1
|
Woo HK, Nam Y, Park HG, Lee H. Bridging laboratory innovation to translational research and commercialization of extracellular vesicle isolation and detection. Biosens Bioelectron 2025; 282:117475. [PMID: 40300344 PMCID: PMC12076185 DOI: 10.1016/j.bios.2025.117475] [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: 08/30/2024] [Revised: 03/04/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
Extracellular vesicles (EVs) have emerged as promising biomarkers for various diseases. Encapsulating biomolecules reflective of their parental cells, EVs are readily accessible in bodily fluids. The prospect for minimally invasive, repeatable molecular testing has stimulated significant research; however, challenges persist in isolating EVs from complex biological matrices and characterizing their limited molecular cargo. Technical advances have been pursued to address these challenges, producing innovative EV-specific platforms. This review highlights recent technological developments, focusing on EV isolation and molecular detection methodologies. Furthermore, it explores the translation of these laboratory innovations to clinical applications through the analysis of patient samples, providing insights into the potential diagnostic and prognostic utility of EV-based technologies.
Collapse
Affiliation(s)
- Hyun-Kyung Woo
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yoonho Nam
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Chemical and Biomolecular Engineering (BK21 Four), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyun Gyu Park
- Department of Chemical and Biomolecular Engineering (BK21 Four), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Zhang G, Huang X, Liu S, Xu Y, Wang N, Yang C, Zhu Z. Demystifying EV heterogeneity: emerging microfluidic technologies for isolation and multiplexed profiling of extracellular vesicles. LAB ON A CHIP 2025; 25:1228-1255. [PMID: 39775292 DOI: 10.1039/d4lc00777h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous lipid containers carrying complex molecular cargoes, including proteins, nucleic acids, glycans, etc. These vesicles are closely associated with specific physiological characteristics, which makes them invaluable in the detection and monitoring of various diseases. However, traditional isolation methods are often labour-intensive, inefficient, and time-consuming. In addition, single biomarker analyses are no longer accurate enough to meet diagnostic needs. Routine isolation and molecular analysis of high-purity EVs in clinical applications is even more challenging. In this review, we discuss a promising solution, microfluidic-based techniques, that combine efficient isolation and multiplex detection of EVs, to further demystify EV heterogeneity. These microfluidic-based EV multiplexing platforms will hopefully facilitate development of liquid biopsies and offer promising opportunities for personalised therapy.
Collapse
Affiliation(s)
- Guihua Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Xiaodan Huang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Sinong Liu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Yiling Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Nan Wang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai 200127, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
3
|
Lan Z, Chen R, Zou D, Zhao C. Microfluidic Nanoparticle Separation for Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411278. [PMID: 39632600 PMCID: PMC11775552 DOI: 10.1002/advs.202411278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/11/2024] [Indexed: 12/07/2024]
Abstract
A deeper understanding of disease heterogeneity highlights the urgent need for precision medicine. Microfluidics, with its unique advantages, such as high adjustability, diverse material selection, low cost, high processing efficiency, and minimal sample requirements, presents an ideal platform for precision medicine applications. As nanoparticles, both of biological origin and for therapeutic purposes, become increasingly important in precision medicine, microfluidic nanoparticle separation proves particularly advantageous for handling valuable samples in personalized medicine. This technology not only enhances detection, diagnosis, monitoring, and treatment accuracy, but also reduces invasiveness in medical procedures. This review summarizes the fundamentals of microfluidic nanoparticle separation techniques for precision medicine, starting with an examination of nanoparticle properties essential for separation and the core principles that guide various microfluidic methods. It then explores passive, active, and hybrid separation techniques, detailing their principles, structures, and applications. Furthermore, the review highlights their contributions to advancements in liquid biopsy and nanomedicine. Finally, it addresses existing challenges and envisions future development spurred by emerging technologies such as advanced materials science, 3D printing, and artificial intelligence. These interdisciplinary collaborations are anticipated to propel the platformization of microfluidic separation techniques, significantly expanding their potential in precision medicine.
Collapse
Affiliation(s)
- Zhenwei Lan
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Rui Chen
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Da Zou
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Chun‐Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| |
Collapse
|
4
|
Seo J, Ha S, Kim SJ. Investigation of Operational Parameters for Nanoelectrokinetic Purification and Preconcentration. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:16443-16453. [PMID: 39048092 DOI: 10.1021/acs.langmuir.4c01773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
This work reports on experimental investigations into the operational parameters of nanoelectrokinetic purification and preconcentration, especially utilizing on ion concentration polarization (ICP). ICP as a nanoscale electrokinetic phenomenon has demonstrated promising advances in various fields utilizing an ion depletion zone (IDZ) with a steep electric field gradient inside the ICP layer. However, the inevitable electrokinetic instability occurring within the IDZ has posed a challenge in operating the ICP system stably. To address the need for a stable and efficient ICP operation in various devices and applications, we propose an operational strategy along with conducted research to determine optimal operating ranges. In order to investigate the operational parameters, a unit voltage (VTH) is introduced as the threshold for initiating ICP. We examined the applicability of VTH across various operating ranges to ensure its effectiveness and versatility. In ICP purification, we categorize three modes (steady, burst, and unsteady) based on IDZ expansion and stability under varying VTH and flow rate conditions, presenting optimal operational conditions that minimize the voltage margin. In ICP preconcentration, a systematic investigation is conducted to observe the influence of background electrolyte concentration and voltage conditions on preconcentration efficiency, offering insights into the correlation between preconcentration factor, electrical conditions, and preconcentration time. Therefore, this research would contribute to the practical understanding of nanoelectrokinetics, providing insight into experimental designs. These findings are expected to offer valuable guidance to researchers aiming to utilize ICP's potential across a spectrum of applications, from purification to preconcentration, in the realm of micro/nanofluidic systems.
Collapse
Affiliation(s)
- Joowon Seo
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungjae Ha
- ProvaLabs, Inc., Seoul 08826, Republic of Korea
| | - Sung Jae Kim
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
- SOFT Foundry Institute, Seoul National University, Seoul 08826, Republic of Korea
- Inter-University Semiconductor Research Center, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
5
|
Mehraji S, DeVoe DL. Microfluidic synthesis of lipid-based nanoparticles for drug delivery: recent advances and opportunities. LAB ON A CHIP 2024; 24:1154-1174. [PMID: 38165786 DOI: 10.1039/d3lc00821e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Microfluidic technologies are revolutionizing the synthesis of nanoscale lipid particles and enabling new opportunities for the production of lipid-based nanomedicines. By harnessing the benefits of microfluidics for controlling diffusive and advective transport within microfabricated flow cells, microfluidic platforms enable unique capabilities for lipid nanoparticle synthesis with precise and tunable control over nanoparticle properties. Here we present an assessment of the current state of microfluidic technologies for lipid-based nanoparticle and nanomedicine production. Microfluidic techniques are discussed in the context of conventional production methods, with an emphasis on the capabilities of microfluidic systems for controlling nanoparticle size and size distribution. Challenges and opportunities associated with the scaling of manufacturing throughput are discussed, together with an overview of emerging microfluidic methods for lipid nanomedicine post-processing. The impact of additive manufacturing on current and future microfluidic platforms is also considered.
Collapse
Affiliation(s)
- Sima Mehraji
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA.
- Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
| | - Don L DeVoe
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA.
- Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
6
|
Bu Y, Wang J, Ni S, Lu Z, Guo Y, Yobas L. High-Performance Gel-Free and Label-Free Size Fractionation of Extracellular Vesicles with Two-Dimensional Electrophoresis in a Microfluidic Artificial Sieve. Anal Chem 2024; 96:3508-3516. [PMID: 38364051 DOI: 10.1021/acs.analchem.3c05290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived particles that exhibit diverse sizes, molecular contents, and clinical implications for various diseases depending on their specific subpopulations. However, fractionation of EV subpopulations with high resolution, efficiency, purity, and yield remains an elusive goal due to their diminutive sizes. In this study, we introduce a novel strategy that effectively separates EV subpopulations in a gel-free and label-free manner, using two-dimensional (2D) electrophoresis in a microfluidic artificial sieve. The microfabricated artificial sieve consists of periodically arranged micro-slit-well structures in a 2D array and generates an anisotropic electric field pattern to size fractionate EVs into discrete streams and steer the subpopulations into designated outlets for collection within a minute. Along with fractionating EV subpopulations, contaminants such as free proteins and short nucleic acids can be simultaneously directed to waste outlets, thus accomplishing both size fractionation and purification of EVs with high performance. Our platform offers a simple, rapid, and versatile solution for EV subpopulation isolation, which can potentially facilitate the discovery of biomarkers for specific EV subtypes and the development of EV-based therapeutics.
Collapse
Affiliation(s)
- Yang Bu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Jinhui Wang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Sheng Ni
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Zechen Lu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Yusong Guo
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Levent Yobas
- Department of Electronic and Computer Engineering, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| |
Collapse
|
7
|
Meng Y, Zhang Y, Bühler M, Wang S, Asghari M, Stürchler A, Mateescu B, Weiss T, Stavrakis S, deMello AJ. Direct isolation of small extracellular vesicles from human blood using viscoelastic microfluidics. SCIENCE ADVANCES 2023; 9:eadi5296. [PMID: 37801500 PMCID: PMC10558121 DOI: 10.1126/sciadv.adi5296] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/05/2023] [Indexed: 10/08/2023]
Abstract
Small extracellular vesicles (sEVs; <200 nm) that contain lipids, nucleic acids, and proteins are considered promising biomarkers for a wide variety of diseases. Conventional methods for sEV isolation from blood are incompatible with routine clinical workflows, significantly hampering the utilization of blood-derived sEVs in clinical settings. Here, we present a simple, viscoelastic-based microfluidic platform for label-free isolation of sEVs from human blood. The separation performance of the device is assessed by isolating fluorescent sEVs from whole blood, demonstrating purities and recovery rates of over 97 and 87%, respectively. Significantly, our viscoelastic-based microfluidic method also provides for a remarkable increase in sEV yield compared to gold-standard ultracentrifugation, with proteomic profiles of blood-derived sEVs purified by both methods showing similar protein compositions. To demonstrate the clinical utility of the approach, we isolate sEVs from blood samples of 20 patients with cancer and 20 healthy donors, demonstrating that elevated sEV concentrations can be observed in blood derived from patients with cancer.
Collapse
Affiliation(s)
- Yingchao Meng
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Yanan Zhang
- Department of Neurology, University Hospital Zürich, 8091 Zürich, Switzerland
- Clinical Neuroscience Center, University of Zürich, 8091 Zürich, Switzerland
| | - Marcel Bühler
- Department of Neurology, University Hospital Zürich, 8091 Zürich, Switzerland
- Clinical Neuroscience Center, University of Zürich, 8091 Zürich, Switzerland
| | - Shuchen Wang
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Mohammad Asghari
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Alessandra Stürchler
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
- Brain Research Institute, University of Zürich, 8057 Zürich, Switzerland
| | - Bogdan Mateescu
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
- Brain Research Institute, University of Zürich, 8057 Zürich, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital Zürich, 8091 Zürich, Switzerland
- Clinical Neuroscience Center, University of Zürich, 8091 Zürich, Switzerland
| | - Stavros Stavrakis
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Andrew J. deMello
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
8
|
Seo J, Jung S, Park J, Kim HY, Kim SJ. Hierarchical Capillarity-Assisted Liquid Invasion in Multilayered Paper Channels for Nanoelectrokinetic Preconcentration. NANO LETTERS 2023; 23:8065-8072. [PMID: 37581872 DOI: 10.1021/acs.nanolett.3c02044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
A nanoelectrokinetic phenomenon called ion concentration polarization (ICP) has been recently applied to microfluidic paper-based devices for the high fold preconcentration of low-abundant analytes. The inherent microstructural characteristics of cellulose papers can sufficiently stabilize the chaotic electroconvection of ICP, which is a significant annoyance for typical engineered microfluidic channels. However, a high electrical voltage to induce ICP in a paper-fluidic channel can increase unavoidable electrophoretic forces over drag forces so that the preconcentrated plug is rapidly receded with severe dispersion. In order to enhance the hydraulic drag force that helps the preconcentration of analytes, here we introduce a multilayered paper structure into paper-fluidic channel. We theoretically and experimentally demonstrate that a hierarchical capillary structure in a multilayered paper-fluidic channel can effectively increase the hydraulic drag force. For the practical utility in the field of diagnostics, the mechanism is verified by a simple example of the immunoassay using biotin-streptavidin complexation.
Collapse
Affiliation(s)
- Joowon Seo
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sohyun Jung
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jihee Park
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho-Young Kim
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
- SOFT Foundry Institute, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Jae Kim
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
- SOFT Foundry Institute, Seoul National University, Seoul 08826, Republic of Korea
- Inter-university Semiconductor Research Center, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
9
|
Paccosi E, Proietti-De-Santis L. Parkinson's Disease: From Genetics and Epigenetics to Treatment, a miRNA-Based Strategy. Int J Mol Sci 2023; 24:ijms24119547. [PMID: 37298496 DOI: 10.3390/ijms24119547] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, characterized by an initial and progressive loss of dopaminergic neurons of the substantia nigra pars compacta via a potentially substantial contribution from protein aggregates, the Lewy bodies, mainly composed of α-Synuclein among other factors. Distinguishing symptoms of PD are bradykinesia, muscular rigidity, unstable posture and gait, hypokinetic movement disorder and resting tremor. Currently, there is no cure for PD, and palliative treatments, such as Levodopa administration, are directed to relieve the motor symptoms but induce severe side effects over time. Therefore, there is an urgency for discovering new drugs in order to design more effective therapeutic approaches. The evidence of epigenetic alterations, such as the dysregulation of different miRNAs that may stimulate many aspects of PD pathogenesis, opened a new scenario in the research for a successful treatment. Along this line, a promising strategy for PD treatment comes from the potential exploitation of modified exosomes, which can be loaded with bioactive molecules, such as therapeutic compounds and RNAs, and can allow their delivery to the appropriate location in the brain, overcoming the blood-brain barrier. In this regard, the transfer of miRNAs within Mesenchymal stem cell (MSC)-derived exosomes has yet to demonstrate successful results both in vitro and in vivo. This review, besides providing a systematic overview of both the genetic and epigenetic basis of the disease, aims to explore the exosomes/miRNAs network and its clinical potential for PD treatment.
Collapse
Affiliation(s)
- Elena Paccosi
- Unit of Molecular Genetics of Aging, Department of Ecology and Biology (DEB), University of Tuscia, 01100 Viterbo, Italy
| | - Luca Proietti-De-Santis
- Unit of Molecular Genetics of Aging, Department of Ecology and Biology (DEB), University of Tuscia, 01100 Viterbo, Italy
| |
Collapse
|
10
|
Bu Y, Wang J, Ni S, Guo Y, Yobas L. Continuous-flow label-free size fractionation of extracellular vesicles through electrothermal fluid rolls and dielectrophoresis synergistically integrated in a microfluidic device. LAB ON A CHIP 2023; 23:2421-2433. [PMID: 36951129 DOI: 10.1039/d2lc01193j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived bioparticles that play significant roles in various biological processes including cell-to-cell communication and intercellular delivery. Additionally, they hold great potential as liquid biopsy biomarkers for pre-diagnostic applications. However, the isolation of EV subpopulations, especially exosomes from a biological fluid remains a challenge due to their submicron range. Here, we demonstrate continuous-flow label-free size fractionation of EVs for the first time through a synergistic combination of electrothermal fluid rolls and dielectrophoresis in a microfluidic device. The device features three dimensional microelectrodes with unique sidewall contours that give rise to effective electrothermal fluid rolls in cooperation with dielectrophoretic forces for the electrokinetic manipulation and size separation of submicron particles. We first validate the device functionality by separating submicron polystyrene particles from binary mixtures with a cut-off size of ∼200 nm and then isolate intact exosomes from cell culture medium or blood serum with a high recovery rate and purity (∼80%). The device operation in a high-conductivity medium renders the method ideal for the purification of target bioparticles directly from physiological fluids, and may offer a robust and versatile platform for EV related diagnostic applications.
Collapse
Affiliation(s)
- Yang Bu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
| | - Jinhui Wang
- Division of Life Sciences, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Sheng Ni
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
| | - Yusong Guo
- Division of Life Sciences, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Levent Yobas
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, SAR, China
| |
Collapse
|
11
|
Zheng F, Wang J, Wang D, Yang Q. Clinical Application of Small Extracellular Vesicles in Gynecologic Malignancy Treatments. Cancers (Basel) 2023; 15:cancers15071984. [PMID: 37046644 PMCID: PMC10093031 DOI: 10.3390/cancers15071984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are the key mediators of intercellular communication. They have the potential for clinical use as diagnostic or therapeutic biomarkers and have been explored as vectors for drug delivery. Identification of reliable and noninvasive biomarkers, such as sEVs, is important for early diagnosis and precise treatment of gynecologic diseases to improve patient prognosis. Previous reviews have summarized routine sEVs isolation and identification methods; however, novel and unconventional methods have not been comprehensively described. This review summarizes a convenient method of isolating sEVs from body fluids and liquid biopsy-related sEV markers for early, minimally invasive diagnosis of gynecologic diseases. In addition, the characteristics of sEVs as drug carriers and in precision treatment and drug resistance are introduced, providing a strong foundation for identifying novel and potential therapeutic targets for sEV therapy. We propose potential directions for further research on the applications of sEVs in the diagnosis and treatment of gynecologic diseases.
Collapse
|
12
|
Zhou Y, Niu J, Zhou Y, Li F. Liquid Plasticine-Based Electrokinetic Enrichment of Proteins. ChemistryOpen 2023; 12:e202200259. [PMID: 36971105 PMCID: PMC10041546 DOI: 10.1002/open.202200259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Protein analysis is an important approach for disease diagnosis, in which sample pretreatment is an essential step since protein samples are often complex and many protein biomarkers are of low abundance. Here, given the good openness and light transmission of liquid plasticine (LP), which is a liquid entity formed by SiO2 nanoparticles and encapsulated aqueous solution, we developed a LP-based field-amplified sample stacking (FASS) system for protein enrichment. The system was composed of a LP container, a sample solution and a Tris-HCl solution containing hydroxyethyl cellulose (HEC). The system design, mechanism investigation, optimization of experimental parameters and characterization of LP-FASS performance for protein enrichment were well studied. Under the optimized experimental conditions of 1 % HEC, 100 mm Tris-HCl and 100 V in the LP-FASS system, a 40-80 times enrichment of proteins was obtained in 40 min using bovine hemoglobin (BHb) as the model protein using the constructed LP-FASS system. The simultaneous enrichment of multiple proteins (phycocyanin, BHb and cytochrome C) was also realized using the system. The LP-FASS system can serve as a new platform for protein enrichment which is easy to be combined with online and offline detections.
Collapse
Affiliation(s)
- Yulin Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, 710049, Xi'an, P. R. China
| | - Jicheng Niu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, 710049, Xi'an, P. R. China
| | - Yan Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, 710049, Xi'an, P. R. China
| | - Fei Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, 710049, Xi'an, P. R. China
| |
Collapse
|
13
|
Hettiarachchi S, Cha H, Ouyang L, Mudugamuwa A, An H, Kijanka G, Kashaninejad N, Nguyen NT, Zhang J. Recent microfluidic advances in submicron to nanoparticle manipulation and separation. LAB ON A CHIP 2023; 23:982-1010. [PMID: 36367456 DOI: 10.1039/d2lc00793b] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Manipulation and separation of submicron and nanoparticles are indispensable in many chemical, biological, medical, and environmental applications. Conventional technologies such as ultracentrifugation, ultrafiltration, size exclusion chromatography, precipitation and immunoaffinity capture are limited by high cost, low resolution, low purity or the risk of damage to biological particles. Microfluidics can accurately control fluid flow in channels with dimensions of tens of micrometres. Rapid microfluidics advancement has enabled precise sorting and isolating of nanoparticles with better resolution and efficiency than conventional technologies. This paper comprehensively studies the latest progress in microfluidic technology for submicron and nanoparticle manipulation. We first summarise the principles of the traditional techniques for manipulating nanoparticles. Following the classification of microfluidic techniques as active, passive, and hybrid approaches, we elaborate on the physics, device design, working mechanism and applications of each technique. We also compare the merits and demerits of different microfluidic techniques and benchmark them with conventional technologies. Concurrently, we summarise seven standard post-separation detection techniques for nanoparticles. Finally, we discuss current challenges and future perspectives on microfluidic technology for nanoparticle manipulation and separation.
Collapse
Affiliation(s)
- Samith Hettiarachchi
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Haotian Cha
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Lingxi Ouyang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | | | - Hongjie An
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Gregor Kijanka
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Navid Kashaninejad
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
14
|
Alternative biological sources for extracellular vesicles production and purification strategies for process scale-up. Biotechnol Adv 2023; 63:108092. [PMID: 36608746 DOI: 10.1016/j.biotechadv.2022.108092] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) are phospholipidic bi-layer enclosed nanoparticles secreted naturally by all cell types. They are attracting increasing attention in the fields of nanomedicine, nutraceutics and cosmetics as biocompatible carriers for drug delivery, with intrinsic properties beneficial to human health. Scientific work now focuses on developing techniques for isolating EVs that can translate into industrial-scale production and meet rigorous clinical requirements. The science of EVs is ongoing, and many pitfalls must be addressed, such as the requirement for standard, reproducible, inexpensive, and Good Manufacturing Practices (GMP) adherent EV processing techniques. Researchers are exploring the use of alternative sources to EVs derived from mammalian cultures, such as plant EVs, as well as the use of bacteria, algae and milk. Regarding the downstream processing of EVs, many alternative techniques to the ultracentrifugation (UC) protocols most commonly used in the laboratory are emerging. In the context of process scale-up, membrane-based processes for isolation and purification of EVs are the most promising, either as stand-alone processes or in combination with chromatographic techniques. This review discusses current trends on EVs source selection and EVs downstream processing techniques, with a focus on plant-derived EVs and membrane-based techniques for EVs enrichment.
Collapse
|
15
|
Bondarenko M, Yaroshchuk A. Computational Design of an Electro-Membrane Microfluidic-Diode System. MEMBRANES 2023; 13:243. [PMID: 36837746 PMCID: PMC9959715 DOI: 10.3390/membranes13020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
This study uses computational design to explore the performance of a novel electro-membrane microfluidic diode consisting of physically conjugated nanoporous and micro-perforated ion-exchange layers. Previously, such structures have been demonstrated to exhibit asymmetric electroosmosis, but the model was unrealistic in several important respects. This numerical study investigates two quantitative measures of performance (linear velocity of net flow and efficiency) as functions of such principal system parameters as perforation size and spacing, the thickness of the nanoporous layer and the zeta potential of the pore surface. All of these dependencies exhibit pronounced maxima, which is of interest for future practical applications. The calculated linear velocities of net flows are in the range of several tens of liters per square meter per hour at realistically applied voltages. The system performance somewhat declines when the perforation size is increased from 2 µm to 128 µm (with a parallel increase of the inter-perforation spacing) but remains quite decent even for the largest perforation size. Such perforations should be relatively easy to generate using inexpensive equipment.
Collapse
Affiliation(s)
- Mykola Bondarenko
- F.D. Ovcharenko Institute of Bio-Colloid Chemistry, National Academy of Sciences of Ukraine, Vernadskiy ave.42, 03142 Kyiv, Ukraine
| | - Andriy Yaroshchuk
- ICREA, pg. L.Companys 23, 08010 Barcelona, Spain
- Department of Chemical Engineering, Polytechnic University of Catalonia–Barcelona Tech, av. Diagonal 647, 08028 Barcelona, Spain
| |
Collapse
|
16
|
Kim M, Kim B. Preconcentration of Fluorescent Dyes in Electromembrane Systems via Electrophoretic Migration. MICROMACHINES 2023; 14:398. [PMID: 36838098 PMCID: PMC9967745 DOI: 10.3390/mi14020398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
Microfluidic preconcentration enables the collection or extraction of low-abundance analytes at specific locations. It has attracted considerable attention as an essential technology in bioengineering, particularly for detection and diagnosis. Herein, we investigated the key parameters in the preconcentration of fluorescent dyes based on electrophoresis in a microfluidic electromembrane system. Commercial ion-exchange membrane (IEM)-integrated polydimethylsiloxane microfluidic devices were fabricated, and Alexa Fluor 488 and Rhodamine 6G were used as fluorescent dyes for sample preconcentration. Through experimental studies, the effect of the channel concentration ratio (CCR, concentration ratio of the main and buffer channels) on the performance of the sample preconcentration was studied. The results show that the preconcentration of the target sample occurs more effectively for a high CCR or high salt concentration of the main channel when the CCR is constant. We also demonstrate a phenomenon that the salt concentration in the electrolyte solution increases as the preconcentration progresses. Our results provide consolidated conditions for electrophoresis-based sample preconcentration in electromembrane systems.
Collapse
Affiliation(s)
- Minsung Kim
- Department of Future Convergence Engineering, Kongju National University, Cheonan 31080, Republic of Korea
| | - Bumjoo Kim
- Department of Future Convergence Engineering, Kongju National University, Cheonan 31080, Republic of Korea
- Department of Mechanical and Automotive Engineering, Kongju National University, Cheonan 31080, Republic of Korea
| |
Collapse
|
17
|
Gong X, Chi H, Strohmer DF, Teichmann AT, Xia Z, Wang Q. Exosomes: A potential tool for immunotherapy of ovarian cancer. Front Immunol 2023; 13:1089410. [PMID: 36741380 PMCID: PMC9889675 DOI: 10.3389/fimmu.2022.1089410] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
Ovarian cancer is a malignant tumor of the female reproductive system, with a very poor prognosis and high mortality rates. Chemotherapy and radiotherapy are the most common treatments for ovarian cancer, with unsatisfactory results. Exosomes are a subpopulation of extracellular vesicles, which have a diameter of approximately 30-100 nm and are secreted by many different types of cells in various body fluids. Exosomes are highly stable and are effective carriers of immunotherapeutic drugs. Recent studies have shown that exosomes are involved in various cellular responses in the tumor microenvironment, influencing the development and therapeutic efficacy of ovarian cancer, and exhibiting dual roles in inhibiting and promoting tumor development. Exosomes also contain a variety of genes related to ovarian cancer immunotherapy that could be potential biomarkers for ovarian cancer diagnosis and prognosis. Undoubtedly, exosomes have great therapeutic potential in the field of ovarian cancer immunotherapy. However, translation of this idea to the clinic has not occurred. Therefore, it is important to understand how exosomes could be used in ovarian cancer immunotherapy to regulate tumor progression. In this review, we summarize the biomarkers of exosomes in different body fluids related to immunotherapy in ovarian cancer and the potential mechanisms by which exosomes influence immunotherapeutic response. We also discuss the prospects for clinical application of exosome-based immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Dorothee Franziska Strohmer
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Tobias Teichmann
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
18
|
McCarthy KP, Go DB, Senapati S, Chang HC. An integrated ion-exchange membrane-based microfluidic device for irreversible dissociation and quantification of miRNA from ribonucleoproteins. LAB ON A CHIP 2023; 23:285-294. [PMID: 36524732 PMCID: PMC10697430 DOI: 10.1039/d2lc00517d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Ribonucleoproteins (RNPs), particularly microRNA-induced silencing complex (miRISC), have been associated with cancer-related gene regulation. Specific RNA-protein associations in miRISC complexes or those found in let-7 lin28A complexes can downregulate tumor-suppressing genes and can be directly linked to cancer. The high protein-RNA electrostatic binding affinity is a particular challenge for the quantification of the associated microRNAs (miRNAs). We report here the first microfluidic point-of-care assay that allows direct quantification of RNP-associated RNAs, which has the potential to greatly advance RNP profiling for liquid biopsy. Key to the technology is an integrated cation-anion exchange membrane (CEM/AEM) platform for rapid and irreversible dissociation (k = 0.0025 s-1) of the RNP (Cas9-miR-21) complex and quantification of its associated miR-21 in 40 minutes. The CEM-induced depletion front is used to concentrate the RNP at the depletion front such that the high electric field (>100 V cm-1) within the concentration boundary layer induces irreversible dissociation of the low KD (∼0.5 nM) complex, with ∼100% dissociation even though the association rate (kon = 6.1 s-1) is 1000 times higher. The high field also electrophoretically drives the dissociated RNA out of the concentrated zone without reassociation. A detection limit of 1.1 nM is achieved for Cy3 labelled miR-21.
Collapse
Affiliation(s)
- Kyle P McCarthy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| | - David B Go
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| |
Collapse
|
19
|
Kumar K, Kim E, Alhammadi M, Umapathi R, Aliya S, Tiwari JN, Park HS, Choi JH, Son CY, Vilian AE, Han YK, Bu J, Huh YS. Recent advances in microfluidic approaches for the isolation and detection of exosomes. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2022.116912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
20
|
Advancement and obstacles in microfluidics-based isolation of extracellular vesicles. Anal Bioanal Chem 2023; 415:1265-1285. [PMID: 36284018 PMCID: PMC9928917 DOI: 10.1007/s00216-022-04362-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/01/2022]
Abstract
There is a great need for techniques which enable reproducible separation of extracellular vesicles (EVs) from biofluids with high recovery, purity and throughput. The development of new techniques for isolation of EVs from minute sample volumes is instrumental in enabling EV-based biomarker profiling in large biobank cohorts and paves the way to improved diagnostic profiles in precision medicine. Recent advances in microfluidics-based devices offer a toolbox for separating EVs from small sample volumes. Microfluidic devices that have been used in EV isolation utilise different fundamental principles and rely largely on benefits of scaling laws as the biofluid processing is miniaturised to chip level. Here, we review the progress in the practicality and performance of both passive devices (such as mechanical filtering and hydrodynamic focusing) and active devices (using magnetic, electric or acoustic fields). As it stands, many microfluidic devices isolate intact EV populations at higher purities than centrifugation, precipitation or size-exclusion chromatography. However, this comes at a cost. We address challenges (in particular low throughput, clogging risks and ability to process biofluids) and highlight the need for more improvements in microfluidic devices. Finally, we conclude that there is a need to refine and standardise these lab-on-a-chip techniques to meet the growing interest in the diagnostic and therapeutic value of purified EVs.
Collapse
|
21
|
Meggiolaro A, Moccia V, Brun P, Pierno M, Mistura G, Zappulli V, Ferraro D. Microfluidic Strategies for Extracellular Vesicle Isolation: Towards Clinical Applications. BIOSENSORS 2022; 13:bios13010050. [PMID: 36671885 PMCID: PMC9855931 DOI: 10.3390/bios13010050] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 05/15/2023]
Abstract
Extracellular vesicles (EVs) are double-layered lipid membrane vesicles released by cells. Currently, EVs are attracting a lot of attention in the biological and medical fields due to their role as natural carriers of proteins, lipids, and nucleic acids. Thus, they can transport useful genomic information from their parental cell through body fluids, promoting cell-to-cell communication even between different organs. Due to their functionality as cargo carriers and their protein expression, they can play an important role as possible diagnostic and prognostic biomarkers in various types of diseases, e.g., cancers, neurodegenerative, and autoimmune diseases. Today, given the invaluable importance of EVs, there are some pivotal challenges to overcome in terms of their isolation. Conventional methods have some limitations: they are influenced by the starting sample, might present low throughput and low purity, and sometimes a lack of reproducibility, being operator dependent. During the past few years, several microfluidic approaches have been proposed to address these issues. In this review, we summarize the most important microfluidic-based devices for EV isolation, highlighting their advantages and disadvantages compared to existing technology, as well as the current state of the art from the perspective of the use of these devices in clinical applications.
Collapse
Affiliation(s)
- Alessio Meggiolaro
- Department of Physics and Astronomy, University of Padua, Via Marzolo 8, 35131 Padua, Italy
| | - Valentina Moccia
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Legnaro, Italy
| | - Paola Brun
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Matteo Pierno
- Department of Physics and Astronomy, University of Padua, Via Marzolo 8, 35131 Padua, Italy
| | - Giampaolo Mistura
- Department of Physics and Astronomy, University of Padua, Via Marzolo 8, 35131 Padua, Italy
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Legnaro, Italy
| | - Davide Ferraro
- Department of Physics and Astronomy, University of Padua, Via Marzolo 8, 35131 Padua, Italy
- Correspondence:
| |
Collapse
|
22
|
Zhang S, Deng J, Li J, Tian F, Liu C, Fang L, Sun J. Advanced microfluidic technologies for isolating extracellular vesicles. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Bai J, Wei X, Zhang X, Wu C, Wang Z, Chen M, Wang J. Microfluidic strategies for the isolation and profiling of exosomes. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
24
|
An integrated lab-on-a-chip platform for pre-concentration and detection of colorectal cancer exosomes using anti-CD63 aptamer as a recognition element. Biosens Bioelectron 2022; 220:114856. [DOI: 10.1016/j.bios.2022.114856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/02/2022] [Accepted: 10/23/2022] [Indexed: 11/23/2022]
|
25
|
Lucotti S, Kenific CM, Zhang H, Lyden D. Extracellular vesicles and particles impact the systemic landscape of cancer. EMBO J 2022; 41:e109288. [PMID: 36052513 PMCID: PMC9475536 DOI: 10.15252/embj.2021109288] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022] Open
Abstract
Intercellular cross talk between cancer cells and stromal and immune cells is essential for tumor progression and metastasis. Extracellular vesicles and particles (EVPs) are a heterogeneous class of secreted messengers that carry bioactive molecules and that have been shown to be crucial for this cell-cell communication. Here, we highlight the multifaceted roles of EVPs in cancer. Functionally, transfer of EVP cargo between cells influences tumor cell growth and invasion, alters immune cell composition and function, and contributes to stromal cell activation. These EVP-mediated changes impact local tumor progression, foster cultivation of pre-metastatic niches at distant organ-specific sites, and mediate systemic effects of cancer. Furthermore, we discuss how exploiting the highly selective enrichment of molecules within EVPs has profound implications for advancing diagnostic and prognostic biomarker development and for improving therapy delivery in cancer patients. Altogether, these investigations into the role of EVPs in cancer have led to discoveries that hold great promise for improving cancer patient care and outcome.
Collapse
Affiliation(s)
- Serena Lucotti
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Candia M Kenific
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Haiying Zhang
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - David Lyden
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
26
|
Bayareh M. Active cell capturing for organ-on-a-chip systems: a review. BIOMED ENG-BIOMED TE 2022; 67:443-459. [PMID: 36062551 DOI: 10.1515/bmt-2022-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that has been proposed as a new powerful cell-based tool to imitate the pathophysiological environment of human organs. For most OOC systems, a pivotal step is to culture cells in microfluidic devices. In active cell capturing techniques, external actuators, such as electrokinetic, magnetic, acoustic, and optical forces, or a combination of these forces, can be applied to trap cells after ejecting cell suspension into the microchannel inlet. This review paper distinguishes the characteristics of biomaterials and evaluates microfluidic technology. Besides, various types of OOC and their fabrication techniques are reported and various active cell capture microstructures are analyzed. Furthermore, their constraints, challenges, and future perspectives are provided.
Collapse
Affiliation(s)
- Morteza Bayareh
- Department of Mechanical Engineering, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
27
|
Mousavi SM, Amin Mahdian SM, Ebrahimi MS, Taghizadieh M, Vosough M, Sadri Nahand J, Hosseindoost S, Vousooghi N, Javar HA, Larijani B, Hadjighassem MR, Rahimian N, Hamblin MR, Mirzaei H. Microfluidics for detection of exosomes and microRNAs in cancer: State of the art. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:758-791. [PMID: 35664698 PMCID: PMC9130092 DOI: 10.1016/j.omtn.2022.04.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Exosomes are small extracellular vesicles with sizes ranging from 30-150 nanometers that contain proteins, lipids, mRNAs, microRNAs, and double-stranded DNA derived from the cells of origin. Exosomes can be taken up by target cells, acting as a means of cell-to-cell communication. The discovery of these vesicles in body fluids and their participation in cell communication has led to major breakthroughs in diagnosis, prognosis, and treatment of several conditions (e.g., cancer). However, conventional isolation and evaluation of exosomes and their microRNA content suffers from high cost, lengthy processes, difficult standardization, low purity, and poor yield. The emergence of microfluidics devices with increased efficiency in sieving, trapping, and immunological separation of small volumes could provide improved detection and monitoring of exosomes involved in cancer. Microfluidics techniques hold promise for advances in development of diagnostic and prognostic devices. This review covers ongoing research on microfluidics devices for detection of microRNAs and exosomes as biomarkers and their translation to point-of-care and clinical applications.
Collapse
Affiliation(s)
- Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeid Ebrahimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women’s Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Akbari Javar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
28
|
Liu L, Yang R, Cui J, Chen P, Ri HC, Sun H, Piao X, Li M, Pu Q, Quinto M, Zhou JL, Shang HB, Li D. Circular Nonuniform Electric Field Gel Electrophoresis for the Separation and Concentration of Nanoparticles. Anal Chem 2022; 94:8474-8482. [PMID: 35652329 DOI: 10.1021/acs.analchem.2c01313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A circular nonuniform electric field strategy coupled with gel electrophoresis was proposed to control the precise separation and efficient concentration of nano- and microparticles. The circular nonuniform electric field has the feature of exponential increase in the electric field intensity along the radius, working with three functional zones of migration, acceleration, and concentration. The distribution form of electric field lines is regulated in functional zones to control the migration behaviors of particles for separation and concentration by altering the relative position of the ring electrode (outside) and rodlike electrode (inner). The circular nonuniform electric field promotes the target-type and high-precision separation of nanoparticles based on the difference in charge-to-size ratio. The concentration multiple of nanoparticles is also controlled randomly with the alternation of radius, taking advantage of vertical extrusion and concentric converging of the migration path. This work provides a brand new insight into the simultaneous separation and concentration of particles and is promising for developing a versatile tool for the separation and preparation of various samples instead of conventional methods.
Collapse
Affiliation(s)
- Lu Liu
- Department of Chemistry, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Ruilin Yang
- Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Jiaxuan Cui
- Department of Chemistry, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Peng Chen
- Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Hyok Chol Ri
- College of Pharmacy, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Huaze Sun
- Department of Chemistry, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Xiangfan Piao
- Department of Electronics, School of Engineering, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Minshu Li
- Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark
| | - Qiaosheng Pu
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, Department of Chemistry, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Maurizio Quinto
- DAFNE - Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, Via Napoli 25, I-71122 Foggia, Italy
| | - John L Zhou
- Centre for Green Technology, School of Civil and Environmental Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Hai-Bo Shang
- Department of Chemistry, Yanbian University, Park Road 977, Yanji 133002, Jilin, China.,Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| | - Donghao Li
- Department of Chemistry, Yanbian University, Park Road 977, Yanji 133002, Jilin, China.,Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University, Park Road 977, Yanji 133002, Jilin, China
| |
Collapse
|
29
|
Burtenshaw D, Regan B, Owen K, Collins D, McEneaney D, Megson IL, Redmond EM, Cahill PA. Exosomal Composition, Biogenesis and Profiling Using Point-of-Care Diagnostics—Implications for Cardiovascular Disease. Front Cell Dev Biol 2022; 10:853451. [PMID: 35721503 PMCID: PMC9198276 DOI: 10.3389/fcell.2022.853451] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Arteriosclerosis is an important age-dependent disease that encompasses atherosclerosis, in-stent restenosis (ISR), pulmonary hypertension, autologous bypass grafting and transplant arteriosclerosis. Endothelial dysfunction and the proliferation of vascular smooth muscle cell (vSMC)-like cells is a critical event in the pathology of arteriosclerotic disease leading to intimal-medial thickening (IMT), lipid retention and vessel remodelling. An important aspect in guiding clinical decision-making is the detection of biomarkers of subclinical arteriosclerosis and early cardiovascular risk. Crucially, relevant biomarkers need to be good indicators of injury which change in their circulating concentrations or structure, signalling functional disturbances. Extracellular vesicles (EVs) are nanosized membraneous vesicles secreted by cells that contain numerous bioactive molecules and act as a means of intercellular communication between different cell populations to maintain tissue homeostasis, gene regulation in recipient cells and the adaptive response to stress. This review will focus on the emerging field of EV research in cardiovascular disease (CVD) and discuss how key EV signatures in liquid biopsies may act as early pathological indicators of adaptive lesion formation and arteriosclerotic disease progression. EV profiling has the potential to provide important clinical information to complement current cardiovascular diagnostic platforms that indicate or predict myocardial injury. Finally, the development of fitting devices to enable rapid and/or high-throughput exosomal analysis that require adapted processing procedures will be evaluated.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology and Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Brian Regan
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Kathryn Owen
- Southern Health and Social Care Trust, Craigavon Area Hospital, Craigavon, United Kingdom
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), Ulster University, Belfast, United Kingdom
| | - David Collins
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - David McEneaney
- Southern Health and Social Care Trust, Craigavon Area Hospital, Craigavon, United Kingdom
| | - Ian L. Megson
- Division of Biomedical Sciences, Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Eileen M. Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Paul Aidan Cahill
- Vascular Biology and Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
- *Correspondence: Paul Aidan Cahill,
| |
Collapse
|
30
|
Park S, Sabbagh B, Abu-Rjal R, Yossifon G. Digital microfluidics-like manipulation of electrokinetically preconcentrated bioparticle plugs in continuous-flow. LAB ON A CHIP 2022; 22:814-825. [PMID: 35080550 DOI: 10.1039/d1lc00864a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Herein, we demonstrate digital microfluidics-like manipulations of preconcentrated biomolecule plugs within a continuous flow that is different from the commonly known digital microfluidics involving discrete (i.e. droplets) media. This is realized using one- and two-dimensional arrays of individually addressable ion-permselective membranes with interconnecting microfluidic channels. The location of powered electrodes, dictates which of the membranes are active and generates either enrichment/depletion diffusion layers, which, in turn, control the location of the preconcentrated plug. An array of such powered membranes enables formation of multiple preconcentrated plugs of the same biosample as well as of preconcentrated plugs of multiple biosample types introduced via different inlets in a selective manner. Moreover, digital-microfluidics operations such as up-down and left-right translation, merging, and splitting, can be realized, but on preconcentrated biomolecule plugs instead of on discrete droplets. This technology, based on nanoscale electrokinetics of ion transport through permselective medium, opens future opportunities for smart and programmable digital-like manipulations of preconcentrated biological particle plugs for various on-chip biological applications.
Collapse
Affiliation(s)
- Sinwook Park
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion - Israel Institute of Technology, Technion City 3200000, Israel.
| | - Barak Sabbagh
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion - Israel Institute of Technology, Technion City 3200000, Israel.
| | - Ramadan Abu-Rjal
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion - Israel Institute of Technology, Technion City 3200000, Israel.
| | - Gilad Yossifon
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion - Israel Institute of Technology, Technion City 3200000, Israel.
| |
Collapse
|
31
|
Berzina B, Kim S, Peramune U, Saurabh K, Ganapathysubramanian B, Anand RK. Out-of-plane faradaic ion concentration polarization: stable focusing of charged analytes at a three-dimensional porous electrode. LAB ON A CHIP 2022; 22:573-583. [PMID: 35023536 DOI: 10.1039/d1lc01011e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Ion concentration polarization (ICP) accomplishes preconcentration for bioanalysis by localized depletion of electrolyte ions, thereby generating a gradient in electric field strength that facilitates electrokinetic focusing of charged analytes by their electromigration against opposing fluid flow. Such ICP focusing has been shown to accomplish up to a million-fold enrichment of nucleic acids and proteins in single-stage preconcentrators. However, the rate at which the sample volume is swept is limited, requiring several hours to achieve these high enrichment factors. This limitation is caused by two factors. First, an ion depleted zone (IDZ) formed at a planar membrane or electrode may not extend across the full channel cross section under the flow rate employed for focusing, thereby allowing the analyte to "leak" past the IDZ. Second, within the IDZ, large fluid vortices lead to mixing, which decreases the efficiency of analyte enrichment and worsens with increased channel dimensions. Here, we address these challenges with faradaic ICP (fICP) at a three-dimensional (3D) electrode comprising metallic microbeads. This 3D-electrode distributes the IDZ, and therefore, the electric field gradient utilized for counter-flow focusing across the full height of the fluidic channel, and its large area, microstructured surface supports smaller vortices. An additional bed of insulating microbeads restricts flow patterns and supplies a large area for surface conduction of ions through the IDZ. Finally, the resistance of this secondary bed enhances focusing by locally strengthening sequestering forces. This easy-to-build platform lays a foundation for the integration of enrichment with user-defined packed bed and electrode materials.
Collapse
Affiliation(s)
- Beatrise Berzina
- The Department of Chemistry, Iowa State University, 2415 Osborn Drive, 1605 Gilman Hall, Ames, Iowa 50011-1021, USA.
| | - Sungu Kim
- The Department of Chemistry, Iowa State University, 2415 Osborn Drive, 1605 Gilman Hall, Ames, Iowa 50011-1021, USA.
- The Department of Mechanical Engineering, Iowa State University, 2043 Black Engineering, 2529 Union Drive, Ames, Iowa 50011-2030, USA
| | - Umesha Peramune
- The Department of Chemistry, Iowa State University, 2415 Osborn Drive, 1605 Gilman Hall, Ames, Iowa 50011-1021, USA.
| | - Kumar Saurabh
- The Department of Mechanical Engineering, Iowa State University, 2043 Black Engineering, 2529 Union Drive, Ames, Iowa 50011-2030, USA
| | - Baskar Ganapathysubramanian
- The Department of Mechanical Engineering, Iowa State University, 2043 Black Engineering, 2529 Union Drive, Ames, Iowa 50011-2030, USA
| | - Robbyn K Anand
- The Department of Chemistry, Iowa State University, 2415 Osborn Drive, 1605 Gilman Hall, Ames, Iowa 50011-1021, USA.
| |
Collapse
|
32
|
Krishnamurthy A, Anand RK. Recent advances in microscale extraction driven by ion concentration polarization. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
33
|
Saad MG, Beyenal H, Dong WJ. Exosomes as Powerful Engines in Cancer: Isolation, Characterization and Detection Techniques. BIOSENSORS 2021; 11:518. [PMID: 34940275 PMCID: PMC8699402 DOI: 10.3390/bios11120518] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/28/2021] [Accepted: 12/02/2021] [Indexed: 06/01/2023]
Abstract
Exosomes, powerful extracellular nanovesicles released from almost all types of living cells, are considered the communication engines (messengers) that control and reprogram physiological pathways inside target cells within a community or between different communities. The cell-like structure of these extracellular vesicles provides a protective environment for their proteins and DNA/RNA cargos, which serve as biomarkers for many malicious diseases, including infectious diseases and cancers. Cancer-derived exosomes control cancer metastasis, prognosis, and development. In addition to the unique structure of exosomes, their nanometer size and tendency of interacting with cells makes them a viable novel drug delivery solution. In recent years, numerous research efforts have been made to quantify and characterize disease-derived exosomes for diagnosis, monitoring, and therapeutic purposes. This review aims to (1) relate exosome biomarkers to their origins, (2) focus on current isolation and detection methods, (3) discuss and evaluate the proposed technologies deriving from exosome research for cancer treatment, and (4) form a conclusion about the prospects of the current exosome research.
Collapse
Affiliation(s)
| | | | - Wen-Ji Dong
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, USA; (M.G.S.); (H.B.)
| |
Collapse
|
34
|
Sharma R, Geranpayehvaghei M, Ejeian F, Razmjou A, Asadnia M. Recent advances in polymeric nanostructured ion selective membranes for biomedical applications. Talanta 2021; 235:122815. [PMID: 34517671 DOI: 10.1016/j.talanta.2021.122815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/30/2022]
Abstract
Nano structured ion-selective membranes (ISMs) are very attractive materials for a wide range of sensing and ion separation applications. The present review focuses on the design principles of various ISMs; nanostructured and ionophore/ion acceptor doped ISMs, and their use in biomedical engineering. Applications of ISMs in the biomedical field have been well-known for more than half a century in potentiometric analysis of biological fluids and pharmaceutical products. However, the emergence of nanotechnology and sophisticated sensing methods assisted in miniaturising ion-selective electrodes to needle-like sensors that can be designed in the form of implantable or wearable devices (smartwatch, tattoo, sweatband, fabric patch) for health monitoring. This article provides a critical review of recent advances in miniaturization, sensing and construction of new devices over last decade (2011-2021). The designing of tunable ISM with biomimetic artificial ion channels offered intensive opportunities and innovative clinical analysis applications, including precise biosensing, controlled drug delivery and early disease diagnosis. This paper will also address the future perspective on potential applications and challenges in the widespread use of ISM for clinical use. Finally, this review details some recommendations and future directions to improve the accuracy and robustness of ISMs for biomedical applications.
Collapse
Affiliation(s)
- Rajni Sharma
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Marzieh Geranpayehvaghei
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia; Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-175, Iran
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, 73441-81746, Iran
| | - Amir Razmjou
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia; Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, 73441-81746, Iran; Centre for Technology in Water and Wastewater, University of Technology Sydney, New South Wales, Australia; UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Mohsen Asadnia
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
35
|
Hassanpour Tamrin S, Sanati Nezhad A, Sen A. Label-Free Isolation of Exosomes Using Microfluidic Technologies. ACS NANO 2021; 15:17047-17079. [PMID: 34723478 DOI: 10.1021/acsnano.1c03469] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Exosomes are cell-derived structures packaged with lipids, proteins, and nucleic acids. They exist in diverse bodily fluids and are involved in physiological and pathological processes. Although their potential for clinical application as diagnostic and therapeutic tools has been revealed, a huge bottleneck impeding the development of applications in the rapidly burgeoning field of exosome research is an inability to efficiently isolate pure exosomes from other unwanted components present in bodily fluids. To date, several approaches have been proposed and investigated for exosome separation, with the leading candidate being microfluidic technology due to its relative simplicity, cost-effectiveness, precise and fast processing at the microscale, and amenability to automation. Notably, avoiding the need for exosome labeling represents a significant advance in terms of process simplicity, time, and cost as well as protecting the biological activities of exosomes. Despite the exciting progress in microfluidic strategies for exosome isolation and the countless benefits of label-free approaches for clinical applications, current microfluidic platforms for isolation of exosomes are still facing a series of problems and challenges that prevent their use for clinical sample processing. This review focuses on the recent microfluidic platforms developed for label-free isolation of exosomes including those based on sieving, deterministic lateral displacement, field flow, and pinched flow fractionation as well as viscoelastic, acoustic, inertial, electrical, and centrifugal forces. Further, we discuss advantages and disadvantages of these strategies with highlights of current challenges and outlook of label-free microfluidics toward the clinical utility of exosomes.
Collapse
Affiliation(s)
- Sara Hassanpour Tamrin
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, CCIT 125, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
| | - Amir Sanati Nezhad
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, CCIT 125, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
| |
Collapse
|
36
|
Diaz-Armas GG, Cervantes-Gonzalez AP, Martinez-Duarte R, Perez-Gonzalez VH. Electrically driven microfluidic platforms for exosome manipulation and characterization. Electrophoresis 2021; 43:327-339. [PMID: 34717000 DOI: 10.1002/elps.202100202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 01/15/2023]
Abstract
Exosomes are small extracellular vesicles that can be obtained from several body fluids such as blood and urine. Since these vesicles can carry biomarkers and other cargo, they have application in healthcare diagnostics and therapeutics, such as liquid biopsies and drug delivery. Yet, their identification and separation from a sample remain challenging due to their high degree of heterogeneity and their co-existence with other bioparticles. In this contribution, we review the state-of-the-art on electrical techniques and methods to displace, selectively trap/isolate, and detect/characterize exosomes in microfluidic devices. Although there are many reviews focused on exosome separation using benchtop equipment, such as ultracentrifugation, there are limited reviews focusing on the use of electrical phenomena in microfluidic devices for exosome manipulation and detection. Here, we highlight contributions published during the past decade and present perspectives for this research field for the near future, outlining challenges to address in years to come.
Collapse
Affiliation(s)
- Gladys G Diaz-Armas
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| | | | - Rodrigo Martinez-Duarte
- Multiscale Manufacturing Laboratory, Department of Mechanical Engineering, Clemson University, Clemson, South Carolina, USA
| | | |
Collapse
|
37
|
Regan B, O'Kennedy R, Collins D. Advances in point-of-care testing for cardiovascular diseases. Adv Clin Chem 2021; 104:1-70. [PMID: 34462053 DOI: 10.1016/bs.acc.2020.09.001] [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: 11/29/2022]
Abstract
Point-of-care testing (POCT) is a specific format of diagnostic testing that is conducted without accompanying infrastructure or sophisticated instrumentation. Traditionally, such rapid sample-to-answer assays provide inferior analytical performances to their laboratory counterparts when measuring cardiac biomarkers. Hence, their potentially broad applicability is somewhat bound by their inability to detect clinically relevant concentrations of cardiac troponin (cTn) in the early stages of myocardial injury. However, the continuous refinement of biorecognition elements, the optimization of detection techniques, and the fabrication of tailored fluid handling systems to manage the sensing process has stimulated the production of commercial assays that can support accelerated diagnostic pathways. This review will present the latest commercial POC assays and examine their impact on clinical decision-making. The individual elements that constitute POC assays will be explored, with an emphasis on aspects that contribute to economically feasible and highly sensitive assays. Furthermore, the prospect of POCT imparting a greater influence on early interventions for medium to high-risk individuals and the potential to re-shape the paradigm of cardiovascular risk assessments will be discussed.
Collapse
Affiliation(s)
- Brian Regan
- School of Biotechnology, Dublin City University, Dublin, Ireland.
| | - Richard O'Kennedy
- School of Biotechnology, Dublin City University, Dublin, Ireland; Research Complex, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - David Collins
- School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
38
|
Di Santo R, Romanò S, Mazzini A, Jovanović S, Nocca G, Campi G, Papi M, De Spirito M, Di Giacinto F, Ciasca G. Recent Advances in the Label-Free Characterization of Exosomes for Cancer Liquid Biopsy: From Scattering and Spectroscopy to Nanoindentation and Nanodevices. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1476. [PMID: 34199576 PMCID: PMC8230295 DOI: 10.3390/nano11061476] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/26/2022]
Abstract
Exosomes (EXOs) are nano-sized vesicles secreted by most cell types. They are abundant in bio-fluids and harbor specific molecular constituents from their parental cells. Due to these characteristics, EXOs have a great potential in cancer diagnostics for liquid biopsy and personalized medicine. Despite this unique potential, EXOs are not yet widely applied in clinical settings, with two main factors hindering their translational process in diagnostics. Firstly, conventional extraction methods are time-consuming, require large sample volumes and expensive equipment, and often do not provide high-purity samples. Secondly, characterization methods have some limitations, because they are often qualitative, need extensive labeling or complex sampling procedures that can induce artifacts. In this context, novel label-free approaches are rapidly emerging, and are holding potential to revolutionize EXO diagnostics. These methods include the use of nanodevices for EXO purification, and vibrational spectroscopies, scattering, and nanoindentation for characterization. In this progress report, we summarize recent key advances in label-free techniques for EXO purification and characterization. We point out that these methods contribute to reducing costs and processing times, provide complementary information compared to the conventional characterization techniques, and enhance flexibility, thus favoring the discovery of novel and unexplored EXO-based biomarkers. In this process, the impact of nanotechnology is systematically highlighted, showing how the effectiveness of these techniques can be enhanced using nanomaterials, such as plasmonic nanoparticles and nanostructured surfaces, which enable the exploitation of advanced physical phenomena occurring at the nanoscale level.
Collapse
Affiliation(s)
- Riccardo Di Santo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
| | - Sabrina Romanò
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica Del Sacro Cuore, 00168 Roma, Italy
| | - Alberto Mazzini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica Del Sacro Cuore, 00168 Roma, Italy
| | - Svetlana Jovanović
- “Vinča” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Giuseppina Nocca
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gaetano Campi
- Rome International Centre Materials Science Superstripes RICMASS, via dei Sabelli 119A, 00185 Rome, Italy;
- Institute of Crystallography, CNR, via Salaria Km 29. 300, Monterotondo Stazione, 00016 Roma, Italy
| | - Massimiliano Papi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica Del Sacro Cuore, 00168 Roma, Italy
| | - Marco De Spirito
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica Del Sacro Cuore, 00168 Roma, Italy
| | - Flavio Di Giacinto
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica Del Sacro Cuore, 00168 Roma, Italy
| | - Gabriele Ciasca
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (R.D.S.); (S.R.); (A.M.); (G.N.); (M.P.); (F.D.G.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica Del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
39
|
Zhiyue M, Xichen Y, Li R, Yang Y, Huicheng F, Peng S. Recent advances in paper-based preconcentrators by utilizing ion concentration polarization. Electrophoresis 2021; 42:1340-1351. [PMID: 33768593 DOI: 10.1002/elps.202000291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 11/09/2022]
Abstract
One of the most cited limitations of biochemical detection is its poor sensitivity, owing to the relatively high complexity of micro-samples. Moreover, some samples cannot be easily self-replicated and their abundance cannot be increased through traditional technologies. Therefore, the preconcentration of low-abundance samples is a key requirement for microfluidic biological analysis. In recent years, the ion-concentration polarization phenomenon has aroused widespread interest in the application of microfluidic technology. In addition, paper-based materials are readily available, easy to modify, and exhibit good hydrophilicity. The study of the ion-concentration polarization preconcentration of micro-samples in paper-based microfluidic chips is of considerable significance. In this review, we discuss the development and applications of ion-concentration polarization paper-based preconcentrator in the past 5 years, with emphasis on key progresses in chip fabrication and performance optimization under different conditions. The current needs and development prospects in this field have also been discussed.
Collapse
Affiliation(s)
- Meng Zhiyue
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, P. R. China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, P. R. China
| | - Yuan Xichen
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, P. R. China.,Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, P. R. China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, P. R. China.,Yangtze River Delta Research Institute of Northwestern Polytechnical University, Taicang, P. R. China
| | - Ren Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, P. R. China.,Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, P. R. China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, P. R. China
| | - Yang Yang
- Ministry of Education Key Laboratory of Low-grade Energy Utilization Technologies and Systems, Chongqing University, Chongqing, P. R. China
| | - Feng Huicheng
- Unmanned System Research Institute, Northwestern Polytechnical University, Xi'an, P. R. China.,MOE Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an, P. R. China
| | - Shang Peng
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, P. R. China.,Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, P. R. China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, P. R. China
| |
Collapse
|
40
|
Wang J, Ma P, Kim DH, Liu BF, Demirci U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. NANO TODAY 2021; 37:101066. [PMID: 33777166 PMCID: PMC7990116 DOI: 10.1016/j.nantod.2020.101066] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Exosomes are a class of cell-secreted, nano-sized extracellular vesicles with a bilayer membrane structure of 30-150 nm in diameter. Their discovery and application have brought breakthroughs in numerous areas, such as liquid biopsies, cancer biology, drug delivery, immunotherapy, tissue repair, and cardiovascular diseases. Isolation of exosomes is the first step in exosome-related research and its applications. Standard benchtop exosome separation and sensing techniques are tedious and challenging, as they require large sample volumes, multi-step operations that are complex and time-consuming, requiring cumbersome and expensive instruments. In contrast, microfluidic platforms have the potential to overcome some of these limitations, owing to their high-precision processing, ability to handle liquids at a microscale, and integrability with various functional units, such as mixers, actuators, reactors, separators, and sensors. These platforms can optimize the detection process on a single device, representing a robust and versatile technique for exosome separation and sensing to attain high purity and high recovery rates with a short processing time. Herein, we overview microfluidic strategies for exosome isolation based on their hydrodynamic properties, size filtration, acoustic fields, immunoaffinity, and dielectrophoretic properties. We focus especially on advances in label-free isolation of exosomes with active biological properties and intact morphological structures. Further, we introduce microfluidic techniques for the detection of exosomal proteins and RNAs with high sensitivity, high specificity, and low detection limits. We summarize the biomedical applications of exosome-mediated therapeutic delivery targeting cancer cells. To highlight the advantages of microfluidic platforms, conventional techniques are included for comparison. Future challenges and prospects of microfluidics towards exosome isolation applications are also discussed. Although the use of exosomes in clinical applications still faces biological, technical, regulatory, and market challenges, in the foreseeable future, recent developments in microfluidic technologies are expected to pave the way for tailoring exosome-related applications in precision medicine.
Collapse
Affiliation(s)
- Jie Wang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Peng Ma
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Daniel H Kim
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Bi-Feng Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| |
Collapse
|
41
|
Lin B, Lei Y, Wang J, Zhu L, Wu Y, Zhang H, Wu L, Zhang P, Yang C. Microfluidic-Based Exosome Analysis for Liquid Biopsy. SMALL METHODS 2021; 5:e2001131. [PMID: 34927834 DOI: 10.1002/smtd.202001131] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/29/2020] [Indexed: 06/14/2023]
Abstract
Liquid biopsy offers non-invasive and real-time molecular profiling of individual patients, and is thus considered a revolutionary technology in precision medicine. Exosomes have been acknowledged as significant biomarkers in liquid biopsy, as they play a central role in cell-cell communication and are closely related to the pathogenesis of most human malignancies. Nevertheless, in biofluids exosomes always co-exist with other particles, and the cargo components of exosomes are highly heterogeneous. Thus, the isolation and molecular characterization of exosomes are still technically challenging. Microfluidics technology effectively addresses this challenge by virtue of its inherent advantages, such as precise manipulation of fluids, low consumption of samples and reagents, and a high level of integration. Recent advances in microfluidics allow in situ exosome capture and molecular detection with unprecedented selectivity and sensitivity. In this review, the state-of-the-art developments in microfluidics-based exosome research, including exosome isolation approaches and molecular detection strategies, with highlights of the characterization of exosomal biomarkers in cancer liquid biopsy is summarized. The major challenges are also discussed and some perspectives for the future directions of exosome-based liquid biopsy in microfluidic systems are presented.
Collapse
Affiliation(s)
- Bingqian Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yanmei Lei
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Junxia Wang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Lin Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yuqi Wu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Huimin Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Lingling Wu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Peng Zhang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
42
|
Bondarenko MP, Bruening ML, Yaroshchuk A. Electro-osmo-dialysis through nanoporous layers physically conjugated to micro-perforated ion-exchange membranes: Highly selective accumulation of trace coions. J Memb Sci 2021. [DOI: 10.1016/j.memsci.2020.119022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Morani M, Mai TD, Krupova Z, van Niel G, Defrenaix P, Taverna M. Recent electrokinetic strategies for isolation, enrichment and separation of extracellular vesicles. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
44
|
Xia Y, Chen T, Zhang L, Zhang X, Shi W, Chen G, Chen W, Lan J, Li C, Sun W, Chen J. Colorimetric detection of exosomal microRNA through switching the visible-light-induced oxidase mimic activity of acridone derivate. Biosens Bioelectron 2021; 173:112834. [DOI: 10.1016/j.bios.2020.112834] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/31/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
|
45
|
Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A 2020; 1636:461773. [PMID: 33316564 DOI: 10.1016/j.chroma.2020.461773] [Citation(s) in RCA: 301] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are heterogenous membrane-bound vesicles released from various origins. EVs play a crucial role in cellular communication and mediate several physiological and pathological processes, highlighting their potential therapeutic and diagnostic applications. Due to the rapid increase in interests and needs to elucidate EV properties and functions, numerous isolation and separation approaches for EVs have been developed to overcome limitations of conventional techniques, such as ultracentrifugation. This review focuses on recently emerging and modern EV isolation and separation techniques, including size-, charge-, and affinity-based techniques while excluding ultracentrifugation and precipitation-based techniques due to their multiple limitations. The advantages and drawbacks of each technique are discussed together with insights into their applications. Emerging approaches all share similar features in terms of being time-effective, easy-to-operate, and capable of providing EVs with suitable and desirable purity and integrity for applications of interest. Combination and hyphenation of techniques have been used for EV isolation and separation to yield EVs with the best quality. The most recent development using an automated on-line system including selective affinity-based trapping unit and asymmetrical flow field-flow fractionation allows reliable isolation and fractionation of EV subpopulations from human plasma.
Collapse
Affiliation(s)
| | - Evgen Multia
- Department of Chemistry, P.O. Box 55, FI-00014 University of Helsinki, Finland
| | | |
Collapse
|
46
|
Yadav V, Chong N, Ellis B, Ren X, Senapati S, Chang HC, Zorlutuna P. Constant-potential environment for activating and synchronizing cardiomyocyte colonies with on-chip ion-depleting perm-selective membranes. LAB ON A CHIP 2020; 20:4273-4284. [PMID: 33090162 DOI: 10.1039/d0lc00809e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, an ion depleted zone created by an ion-selective membrane was used to impose a high and uniform constant extracellular potential over an entire ∼1000 cell rat cardiomyocyte (rCM) colony on-a-chip to trigger synchronized voltage-gated ion channel activities while preserving cell viability, thus extending single-cell voltage-clamp ion channel studies to an entire normalized colony. Image analysis indicated that rCM beating was strengthened and accelerated (by a factor of ∼2) within minutes of ion depletion and the duration of contraction and relaxation phases was significantly reduced. After the initial synchronization, the entire colony responds collectively to external potential changes such that beating over the entire colony can be activated or deactivated within 0.1 s. These newly observed collective dynamic responses, due to simultaneous ion channel activation/deactivation by a uniform constant-potential extracellular environment, suggest that perm-selective membrane modules on cell culture chips can facilitate studies of extracellular cardiac cell electrical communication and how ion-channel related pathologies affect cardiac cell synchronization. The future applications of this new technology can lead to better drug screening platforms for cardiotoxicity as well as platforms that can facilitate synchronized maturation of pluripotent stem cells into colonies with high electrical connectivity.
Collapse
Affiliation(s)
- Vivek Yadav
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nicholas Chong
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Bradley Ellis
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA and Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
47
|
Wang C, Senapati S, Chang HC. Liquid biopsy technologies based on membrane microfluidics: High-yield purification and selective quantification of biomarkers in nanocarriers. Electrophoresis 2020; 41:1878-1892. [PMID: 32180242 PMCID: PMC7492446 DOI: 10.1002/elps.202000015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022]
Abstract
Liquid biopsy, screening cancer non-invasively and frequently by detecting and quantifying molecular markers in physiological fluids, would significantly improve cancer survival rate but it remains a distant goal. The key obstacles presented by the highly heterogeneous samples are rapid/high-yield purification and precise/selective marker capture by their antibody and oligo probes. As irregular expressions of these molecular biomarkers are the key signals, quantifying only those from the cancer cells would greatly enhance the performance of the screening tests. The recent discovery that the biomarkers are carried by nanocarriers, such as exosomes, with cell-specific membrane proteins suggests that such selection may be possible, although a new suite of fractionation and quantification technologies would need to be developed. Although under-appreciated, membrane microfluidics has made considerable contributions to resolving these issues. We review the progress made so far, based on ion-selective, track-etched, and gel membranes and advanced electrophoretic and nano-filtration designs, in this perspective and suggest future directions.
Collapse
Affiliation(s)
- Ceming Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| |
Collapse
|
48
|
Tutorial review: Enrichment and separation of neutral and charged species by ion concentration polarization focusing. Anal Chim Acta 2020; 1128:149-173. [PMID: 32825899 DOI: 10.1016/j.aca.2020.06.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 01/06/2023]
Abstract
Ion concentration polarization focusing (ICPF) is an electrokinetic technique, in which analytes are enriched and separated along a localized electric field gradient in the presence of a counter flow. This field gradient is generated by depletion of ions of the background electrolyte at an ion permselective junction. In this tutorial review, we summarize the fundamental principles and experimental parameters that govern selective ion transport and the stability of the enriched analyte plug. We also examine faradaic ICP (fICP), in which local ion concentration is modulated via electrochemical reactions as an attractive alternative to ICP that achieves similar performance with a decrease in both power consumption and Joule heating. The tutorial covers important challenges to the broad application of ICPF including undesired pH gradients, low volumetric throughput, samples that induce biofouling or are highly conductive, and limited approaches to on- or off-chip analysis. Recent developments in the field that seek to address these challenges are reviewed along with new approaches to maximize enrichment, focus uncharged analytes, and achieve enrichment and separation in water-in-oil droplets. For new practitioners, we discuss practical aspects of ICPF, such as strategies for device design and fabrication and the relative advantages of several types of ion selective junctions and electrodes. Lastly, we summarize tips and tricks for tackling common experimental challenges in ICPF.
Collapse
|
49
|
Han W, Chen X. A review: applications of ion transport in micro‐nanofluidic systems based on ion concentration polarization. JOURNAL OF CHEMICAL TECHNOLOGY & BIOTECHNOLOGY 2020; 95:1622-1631. [DOI: 10.1002/jctb.6288] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/22/2019] [Indexed: 01/12/2025]
Abstract
AbstractLab‐on‐a‐chip has been used widely in rapid, high‐throughput and low‐consumption analysis of samples in biochemistry. The ion concentration polarization (ICP) produced by ion‐selective transport of nanochannels provides a novel solution for problems in ultra‐low concentration sample detection, systems biology and desalination. This paper reviews the applications of ion transport based on the principle of ICP in micro‐nanofluidic systems. First, the fundamental governing equations of ICP are described. Then, the applications of nano‐electrokinetic ion enrichment and ion current rectification (ICR) are introduced. Nano‐electrokinetic ion enrichment is used mainly in the fields of molecular enrichment, ultra‐low concentration sample detection and seawater desalination. ICR is applied mainly to the sensitive detection of analytical substances such as proteins, nucleic acids and small molecules. The application of ion transport based on ICP principle is summarized and the possible directions worthy of further research are proposed. © 2019 Society of Chemical Industry
Collapse
Affiliation(s)
- Wenbo Han
- Faculty of Mechanical Engineering and Automation Liaoning University of Technology Jinzhou China
| | - Xueye Chen
- Faculty of Mechanical Engineering and Automation Liaoning University of Technology Jinzhou China
| |
Collapse
|
50
|
Ragab MAA, El-Kimary EI. Recent Advances and Applications of Microfluidic Capillary Electrophoresis: A Comprehensive Review (2017-Mid 2019). Crit Rev Anal Chem 2020; 51:709-741. [PMID: 32447968 DOI: 10.1080/10408347.2020.1765729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Microfluidic capillary electrophoresis (MCE) is the novel technique resulted from the CE mininaturization as planar separation and analysis device. This review presents and discusses various application fields of this advanced technology published in the period 2017 till mid-2019 in eight different sections including clinical, biological, single cell analysis, environmental, pharmaceuticals, food analysis, forensic and ion analysis. The need for miniaturization of CE and the consequence advantages achieved are also discussed including high-throughput, miniaturized detection, effective separation, portability and the need for micro- or even nano-volume of samples. Comprehensive tables for the MCE applications in the different studied fields are provided. Also, figure comparing the number of the published papers applying MCE in the eight discussed fields within the studied period is included. The future investigation should put into consideration the possibility of replacing conventional CE with the MCE after proper validation. Suitable validation parameters with their suitable accepted ranges should be tailored for analysis methods utilizing such unique technique (MCE).
Collapse
Affiliation(s)
- Marwa A A Ragab
- Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, Alexandria University, El-Messalah, Alexandria, Egypt
| | - Eman I El-Kimary
- Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, Alexandria University, El-Messalah, Alexandria, Egypt
| |
Collapse
|