiliation(s)
- Adnan Shami-Shah
- Department of Pathology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Maia Norman
- Department of Pathology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA; Tufts University School of Medicine, Boston, MA
| | - David R Walt
- Department of Pathology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA.
| |
Collapse
|
34
|
Valdemar-Aguilar CM, Manisekaran R, Acosta-Torres LS, López-Marín LM. Spotlight on mycobacterial lipid exploitation using nanotechnology for diagnosis, vaccines, and treatments. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102653. [PMID: 36646193 PMCID: PMC9839462 DOI: 10.1016/j.nano.2023.102653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/24/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Tuberculosis (TB), historically the most significant cause of human morbidity and mortality, has returned as the top infectious disease worldwide, under circumstances worsened by the COVID-19 pandemic's devastating effects on public health. Although Mycobacterium tuberculosis, the causal agent, has been known of for more than a century, the development of tools to control it has been largely neglected. With the advancement of nanotechnology, the possibility of engineering tools at the nanoscale creates unique opportunities to exploit any molecular type. However, little attention has been paid to one of the major attributes of the pathogen, represented by the atypical coat and its abundant lipids. In this review, an overview of the lipids encountered in M. tuberculosis and interest in exploiting them for the development of TB control tools are presented. Then, the amalgamation of nanotechnology with mycobacterial lipids from both reported and future works are discussed.
Collapse
Affiliation(s)
- Carlos M. Valdemar-Aguilar
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230 Querétaro, Mexico,Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - Ravichandran Manisekaran
- Interdisciplinary Research Laboratory (LII), Nanostructures and Biomaterials Area, Escuela Nacional de Estudios Superiores Unidad León, Universidad Nacional Autónoma de México, Predio el Saucillo y el Potrero, Comunidad de los Tepetates, 37689 León, Mexico.
| | - Laura S. Acosta-Torres
- Interdisciplinary Research Laboratory (LII), Nanostructures and Biomaterials Area, Escuela Nacional de Estudios Superiores Unidad León, Universidad Nacional Autónoma de México, Predio el Saucillo y el Potrero, Comunidad de los Tepetates, 37689 León, Mexico
| | - Luz M. López-Marín
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230 Querétaro, Mexico,Corresponding authors
| |
Collapse
|
35
|
Wang S, Zheng W, Wang R, Zhang L, Yang L, Wang T, Saliba JG, Chandra S, Li CZ, Lyon CJ, Hu TY. Monocrystalline Labeling Enables Stable Plasmonic Enhancement for Isolation-Free Extracellular Vesicle Analysis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204298. [PMID: 36354195 PMCID: PMC9839537 DOI: 10.1002/smll.202204298] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/14/2022] [Indexed: 05/20/2023]
Abstract
Sensitive detection of extracellular vesicles (EVs) as emerging biomarkers has shown great promises for disease diagnosis. Plasmonic metal nanostructures conjugated with molecules that bind specific biomarker targets are widely used for EVs sensing but involve tradeoffs between particle-size-dependent signal intensity and conjugation efficiency. One solution to this problem would be to induce nucleation on nanoparticles that have successfully bound a target biomarker to permit in situ nanoparticle growth for signal amplification, but approaches that are evaluated to date require harsh conditions or lack nucleation specificity, prohibiting their effective use with most biological specimens. This study describes a one-step in situ strategy to induce monocrystalline copper shell growth on gold nanorod probes without decreasing signal by disrupting probe-target interactions or lipid bilayer integrity to enable EV biomarker detections. This approach increases the detected nanoparticle signal about two orders of magnitude after a 10 min copper nanoshell growth reaction. This has significant implications for improved disease detection, as indicated by the ability of a novel immunoassay using this approach to detect low abundance EVs carrying a pathogen-derived biomarker, after their direct capture from serum, to facilitate the diagnosis of tuberculosis cases in a diagnostically challenging pediatric cohort.
Collapse
Affiliation(s)
- Shu Wang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Wenshu Zheng
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Ruixuan Wang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Lili Zhang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Li Yang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Tao Wang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Julian G Saliba
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, 6823 St. Charles Ave, New Orleans, LA, 70118, USA
| | - Sutapa Chandra
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Chen-Zhong Li
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Christopher J Lyon
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Tony Y Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| |
Collapse
|
36
|
Liu Y, Li B, Liu B, Zhang K. Single-Particle Optical Imaging for Ultrasensitive Bioanalysis. BIOSENSORS 2022; 12:1105. [PMID: 36551072 PMCID: PMC9775667 DOI: 10.3390/bios12121105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
The quantitative detection of critical biomolecules and in particular low-abundance biomarkers in biofluids is crucial for early-stage diagnosis and management but remains a challenge largely owing to the insufficient sensitivity of existing ensemble-sensing methods. The single-particle imaging technique has emerged as an important tool to analyze ultralow-abundance biomolecules by engineering and exploiting the distinct physical and chemical property of individual luminescent particles. In this review, we focus and survey the latest advances in single-particle optical imaging (OSPI) for ultrasensitive bioanalysis pertaining to basic biological studies and clinical applications. We first introduce state-of-the-art OSPI techniques, including fluorescence, surface-enhanced Raman scattering, electrochemiluminescence, and dark-field scattering, with emphasis on the contributions of various metal and nonmetal nano-labels to the improvement of the signal-to-noise ratio. During the discussion of individual techniques, we also highlight their applications in spatial-temporal measurement of key biomarkers such as proteins, nucleic acids and extracellular vesicles with single-entity sensitivity. To that end, we discuss the current challenges and prospective trends of single-particle optical-imaging-based bioanalysis.
Collapse
Affiliation(s)
- Yujie Liu
- Shanghai Institute of Pediatric Research, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Binxiao Li
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Kun Zhang
- Shanghai Institute of Pediatric Research, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|