101
|
Su W, Li H, Chen W, Qin J. Microfluidic strategies for label-free exosomes isolation and analysis. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
102
|
Seyfoori A, Seyyed Ebrahimi SA, Samiei E, Akbari M. Multifunctional Hybrid Magnetic Microgel Synthesis for Immune-Based Isolation and Post-Isolation Culture of Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:24945-24958. [PMID: 31268286 DOI: 10.1021/acsami.9b02959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Circulating tumor cells are of utmost importance among various biomarkers in liquid biopsies as a prognosis indicator of metastasis as well as in chemotherapeutic monitoring. This study introduces an efficient tool composed of soft nano/hybrid immune microgels for magnetic isolation of targeted tumor cells. The development process involves the in situ synthesis of magnetic nanoparticles within the three-dimensional matrix of thermoresponsive microgels. Surface modification and anti-EpCAM conjugation are adjusted by changing the temperature, and a conjugation efficiency of around 70% is achieved by using a protein G linker. Anti-EpCAM-conjugated nano/hybrid magnetic microgels are used to isolate EpCAM-expressing breast adenocarcinoma MCF-7 cells from culture media and whole blood with an efficiency of 75 and 70%, respectively. Furthermore, we demonstrate the ability of the hybrid microgels to isolate cancer cells with a purity of 65% and culture the cells post-isolation for further drug studies. The multifunctional hybrid microcarriers reported in this work can be potentially used for continuous monitoring of cancers and in personalized medicine.
Collapse
Affiliation(s)
- Amir Seyfoori
- Advanced Magnetic Materials Research Center, College of Engineering , University of Tehran , Tehran 14399-57131 , Iran
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute , ACECR , Tehran 1665659911 , Iran
| | - S A Seyyed Ebrahimi
- Advanced Magnetic Materials Research Center, College of Engineering , University of Tehran , Tehran 14399-57131 , Iran
| | | | | |
Collapse
|
103
|
Majarikar V, Takehara H, Ichiki T. Adsorption Phenomena of Anionic and Cationic Nanoliposomes on the Surface of Poly(dimethylsiloxane) Microchannel. J PHOTOPOLYM SCI TEC 2019. [DOI: 10.2494/photopolymer.32.107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Virendra Majarikar
- Department of Materials Engineering, School of Engineering, The University of Tokyo
| | - Hiroaki Takehara
- Department of Materials Engineering, School of Engineering, The University of Tokyo
- Innovation Center of NanoMedicine, Institute of Industry Promotion-Kawasaki
| | - Takanori Ichiki
- Department of Materials Engineering, School of Engineering, The University of Tokyo
- Innovation Center of NanoMedicine, Institute of Industry Promotion-Kawasaki
| |
Collapse
|
104
|
Zhang P, Zhou X, Zeng Y. Multiplexed immunophenotyping of circulating exosomes on nano-engineered ExoProfile chip towards early diagnosis of cancer. Chem Sci 2019; 10:5495-5504. [PMID: 31293733 PMCID: PMC6544119 DOI: 10.1039/c9sc00961b] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/19/2019] [Indexed: 12/28/2022] Open
Abstract
Circulating exosomes have been studied as a promising biomarker for non-invasive cancer diagnosis, as they are implicated in tumor initiation, progression, and metastasis. However, the clinical significance of circulating exosomes has not been revealed thoroughly, due to the technical limitation in sensitive and multiplexed detection of cargoes on exosomes, such as proteins and nucleic acids. Herein we developed an integrated exosome profiling platform (ExoProfile chip) to afford superior sensitivity and multiplexed capability for quantitative detection of a panel of surface protein markers on exosomes. To achieve this goal, we innovatively constructed 3D porous serpentine nanostructures via patterned colloidal self-assembly to provide enormous reaction sites and improve biosensing efficiency of exosomes. Meanwhile, the switchable microfluidic design enabled the simultaneous detection of eight markers on single addition of exosome samples. The ExoProfile chip was validated with purified exosomes from SKOV3 cells, which yielded a limit of detection of 21 exosomes per μL. We applied the ExoProfile chip to clinical analysis of circulating exosomes using only 10 μL ovarian cancer plasma and completing the analysis within 3 h. The diagnostic power of seven markers (EGFR, HER2, CA125, FRα, CD24, EpCAM, and CD9 plus CD63) were evaluated with receiver operator characteristic curve and heatmap clustering. Compared to single biomarker, the combined assessment of a biomarker panel was demonstrated to display improved accuracy in distinguishing early and late stage cancer. The results suggested the ExoProfile chip as a promising platform for molecular fingerprinting of circulating exosomes towards early cancer diagnosis.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Chemistry , Ralph N Adams Institute for Bioanalytical Chemistry , University of Kansas , Lawrence , KS 66045 , USA .
| | - Xin Zhou
- Department of Chemistry , Ralph N Adams Institute for Bioanalytical Chemistry , University of Kansas , Lawrence , KS 66045 , USA .
| | - Yong Zeng
- Department of Chemistry , Ralph N Adams Institute for Bioanalytical Chemistry , University of Kansas , Lawrence , KS 66045 , USA .
- University of Kansas Cancer Center , Kansas City , KS 66160 , USA
| |
Collapse
|
105
|
Monguió-Tortajada M, Gálvez-Montón C, Bayes-Genis A, Roura S, Borràs FE. Extracellular vesicle isolation methods: rising impact of size-exclusion chromatography. Cell Mol Life Sci 2019; 76:2369-2382. [PMID: 30891621 PMCID: PMC11105396 DOI: 10.1007/s00018-019-03071-y] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) include a variety of nanosized vesicles released to the extracellular microenvironment by the vast majority of cells transferring bioactive lipids, proteins, mRNA, miRNA or non-coding RNA, as means of intercellular communication. Remarkably, among other fields of research, their use has become promising for immunomodulation, tissue repair and as source for novel disease-specific molecular signatures or biomarkers. However, a major challenge is to define accurate, reliable and easily implemented techniques for EV isolation due to their nanoscale size and high heterogeneity. In this context, differential ultracentrifugation (dUC) has been the most widely used laboratory methodology, but alternative procedures have emerged to allow purer EV preparations with easy implementation. Here, we present and discuss the most used of the different EV isolation methods, focusing on the increasing impact of size exclusion chromatography (SEC) on the resulting EV preparations from in vitro cultured cells-conditioned medium and biological fluids. Comparatively, low protein content and cryo-electron microscopy analysis show that SEC removes most of the overabundant soluble plasma proteins, which are not discarded using dUC or precipitating agents, while being more user friendly and less time-consuming than gradient-based EV isolation. Also, SEC highly maintains the major EVs' characteristics, including vesicular structure and content, which guarantee forthcoming applications. In sum, together with scaling-up possibilities to increase EV recovery and manufacturing following high-quality standards, SEC could be easily adapted to most laboratories to assist EV-associated biomarker discovery and to deliver innovative cell-free immunomodulatory and pro-regenerative therapies.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, UAB, Barcelona, Spain
| | - Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.
| | - Francesc E Borràs
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.
| |
Collapse
|
106
|
The role of miRNAs as biomarkers in prostate cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2019; 781:165-174. [PMID: 31416574 DOI: 10.1016/j.mrrev.2019.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/13/2022]
Abstract
There is an urged need of non-invasive biomarkers for the implementation of precision medicine. These biomarkers are required to these days for improving prostate cancer (PCa) screening, treatment or stratification in current clinical strategies. There are several commercial kits (Oncotype DX genomic prostate score®, Prolaris®, among others) that use genomic changes, rearrangement or even non-coding RNA events. However, none of them are currently used in the routine clinical practice. Many recent studies indicate that miRNAs are relevant molecules (small single-stranded non-coding RNAs that regulate gene expression of more than 30% of human genes) to be implement non-invasive biomarkers. However, contrasting to others tumors, such as breast cancer where miR-21 seems to be consistently upregulated; PCa data are controversial. Here we reported an extended revision about the role of miRNAs in PCa including data of AR signaling, cell cycle, EMT process, CSCs regulation and even the role of miRNAs as PCa diagnostic, prognostic and predictive tool. It is known that current biomedical research uses big-data analysis like Next Generation Sequencing (NGS) analysis. We also conducted an extensive online search, including the main platforms and kits for miRNAs massive analysis (like MiSeq, Nextseq 550, or Ion S5™ systems) indicating their pros, cons and including pre-analytical and analytical issues of miRNA studies.
Collapse
|
107
|
Rojalin T, Phong B, Koster HJ, Carney RP. Nanoplasmonic Approaches for Sensitive Detection and Molecular Characterization of Extracellular Vesicles. Front Chem 2019; 7:279. [PMID: 31134179 PMCID: PMC6514246 DOI: 10.3389/fchem.2019.00279] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
All cells release a multitude of nanoscale extracellular vesicles (nEVs) into circulation, offering immense potential for new diagnostic strategies. Yet, clinical translation for nEVs remains a challenge due to their vast heterogeneity, our insufficient ability to isolate subpopulations, and the low frequency of disease-associated nEVs in biofluids. The growing field of nanoplasmonics is poised to address many of these challenges. Innovative materials engineering approaches based on exploiting nanoplasmonic phenomena, i.e., the unique interaction of light with nanoscale metallic materials, can achieve unrivaled sensitivity, offering real-time analysis and new modes of medical and biological imaging. We begin with an introduction into the basic structure and function of nEVs before critically reviewing recent studies utilizing nanoplasmonic platforms to detect and characterize nEVs. For the major techniques considered, surface plasmon resonance (SPR), localized SPR, and surface enhanced Raman spectroscopy (SERS), we introduce and summarize the background theory before reviewing the studies applied to nEVs. Along the way, we consider notable aspects, limitations, and considerations needed to apply plasmonic technologies to nEV detection and analysis.
Collapse
Affiliation(s)
- Tatu Rojalin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Brian Phong
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Hanna J. Koster
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Randy P. Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
108
|
Doan M, Carpenter AE. Leveraging machine vision in cell-based diagnostics to do more with less. NATURE MATERIALS 2019; 18:414-418. [PMID: 31000804 DOI: 10.1038/s41563-019-0339-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Affiliation(s)
- Minh Doan
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
109
|
Holliday LS, Truzman E, Zuo J, Han G, Torres-Medina R, Rody WJ. Extracellular vesicle identification in tooth movement models. Orthod Craniofac Res 2019; 22 Suppl 1:101-106. [PMID: 31074148 PMCID: PMC6512852 DOI: 10.1111/ocr.12287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) are 30-150 nm in diameter vesicles released by cells that serve important intercellular regulatory functions. EVs include exosomes and microvesicles. Exosomes form in multivesicular bodies and are released extracellularly as the multivesicular bodies fuse with the plasma membrane. Microvesicles bud directly from the plasma membrane. Here, we examine methods that are available or emerging to detect and study EVs during orthodontic tooth movement (OTM). EV's involvement in regulating bone remodelling associated with OTM may be demonstrated by adding isolated EVs to an animal model to change the rate of tooth movement. Exosomes in multivesicular bodies might be detected by immunogold labelling of markers in sections from the tooth and jaw and detection by electron microscopy. Gingival crevicular fluid (GCF) is enriched in EVs. Detection and characterization of EVs released by osteoclasts during resorption have been described, and this information could be used to analyse EVs in OTM models. Regulatory EVs may be enriched in the GCF from teeth that are being moved or are undergoing root resorption. Emerging approaches, including nanoparticle tracking, ExoView and micro- and nanofluidics, show promise for studying EVs in the GCF. Techniques that amplify signal, including polymerase chain reaction (PCR), provide the sensitivity necessary to utilize EVs from GCF as biomarkers. Studies of the role of EVs in OTM will provide fresh insight that may identify means for enhancing OTM procedures. EVs in GCF may include biomarkers for bone remodelling during OTM, orthodontic-associated root resorption, and other dental pathologies.
Collapse
Affiliation(s)
- L Shannon Holliday
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
- Department of Anatomy & Cell Biology, University of Florida College of Medicine, Gainesville, Florida
| | - Estella Truzman
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
| | - Jian Zuo
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
| | - Guanghong Han
- Department of Oral Geriatrics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Rosemarie Torres-Medina
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
| | - Wellington J Rody
- Department of Orthodontics and Pediatric Dentistry, Stony Brook School of Dental Medicine, Stony Brook, New York
| |
Collapse
|
110
|
Xie C, Ji N, Tang Z, Li J, Chen Q. The role of extracellular vesicles from different origin in the microenvironment of head and neck cancers. Mol Cancer 2019; 18:83. [PMID: 30954079 PMCID: PMC6451295 DOI: 10.1186/s12943-019-0985-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
The proliferation and metastasis ability of tumors are mediate by the "mutual dialogue" between cells in the tumor microenvironment (TME). Extracellular vesicles (EVs), mainly exosomes and microvesicles, play an important role in achieving intercellular substance transport and information transfer in the TME. Initially considered "garbage dumpsters" and later referred to as "signal boxes", EVs carry "cargo" (proteins, lipids, or nucleic acids) that can redirect the function of a recipient cell. Currently, the molecular mechanisms and clinical applications of EVs in head and neck cancers (HNCs) are still at an early stage and need to be further investigate. In this review, we provide insight into the TME of HNCs, classifying and summarizing EVs derived from different cell types and illuminating their complex signaling networks involved in mediating tumor proliferation, invasion and metastasis, vascular angiogenesis and cancer drug resistance. In addition, we highlight the application of EVs in HNCs, underlining the special pathological and physiological environment of HNCs. The application of tumor heterogeneous EVs in saliva and circulating blood diagnostics will provide a new perspective for the early screening, real-time monitoring and prognostic risk assessment of HNCs. Given the concept of precise and individual therapy, nanostructured EVs are equipped with superior characteristics of biocompatibility, low immunogenicity, loadability and modification ability, making these molecules one of the new strategies for HNCs treatment.
Collapse
Affiliation(s)
- Changqing Xie
- Department of Oral and Maxillofacial Surgery, Xiangya Stomalogical Hospital & School of Stomatology, Central South University, Changsha, 410078, Hunan, China.,State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhangui Tang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomalogical Hospital & School of Stomatology, Central South University, Changsha, 410078, Hunan, China.
| | - Jing Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
111
|
Abstract
The clinical utility of tissue biopsies in cancer management will continue to expand, especially with the evolving role of targeted therapies. "Liquid biopsy" refers to testing a patient's biofluid samples such as blood or urine to detect tumor-derived molecules and cells that can be used diagnostically and prognostically in the assessment of cancer. Many proof-of-concept and pilot studies have shown the clinical potential of liquid biopsies as diagnostic and prognostic markers which would provide a surrogate for the conventional "solid biopsy". In this review, we focus on three methods of liquid biopsy-circulating tumor cells, extracellular vesicles, and circulating tumor DNA-to provide a landscape view of their clinical applicability in cancer management and research.
Collapse
Affiliation(s)
- Matthew Scarlotta
- 1 Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Cem Simsek
- 2 Division of Gastroenterology and Hepatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Amy K Kim
- 2 Division of Gastroenterology and Hepatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
112
|
Lin S, Zhi X, Chen D, Xia F, Shen Y, Niu J, Huang S, Song J, Miao J, Cui D, Ding X. A flyover style microfluidic chip for highly purified magnetic cell separation. Biosens Bioelectron 2019; 129:175-181. [DOI: 10.1016/j.bios.2018.12.058] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/10/2018] [Accepted: 12/29/2018] [Indexed: 02/07/2023]
|
113
|
Li X, Corbett AL, Taatizadeh E, Tasnim N, Little JP, Garnis C, Daugaard M, Guns E, Hoorfar M, Li ITS. Challenges and opportunities in exosome research-Perspectives from biology, engineering, and cancer therapy. APL Bioeng 2019; 3:011503. [PMID: 31069333 PMCID: PMC6481742 DOI: 10.1063/1.5087122] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/08/2019] [Indexed: 12/11/2022] Open
Abstract
Exosomes are small (∼30-140 nm) lipid bilayer-enclosed particles of endosomal origin. They are a subset of extracellular vesicles (EVs) that are secreted by most cell types. There has been growing interest in exosome research in the last decade due to their emerging role as intercellular messengers and their potential in disease diagnosis. Indeed, exosomes contain proteins, lipids, and RNAs that are specific to their cell origin and could deliver cargo to both nearby and distant cells. As a result, investigation of exosome cargo contents could offer opportunities for disease detection and treatment. Moreover, exosomes have been explored as natural drug delivery vehicles since they can travel safely in extracellular fluids and deliver cargo to destined cells with high specificity and efficiency. Despite significant efforts made in this relatively new field of research, progress has been held back by challenges such as inefficient separation methods, difficulties in characterization, and lack of specific biomarkers. In this review, we summarize the current knowledge in exosome biogenesis, their roles in disease progression, and therapeutic applications and opportunities in bioengineering. Furthermore, we highlight the established and emerging technological developments in exosome isolation and characterization. We aim to consider critical challenges in exosome research and provide directions for future studies.
Collapse
Affiliation(s)
- Xia Li
- Department of Chemistry, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Alexander L. Corbett
- Department of Chemistry, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | | | - Nishat Tasnim
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Jonathan P. Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Cathie Garnis
- Department of Integrative Oncology, BC Cancer Agency, Vancouver, British Columbia, V5Z 1L3, Canada, and Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada, and Department of Urologic Sciences, University of British Columbia, Vancouver, Vancouver, BC V5Z 1M9, Canada
| | - Emma Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada, and Department of Urologic Sciences, University of British Columbia, Vancouver, Vancouver, BC V5Z 1M9, Canada
| | - Mina Hoorfar
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Isaac T. S. Li
- Department of Chemistry, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| |
Collapse
|
114
|
He D, Ho SL, Chan HN, Wang H, Hai L, He X, Wang K, Li HW. Molecular-Recognition-Based DNA Nanodevices for Enhancing the Direct Visualization and Quantification of Single Vesicles of Tumor Exosomes in Plasma Microsamples. Anal Chem 2019; 91:2768-2775. [PMID: 30644724 DOI: 10.1021/acs.analchem.8b04509] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor exosomes (Exo) are presumed to expedite both the growth and metastasis of tumors by actively participating in nearly all aspects of cancer development. Tumor-derived Exos are thus proposed as a resource for diagnostic biomarkers in bodily fluids. However, most Exo assays require large samples and are time-consuming, complicated, and costly, and thus unsuited for practical applications. Herein, we show an ultrasensitive assay that can directly visualize and quantify tumor Exos in plasma microsamples (1 μL) at the single-vesicle level. The assay uses the specific binding of activatable aptamer probes (AAP) to target Exos captured by Exo-specific antibodies on the surface of a flow cell to produce activated fluorescence. Furthermore, the bound AAP triggers in situ assembly of a DNA nanodevice with enhanced fluorescence that improves the Exo-detection sensitivity. By identifying tyrosine-protein-kinase-like 7 (PTK7), a total-internal-reflection-fluorescence (TIRF) assay for PTK7-Exo distinguishes target tumors from control subjects. This assay is also informative in monitoring tumor progression and early responses to therapy. The developed assay can be readily adapted for diagnosis and monitoring of other disease-associated Exo biomarkers.
Collapse
Affiliation(s)
- Dinggeng He
- Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong , China.,State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha 410006 , China
| | - See-Lok Ho
- Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong , China
| | - Hei-Nga Chan
- Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong , China
| | - Huizhen Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha 410006 , China
| | - Luo Hai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha 410006 , China
| | - Xiaoxiao He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha 410006 , China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering , Hunan University , Changsha 410006 , China
| | - Hung-Wing Li
- Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong , China
| |
Collapse
|
115
|
Seyfoori A, Seyyed Ebrahimi SA, Yousefi A, Akbari M. Efficient targeted cancer cell detection, isolation and enumeration using immuno-nano/hybrid magnetic microgels. Biomater Sci 2019; 7:3359-3372. [DOI: 10.1039/c9bm00552h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Magnetic nano/hybrid structures have drawn ample attention in the field of biotechnology due to their excellent magnetic properties and biocompatibility.
Collapse
Affiliation(s)
- Amir Seyfoori
- Advanced Magnetic Materials Research Center
- College of Engineering
- University of Tehran
- Tehran
- Iran
| | - S. A. Seyyed Ebrahimi
- Advanced Magnetic Materials Research Center
- College of Engineering
- University of Tehran
- Tehran
- Iran
| | - Arman Yousefi
- Advanced Magnetic Materials Research Center
- College of Engineering
- University of Tehran
- Tehran
- Iran
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME)
- Department of Mechanical Engineering
- University of Victoria
- Canada
- Center for Biomedical Research
| |
Collapse
|
116
|
Ayala-Mar S, Gallo-Villanueva RC, González-Valdez J. Dielectrophoretic manipulation of exosomes in a multi-section microfluidic device. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.matpr.2019.03.162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
117
|
Kalishwaralal K, Kwon WY, Park KS. Exosomes for Non-Invasive Cancer Monitoring. Biotechnol J 2018; 14:e1800430. [PMID: 30358137 DOI: 10.1002/biot.201800430] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Exosomes, membrane-bound phospholipid vesicles having diameters of 50-200 nm, are secreted by all cell types and circulate in human body fluids. These vesicles are known to carry cellular constituents that are specific to the originating cells (e.g., cytoplasmic/membrane proteins, RNA, and DNA). Thus, exosomes, which are both structurally stable and abundant, are robust indicators of cancers and, as a result, they have been utilized to monitor this disease in a manner that is less invasive than gold standard tissue biopsies. In this review, the history of exosomes and the specific biomarkers present in exosomes that enable accurate monitoring of various diseases are described. In addition, methods for analysis of exosomes and identification of biomarkers are presented with special emphasis being given to isolation and signaling strategies. Lastly, integrated, microfluidic systems developed for exosome-based cancer diagnosis are described and future directions that research in this area will likely take are presented.
Collapse
Affiliation(s)
- Kalimuthu Kalishwaralal
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Woo Young Kwon
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ki Soo Park
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| |
Collapse
|
118
|
Zhang P, Samuel G, Crow J, Godwin AK, Zeng Y. Molecular assessment of circulating exosomes toward liquid biopsy diagnosis of Ewing sarcoma family of tumors. Transl Res 2018; 201:136-153. [PMID: 30031766 PMCID: PMC6424494 DOI: 10.1016/j.trsl.2018.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/18/2018] [Accepted: 05/27/2018] [Indexed: 12/16/2022]
Abstract
Ewing sarcoma was first described in 1921 in the Proceedings of the New York Pathological Society by an eminent American pathologist from Cornell named James R. Ewing as a "diffuse endothelioma of bone." Since this initial description, more has been discovered regarding Ewing sarcoma and in the 1980's both Ewing sarcoma and peripheral primitive neuroectodermal tumors due to their similar features and shared identical genetic abnormality were grouped into a class of cancers entitled Ewing sarcoma family of tumors (ESFTs). Ewing sarcoma is the second most common pediatric osseous malignancy followed by osteosarcoma, with highest incidence among 10-20 years old. Ewing sarcoma is consistently associated with chromosomal translocation and functional fusion of the EWSR1 gene to any of several structurally related transcription factor genes of the E26 transformation-specific family. These tumor-specific molecular rearrangements are useful for primary diagnosis, may provide prognostic information, and present potential therapeutic targets. Therefore, ways to rapidly and efficiently detect these defining genomic alterations are of clinical relevance. Within the past decade, liquid biopsies including extracellular vesicles (EVs), have emerged as a promising alternative and/or complimentary approach to standard tumor biopsies. It was recently reported that fusion mRNAs from tumor-specific chromosome translocations can be detected in Ewing sarcoma cell-derived exosomes. Within this review, we overview the current advances in Ewing sarcoma and the opportunities and challenges in exploiting circulating exosomes, primarily small bioactive EVs (30-180 nm), as developing sources of biomarkers for diagnosis and therapeutic response monitoring in children and young adult patients with ESFT.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Chemistry, University of Kansas, Lawrence, Kansas
| | - Glenson Samuel
- Division of Hematology, Oncology and Bone Marrow Transplant, Children's Mercy Hospitals & Clinics, Kansas City, Missouri
| | - Jennifer Crow
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas; University of Kansas Cancer Center, Kansas City, Kansas.
| | - Yong Zeng
- Department of Chemistry, University of Kansas, Lawrence, Kansas; University of Kansas Cancer Center, Kansas City, Kansas.
| |
Collapse
|
119
|
Hirano K, Iwaki T, Ishido T, Yoshikawa Y, Naruse K, Yoshikawa K. Stretching of single DNA molecules caused by accelerating flow on a microchip. J Chem Phys 2018; 149:165101. [PMID: 30384753 DOI: 10.1063/1.5040564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
DNA elongation induced by fluidic stress was investigated on a microfluidic chip composed of a large inlet pool and a narrow channel. Through single-DNA observation with fluorescence microscopy, the manner of stretching of individual T4 DNA molecules (166 kbp) was monitored near the area of accelerating flow with narrowing streamlines. The results showed that the DNA long-axis length increased in a sigmoidal manner depending on the magnitude of flow acceleration, or shear, along the DNA chain. To elucidate the physical mechanism of DNA elongation, we performed a theoretical study by adopting a model of a coarse-grained nonlinear elastic polymer chain elongated by shear stress due to acceleration flow along the chain direction.
Collapse
Affiliation(s)
- Ken Hirano
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395 Japan
| | - Takafumi Iwaki
- Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Tomomi Ishido
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395 Japan
| | - Yuko Yoshikawa
- Faculty of Life and Medical Science, Doshisha Universiy, Kyotanabe, Kyoto 610-0321, Japan
| | - Keiji Naruse
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Kenichi Yoshikawa
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395 Japan
| |
Collapse
|
120
|
Tengattini S. Chromatographic Approaches for Purification and Analytical Characterization of Extracellular Vesicles: Recent Advancements. Chromatographia 2018. [DOI: 10.1007/s10337-018-3637-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
121
|
Hisey CL, Dorayappan KDP, Cohn DE, Selvendiran K, Hansford DJ. Microfluidic affinity separation chip for selective capture and release of label-free ovarian cancer exosomes. LAB ON A CHIP 2018; 18:3144-3153. [PMID: 30191215 DOI: 10.1039/c8lc00834e] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Exosomes are nanoscale vesicles found in many bodily fluids which play a significant role in cell-to-cell signaling and contain biomolecules indicative of their cells of origin. Recently, microfluidic devices have provided the ability to efficiently capture exosomes based on specific membrane biomarkers, but releasing the captured exosomes intact and label-free for downstream characterization and experimentation remains a challenge. We present a herringbone-grooved microfluidic device which is covalently functionalized with antibodies against general and cancer exosome membrane biomarkers (CD9 and EpCAM) to isolate exosomes from small volumes of high-grade serous ovarian cancer (HGSOC) serum. Following capture, intact exosomes are released label-free using a low pH buffer and immediately neutralized downstream to ensure their stability. Characterization of captured and released exosomes was performed using fluorescence microscopy, nanoparticle tracking analysis, flow-cytometry, and SEM. Our results demonstrate the successful isolation of intact and label-free exosomes, indicate that the amount of both total and EpCAM+ exosomes increases with HGSOC disease progression, and demonstrate the downstream internalization of isolated exosomes by OVCAR8 cells. This device and approach can be utilized for a nearly limitless range of downstream exosome analytical and experimental techniques, both on and off-chip.
Collapse
Affiliation(s)
- Colin L Hisey
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| | | | | | | | | |
Collapse
|
122
|
Gong Y, Fan N, Yang X, Peng B, Jiang H. New advances in microfluidic flow cytometry. Electrophoresis 2018; 40:1212-1229. [PMID: 30242856 DOI: 10.1002/elps.201800298] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/07/2018] [Accepted: 09/15/2018] [Indexed: 01/22/2023]
Abstract
In recent years, researchers are paying the increasing attention to the development of portable microfluidic diagnostic devices including microfluidic flow cytometry for the point-of-care testing. Microfluidic flow cytometry, where microfluidics and flow cytometry work together to realize novel functionalities on the microchip, provides a powerful tool for measuring the multiple characteristics of biological samples. The development of a portable, low-cost, and compact flow cytometer can benefit the health care in underserved areas such as Africa or Asia. In this article, we review recent advancements of microfluidics including sample pumping, focusing and sorting, novel detection approaches, and data analysis in the field of flow cytometry. The challenge of microfluidic flow cytometry is also examined briefly.
Collapse
Affiliation(s)
- Yanli Gong
- School of Mechanical and Electrical Engineering, University of Electronic Science and Technology of China, Chengdu, P. R. China
| | - Na Fan
- School of Mechanical and Electrical Engineering, University of Electronic Science and Technology of China, Chengdu, P. R. China
| | - Xu Yang
- School of Mechanical and Electrical Engineering, University of Electronic Science and Technology of China, Chengdu, P. R. China
| | - Bei Peng
- School of Mechanical and Electrical Engineering, University of Electronic Science and Technology of China, Chengdu, P. R. China
| | - Hai Jiang
- School of Mechanical and Electrical Engineering, University of Electronic Science and Technology of China, Chengdu, P. R. China
| |
Collapse
|
123
|
Phelps J, Sanati-Nezhad A, Ungrin M, Duncan NA, Sen A. Bioprocessing of Mesenchymal Stem Cells and Their Derivatives: Toward Cell-Free Therapeutics. Stem Cells Int 2018; 2018:9415367. [PMID: 30275839 PMCID: PMC6157150 DOI: 10.1155/2018/9415367] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/30/2018] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted tremendous research interest due to their ability to repair tissues and reduce inflammation when implanted into a damaged or diseased site. These therapeutic effects have been largely attributed to the collection of biomolecules they secrete (i.e., their secretome). Recent studies have provided evidence that similar effects may be produced by utilizing only the secretome fraction containing extracellular vesicles (EVs). EVs are cell-derived, membrane-bound vesicles that contain various biomolecules. Due to their small size and relative mobility, they provide a stable mechanism to deliver biomolecules (i.e., biological signals) throughout an organism. The use of the MSC secretome, or its components, has advantages over the implantation of the MSCs themselves: (i) signals can be bioengineered and scaled to specific dosages, and (ii) the nonliving nature of the secretome enables it to be efficiently stored and transported. However, since the composition and therapeutic benefit of the secretome can be influenced by cell source, culture conditions, isolation methods, and storage conditions, there is a need for standardization of bioprocessing parameters. This review focuses on key parameters within the MSC culture environment that affect the nature and functionality of the secretome. This information is pertinent to the development of bioprocesses aimed at scaling up the production of secretome-derived products for their use as therapeutics.
Collapse
Affiliation(s)
- Jolene Phelps
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
| | - Amir Sanati-Nezhad
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
| | - Mark Ungrin
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Faculty of Veterinary Medicine, Heritage Medical Research Building, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB, Canada T2N 4N1
| | - Neil A. Duncan
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Musculoskeletal Mechanobiology and Multiscale Mechanics Bioengineering Lab, Department of Civil Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
| | - 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, AB, Canada T2N 1N4
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, Canada T2N 1N4
| |
Collapse
|
124
|
Wu X, Li L, Iliuk A, Tao WA. Highly Efficient Phosphoproteome Capture and Analysis from Urinary Extracellular Vesicles. J Proteome Res 2018; 17:3308-3316. [PMID: 30080416 PMCID: PMC7236337 DOI: 10.1021/acs.jproteome.8b00459] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Analysis of protein phosphorylation in extracellular vesicles (EVs) offers an unprecedented potential for understanding cancer signaling and early stage disease diagnosis. However, prior to the phosphoproteome analysis step, the isolation of EVs from biofluids remains a challenging issue to overcome due to the low yield and impurity from current isolation methods. Here, we carry out an extensive assessment of several EV isolation methods including a novel rapid isolation method EVTRAP for highly efficient capture of extracellular vesicles from human urine sample. We demonstrate that over 95% recovery yield can be consistently achieved by EVTRAP, a significant improvement over current standard techniques. We then applied EVTRAP to identify over 16 000 unique peptides representing 2000 unique EV proteins from 200 μL urine sample, including all known EV markers with substantially increased recovery levels over ultracentrifugation. Most importantly, close to 2000 unique phosphopeptides were identified from more than 860 unique phosphoproteins using 10 mL of urine. The data demonstrated that EVTRAP is a highly effective and potentially widely implementable clinical isolation method for analysis of EV protein phosphorylation.
Collapse
Affiliation(s)
- Xiaofeng Wu
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Li Li
- Tymora Analytical Operations, West Lafayette, Indiana 47906, United States
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, Indiana 47906, United States
| | - W. Andy Tao
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
- Tymora Analytical Operations, West Lafayette, Indiana 47906, United States
| |
Collapse
|
125
|
Pick H, Alves AC, Vogel H. Single-Vesicle Assays Using Liposomes and Cell-Derived Vesicles: From Modeling Complex Membrane Processes to Synthetic Biology and Biomedical Applications. Chem Rev 2018; 118:8598-8654. [PMID: 30153012 DOI: 10.1021/acs.chemrev.7b00777] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The plasma membrane is of central importance for defining the closed volume of cells in contradistinction to the extracellular environment. The plasma membrane not only serves as a boundary, but it also mediates the exchange of physical and chemical information between the cell and its environment in order to maintain intra- and intercellular functions. Artificial lipid- and cell-derived membrane vesicles have been used as closed-volume containers, representing the simplest cell model systems to study transmembrane processes and intracellular biochemistry. Classical examples are studies of membrane translocation processes in plasma membrane vesicles and proteoliposomes mediated by transport proteins and ion channels. Liposomes and native membrane vesicles are widely used as model membranes for investigating the binding and bilayer insertion of proteins, the structure and function of membrane proteins, the intramembrane composition and distribution of lipids and proteins, and the intermembrane interactions during exo- and endocytosis. In addition, natural cell-released microvesicles have gained importance for early detection of diseases and for their use as nanoreactors and minimal protocells. Yet, in most studies, ensembles of vesicles have been employed. More recently, new micro- and nanotechnological tools as well as novel developments in both optical and electron microscopy have allowed the isolation and investigation of individual (sub)micrometer-sized vesicles. Such single-vesicle experiments have revealed large heterogeneities in the structure and function of membrane components of single vesicles, which were hidden in ensemble studies. These results have opened enormous possibilities for bioanalysis and biotechnological applications involving unprecedented miniaturization at the nanometer and attoliter range. This review will cover important developments toward single-vesicle analysis and the central discoveries made in this exciting field of research.
Collapse
Affiliation(s)
- Horst Pick
- Institute of Chemical Sciences and Engineering , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| | - Ana Catarina Alves
- Institute of Chemical Sciences and Engineering , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| | - Horst Vogel
- Institute of Chemical Sciences and Engineering , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| |
Collapse
|
126
|
Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine - a new paradigm for tissue repair. Biomater Sci 2018; 6:60-78. [PMID: 29184934 DOI: 10.1039/c7bm00479f] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue regeneration by stem cells is driven by the paracrine activity of shedding vesicles and exosomes, which deliver specific cargoes to the recipient cells. Proteins, RNA, cytokines and subsequent gene expression, orchestrate the regeneration process by improving the microenvironment to promote cell survival, controlling inflammation, repairing injury and enhancing the healing process. The action of microRNA is widely accepted as an essential driver of the regenerative process through its impact on multiple downstream biological pathways, and its ability to regulate the host immune response. Here, we present an overview of the recent potential uses of exosomes for regenerative medicine and tissue engineering. We also highlight the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal microRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.
Collapse
Affiliation(s)
- I M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | | | | | | | | |
Collapse
|
127
|
Dickhout A, Koenen RR. Extracellular Vesicles as Biomarkers in Cardiovascular Disease; Chances and Risks. Front Cardiovasc Med 2018; 5:113. [PMID: 30186839 PMCID: PMC6113364 DOI: 10.3389/fcvm.2018.00113] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/06/2018] [Indexed: 02/06/2023] Open
Abstract
The field of extracellular vesicles (EV) is rapidly expanding, also within cardiovascular diseases. Besides their exciting roles in cell-to-cell communication, EV have the potential to serve as excellent biomarkers, since their counts, content, and origin might provide useful information about the pathophysiology of cardiovascular disorders. Various studies have already indicated associations of EV counts and content with cardiovascular diseases. However, EV research is complicated by several factors, most notably the small size of EV. In this review, the advantages and drawbacks of EV-related methods and applications as biomarkers are highlighted.
Collapse
Affiliation(s)
- Annemiek Dickhout
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
128
|
Zhu Q, Heon M, Zhao Z, He M. Microfluidic engineering of exosomes: editing cellular messages for precision therapeutics. LAB ON A CHIP 2018; 18:1690-1703. [PMID: 29780982 PMCID: PMC5997967 DOI: 10.1039/c8lc00246k] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Study of extracellular vesicles (EVs), particularly exosomes, holds significant promise; however, it is technically challenging to define these small and molecularly diverse nanovesicles. With intrinsic molecular payload and biodegradability, molecular engineering of exosomes opens new avenues for mediating cellular responses and developing novel nano-delivery systems in precision therapeutics. Microfluidic lab-on-chip technology is playing pivotal roles in this emerging field. In this review, we have examined scientific advancements of microfluidic technology for engineering exosomes and assessed future applications and perspectives in developing precision therapeutics; this can serve the community via identification of potential new research areas or technologies that are urgently needed in precision therapeutics.
Collapse
Affiliation(s)
- Qingfu Zhu
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, USA.
| | | | | | | |
Collapse
|
129
|
Willms E, Cabañas C, Mäger I, Wood MJA, Vader P. Extracellular Vesicle Heterogeneity: Subpopulations, Isolation Techniques, and Diverse Functions in Cancer Progression. Front Immunol 2018; 9:738. [PMID: 29760691 PMCID: PMC5936763 DOI: 10.3389/fimmu.2018.00738] [Citation(s) in RCA: 599] [Impact Index Per Article: 99.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022] Open
Abstract
Cells release membrane enclosed nano-sized vesicles termed extracellular vesicles (EVs) that function as mediators of intercellular communication by transferring biological information between cells. Tumor-derived EVs have emerged as important mediators in cancer development and progression, mainly through transfer of their bioactive content which can include oncoproteins, oncogenes, chemokine receptors, as well as soluble factors, transcripts of proteins and miRNAs involved in angiogenesis or inflammation. This transfer has been shown to influence the metastatic behavior of primary tumors. Moreover, tumor-derived EVs have been shown to influence distant cellular niches, establishing favorable microenvironments that support growth of disseminated cancer cells upon their arrival at these pre-metastatic niches. It is generally accepted that cells release a number of major EV populations with distinct biophysical properties and biological functions. Exosomes, microvesicles, and apoptotic bodies are EV populations most widely studied and characterized. They are discriminated based primarily on their intracellular origin. However, increasing evidence suggests that even within these EV populations various subpopulations may exist. This heterogeneity introduces an extra level of complexity in the study of EV biology and function. For example, EV subpopulations could have unique roles in the intricate biological processes underlying cancer biology. Here, we discuss current knowledge regarding the role of subpopulations of EVs in cancer development and progression and highlight the relevance of EV heterogeneity. The position of tetraspanins and integrins therein will be highlighted. Since addressing EV heterogeneity has become essential for the EV field, current and novel techniques for isolating EV subpopulations will also be discussed. Further dissection of EV heterogeneity will advance our understanding of the critical roles of EVs in health and disease.
Collapse
Affiliation(s)
- Eduard Willms
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Carlos Cabañas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Department of Microbiology I (Immunology), Faculty of Medicine, Universidad Complutense, Madrid, Spain
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Institute of Technology, University of Tartu, Tartu, Estonia
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pieter Vader
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
130
|
Nogués L, Benito-Martin A, Hergueta-Redondo M, Peinado H. The influence of tumour-derived extracellular vesicles on local and distal metastatic dissemination. Mol Aspects Med 2018; 60:15-26. [PMID: 29196097 PMCID: PMC5856602 DOI: 10.1016/j.mam.2017.11.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/18/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are key mediators of intercellular communication that have been ignored for decades. Tumour cells benefit from the secretion of vesicles as they can influence the behaviour of neighbouring tumour cells within the tumour microenvironment. Several studies have shown that extracellular vesicles play an active role in pre-metastatic niche formation and importantly, they are involved in the metastatic organotropism of different tumour types. Tumour-derived EVs carry and transfer molecules to recipient cells, modifying their behaviour through a process defined as "EV-driven education". EVs favour metastasis to sentinel lymph nodes and distal organs by reinforcing angiogenesis, inflammation and lymphangiogenesis. Hence, in this review we will summarize the main mechanisms by which tumour-derived EVs regulate lymph node and distal organ metastasis. Moreover, since some cancers metastasize through the lymphatic system, we will discuss recent discoveries about the presence and function of tumour EVs in the lymph. Finally, we will address the potential value of tumour EVs as prognostic biomarkers in liquid biopsies, specially blood and lymphatic fluid, and the use of these tools as early detectors of metastases.
Collapse
Affiliation(s)
- Laura Nogués
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Alberto Benito-Martin
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Marta Hergueta-Redondo
- Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain
| | - Héctor Peinado
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA; Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain.
| |
Collapse
|
131
|
Advances in microfluidics for lipid nanoparticles and extracellular vesicles and applications in drug delivery systems. Adv Drug Deliv Rev 2018; 128:84-100. [PMID: 29567396 DOI: 10.1016/j.addr.2018.03.008] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023]
Abstract
Lipid-based nanobiomaterials as liposomes and lipid nanoparticles (LNPs) are the most widely used nanocarriers for drug delivery systems (DDSs). Extracellular vesicles (EVs) and exosomes are also expected to be applied as DDS nanocarriers. The performance of nanomedicines relies on their components such as lipids, targeting ligands, encapsulated DNA, encapsulated RNA, and drugs. Recently, the importance of the nanocarrier sizes smaller than 100nm is attracting attention as a means to improve nanomedicine performance. Microfluidics and lab-on-a chip technologies make it possible to produce size-controlled LNPs by a simple continuous flow process and to separate EVs from blood samples by using a surface marker, ligand, or electric charge or by making a mass or particle size discrimination. Here, we overview recent advances in microfluidic devices and techniques for liposomes, LNPs, and EVs and their applications for DDSs.
Collapse
|
132
|
Abstract
This chapter will discuss the potential use of microRNAs, particularly those located in peripherally-isolated exosomes, as biomarkers in neuropsychiatric disorders. These extracellular vesicles are released as a form of cell-to-cell communication and may mediate the soma-to-germline transmission of brain-relevant information, thereby potentially contributing to the inter- or transgenerational transmission of behavioral traits. Recent novel methods allow for the enrichment of peripheral exosomes specifically released by neurons and astrocytes and may provide valuable brain-relevant biosignatures of disease.
Collapse
Affiliation(s)
- Gabriel R Fries
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | - Joao Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| |
Collapse
|
133
|
Li A, Zhang T, Zheng M, Liu Y, Chen Z. Exosomal proteins as potential markers of tumor diagnosis. J Hematol Oncol 2017; 10:175. [PMID: 29282096 PMCID: PMC5745959 DOI: 10.1186/s13045-017-0542-8] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
Liquid biopsy especially that of exosomes carries tumor-specific molecules and provides useful information during tumor development and progression in "real time." Exosomes are membrane-encapsulated vesicles, constantly released by multiple cells, including cancer cells, in large quantities, and are widely present in body fluids. Tumor exosomes can remodel a tumor-supportive microenvironment via cross-talk with target cells. Recent research has mainly focused on exosomal miRNAs and to a small degree on proteins. However, detecting the genome output (active proteins such as phosphoproteins) can provide more direct information about disease progression, such as in the early discovery and monitoring of cancers. This review highlights the unique features of exosomal proteins over traditional serological markers and summarizes their recent use in cancer diagnosis and prognosis. Furthermore, we describe the general protocols of research on exosome proteomics with an emphasis on their clinical use.
Collapse
Affiliation(s)
- Aichun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Tianbao Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China.
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China.
| |
Collapse
|
134
|
Focus on Mesenchymal Stem Cell-Derived Exosomes: Opportunities and Challenges in Cell-Free Therapy. Stem Cells Int 2017; 2017:6305295. [PMID: 29410682 PMCID: PMC5749272 DOI: 10.1155/2017/6305295] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/05/2017] [Accepted: 11/22/2017] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells have been at the forefront of regenerative medicine for many years. Exosomes, which are nanovesicles involved in intercellular communication and the transportation of genetic material transportation that can be released by mesenchymal stem cells, have been recently reported to play a role in cell-free therapy of many diseases, including myocardial infarction, drug addiction, and status epilepticus. They are also thought to help ameliorate inflammation-induced preterm brain injury, liver injury, and various types of cancer. This review highlights recent advances in the exploration of mesenchymal stem cell-derived exosomes in therapeutic applications. The natural contents, drug delivery potency, modification methods, and drug loading methods of exosomes are also discussed.
Collapse
|
135
|
Chen X, Lan J, Liu Y, Li L, Yan L, Xia Y, Wu F, Li C, Li S, Chen J. A paper-supported aptasensor based on upconversion luminescence resonance energy transfer for the accessible determination of exosomes. Biosens Bioelectron 2017; 102:582-588. [PMID: 29241062 DOI: 10.1016/j.bios.2017.12.012] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/15/2017] [Accepted: 12/06/2017] [Indexed: 01/13/2023]
Abstract
Exosomes, as potential cancer diagnostic markers have received close attention in recent years. However, there is still a lack of simple and convenient methods to detect and quantitate exosomes. Herein, we used a simple paper-supported aptasensor based on luminescence resonance energy transfer (LRET) from upconversion nanoparticles (UCNPs) to gold nanorods (Au NRs) for the accessible determination of exosomes. When exosomes are present, the two sections of the aptamer can combine with the CD63 protein on the surface of exosomes and form a conjugation to close the distance between UCNPs and Au NRs, which initiates the LRET and promotes luminescence quenching. These variations can be monitored by the homemade image system, and the green channel intensities of obtained colored images were extracted with photoshop software to quantify the luminescence. As a result, the quenching of the luminescence of the UCNPs is linearly correlated to the concentration of the exosomes (in the range of 1.0 × 104 ~ 1.0 × 108 particles/μL), enabling the detection and quantification of the exosomes. Such approach can reach a low detection limit of exosomes (1.1 × 103 particles/μL) and effectively reduce the background signal by using UCNPs as a luminescent material. This study provides an efficient and practical approach to the detection of exosomes, which should lead to point-of-care testing in clinical applications.
Collapse
Affiliation(s)
- Xiaosong Chen
- Department of Plastic Surgery, The Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, PR China.
| | - Jianming Lan
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China
| | - Yingxin Liu
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China
| | - Li Li
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China
| | - Liu Yan
- Department of Plastic Surgery, The Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, PR China
| | - Yaokun Xia
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China
| | - Fang Wu
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China
| | - Chunyan Li
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China
| | - Shirong Li
- Department of Plastic Surgery, The Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, PR China; Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, PR China
| | - Jinghua Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350108, PR China.
| |
Collapse
|
136
|
A comprehensive overview of exosomes in ovarian cancer: emerging biomarkers and therapeutic strategies. J Ovarian Res 2017; 10:73. [PMID: 29100532 PMCID: PMC5670635 DOI: 10.1186/s13048-017-0368-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
Exosomes are nanoparticles(40-100 nm) secreted by most cells in the body, which can be isolated from several types of extracellular fluids. It has been shown that exosomes play a key role in intercellular communication and in transportation of genetic information. Emerging evidence shows that exosomes are mediators of metastasis in tumour cells, stromal cells and the extracellular matrix component through the shuttling of cargo, such as proteins, lipids, RNAs, double-stranded DNAs, non-transcribed RNAs, and microRNAs. This phenomenon has been indicated in both tumourigenesis and drug resistance. In this review, we introduce new methods of exosome extraction, focusing on the emerging role of exosomes in ovarian cancer, and discuss their potential clinical applications.
Collapse
|
137
|
Contreras-Naranjo JC, Wu HJ, Ugaz VM. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. LAB ON A CHIP 2017; 17:3558-3577. [PMID: 28832692 PMCID: PMC5656537 DOI: 10.1039/c7lc00592j] [Citation(s) in RCA: 398] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Exosomes, the smallest sized extracellular vesicles (∽30-150 nm) packaged with lipids, proteins, functional messenger RNAs and microRNAs, and double-stranded DNA from their cells of origin, have emerged as key players in intercellular communication. Their presence in bodily fluids, where they protect their cargo from degradation, makes them attractive candidates for clinical application as innovative diagnostic and therapeutic tools. But routine isolation and analysis of high purity exosomes in clinical settings is challenging, with conventional methods facing a number of drawbacks including low yield and/or purity, long processing times, high cost, and difficulties in standardization. Here we review a promising solution, microfluidic-based technologies that have incorporated a host of separation and sensing capabilities for exosome isolation, detection, and analysis, with emphasis on point-of-care and clinical applications. These new capabilities promise to advance fundamental research while paving the way toward routine exosome-based liquid biopsy for personalized medicine.
Collapse
Affiliation(s)
- Jose C Contreras-Naranjo
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA.
| | | | | |
Collapse
|
138
|
Samandari M, Abrinia K, Sanati-Nezhad A. Acoustic Manipulation of Bio-Particles at High Frequencies: An Analytical and Simulation Approach. MICROMACHINES 2017; 8:mi8100290. [PMID: 30400480 PMCID: PMC6190359 DOI: 10.3390/mi8100290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/16/2017] [Accepted: 09/21/2017] [Indexed: 01/09/2023]
Abstract
Manipulation of micro and nano particles in microfluidic devices with high resolution is a challenge especially in bioengineering applications where bio-particles (BPs) are separated or patterned. While acoustic forces have been used to control the position of BPs, its theoretical aspects need further investigation particularly for high-resolution manipulation where the wavelength and particle size are comparable. In this study, we used a finite element method (FEM) to amend analytical calculations of acoustic radiation force (ARF) arising from an imposed standing ultrasound field. First, an acoustic solid interaction (ASI) approach was implemented to calculate the ARF exerted on BPs and resultant deformation induced to them. The results were then used to derive a revised expression for the ARF beyond the small particle assumption. The expression was further assessed in numerical simulations of one- and multi-directional standing acoustic waves (SAWs). Furthermore, a particle tracing scheme was used to investigate the effect of actual ARF on separation and patterning applications under experimentally-relevant conditions. The results demonstrated a significant mismatch between the actual force and previous analytical predictions especially for high frequencies of manipulation. This deviation found to be not only because of the shifted ARF values but also due to the variation in force maps in multidirectional wave propagation. Findings of this work can tackle the simulation limitations for spatiotemporal control of BPs using a high resolution acoustic actuation.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- School of Mechanical Engineering, College of Engineering, University of Tehran, North Kargar St., Tehran 14395-515, Iran.
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK.
| | - Karen Abrinia
- School of Mechanical Engineering, College of Engineering, University of Tehran, North Kargar St., Tehran 14395-515, Iran.
| | - Amir Sanati-Nezhad
- Center for Bioengineering Research and Education, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
139
|
Phipps EA, Khankin EV. Extracellular Vesicles in Preeclampsia: Evolving Contributors to Proteinuria. J Am Soc Nephrol 2017; 28:3135-3137. [PMID: 28928135 DOI: 10.1681/asn.2017070789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Elizabeth A Phipps
- Renal Division, Brigham and Women's Hospital and.,Harvard Medical School, Boston, Massachusetts; and.,Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts
| | - Eliyahu V Khankin
- Harvard Medical School, Boston, Massachusetts; and .,Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
140
|
Kang YT, Kim YJ, Bu J, Cho YH, Han SW, Moon BI. High-purity capture and release of circulating exosomes using an exosome-specific dual-patterned immunofiltration (ExoDIF) device. NANOSCALE 2017; 9:13495-13505. [PMID: 28862274 DOI: 10.1039/c7nr04557c] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We present a microfluidic device for the capture and release of circulating exosomes from human blood. The exosome-specific dual-patterned immunofiltration (ExoDIF) device is composed of two distinct immuno-patterned layers, and is capable of enhancing the chance of binding between the antibody and exosomes by generating mechanical whirling, thus achieving high-throughput exosome isolation with high specificity. Moreover, follow-up recovery after the immuno-affinity based isolation, via cleavage of a linker, enables further downstream analysis. We verified the performance of the present device using MCF-7 secreted exosomes and found that both the concentration and proportion of exosome-sized vesicles were higher than in the samples obtained from the conventional exosome isolation kit. We then isolated exosomes from the human blood samples with our device to compare the exosome level between cancer patients and healthy donors. Cancer patients show a significantly higher exosome level with higher selectivity when validating the exosome-sized vesicles using both electron microscopy and nanoparticle tracking analysis. The captured exosomes from cancer patients also express abundant cancer-associated antigens, the epithelial cell adhesion molecule (EpCAM) on their surface. Our simple and rapid exosome recovery technique has huge potential to elucidate the function of exosomes in cancer patients and can thus be applied for various exosome-based cancer research studies.
Collapse
Affiliation(s)
- Yoon-Tae Kang
- Cell Bench Research Center, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
141
|
Parisse P, Rago I, Ulloa Severino L, Perissinotto F, Ambrosetti E, Paoletti P, Ricci M, Beltrami AP, Cesselli D, Casalis L. Atomic force microscopy analysis of extracellular vesicles. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:813-820. [PMID: 28866771 DOI: 10.1007/s00249-017-1252-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 08/23/2017] [Accepted: 08/27/2017] [Indexed: 12/17/2022]
Affiliation(s)
- P Parisse
- INSTM-ST Unit, Trieste, Italy.
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy.
| | - I Rago
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy
- University of Trieste, Trieste, Italy
| | - L Ulloa Severino
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy
- University of Trieste, Trieste, Italy
| | - F Perissinotto
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy
- University of Trieste, Trieste, Italy
| | - E Ambrosetti
- INSTM-ST Unit, Trieste, Italy
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy
- University of Trieste, Trieste, Italy
| | - P Paoletti
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy
- SISSA, Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy
| | - M Ricci
- Biological and Soft Systems, Cavendish Laboratory, Cambridge University, Cambridge, UK
| | - A P Beltrami
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - D Cesselli
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - L Casalis
- INSTM-ST Unit, Trieste, Italy
- Elettra, Sincrotrone Trieste S.C.p.A., Trieste, Italy
| |
Collapse
|
142
|
Pedde RD, Li H, Borchers CH, Akbari M. Microfluidic-Mass Spectrometry Interfaces for Translational Proteomics. Trends Biotechnol 2017; 35:954-970. [PMID: 28755975 DOI: 10.1016/j.tibtech.2017.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/05/2017] [Accepted: 06/09/2017] [Indexed: 12/29/2022]
Abstract
Interfacing mass spectrometry (MS) with microfluidic chips (μchip-MS) holds considerable potential to transform a clinician's toolbox, providing translatable methods for the early detection, diagnosis, monitoring, and treatment of noncommunicable diseases by streamlining and integrating laborious sample preparation workflows on high-throughput, user-friendly platforms. Overcoming the limitations of competitive immunoassays - currently the gold standard in clinical proteomics - μchip-MS can provide unprecedented access to complex proteomic assays having high sensitivity and specificity, but without the labor, costs, and complexities associated with conventional MS sample processing. This review surveys recent μchip-MS systems for clinical applications and examines their emerging role in streamlining the development and translation of MS-based proteomic assays by alleviating many of the challenges that currently inhibit widespread clinical adoption.
Collapse
Affiliation(s)
- R Daniel Pedde
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Rd., Victoria, BC, V8P 5C2, Canada; University of Victoria-Genome British Columbia Proteomics Centre, University of Victoria, 3101-4464 Markham St., Victoria, BC, V8Z 7X8, Canada
| | - Huiyan Li
- University of Victoria-Genome British Columbia Proteomics Centre, University of Victoria, 3101-4464 Markham St., Victoria, BC, V8Z 7X8, Canada
| | - Christoph H Borchers
- University of Victoria-Genome British Columbia Proteomics Centre, University of Victoria, 3101-4464 Markham St., Victoria, BC, V8Z 7X8, Canada; Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Rd., Victoria, BC, V8P 5C2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montreal, QC, H4A 3T2, Canada; Proteomics Centre, Jewish General Hospital, McGill University, 3755 Cote-Ste-Catherine Road, Montreal, QC, H3T 1E2, Canada.
| | - Mohsen Akbari
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Rd., Victoria, BC, V8P 5C2, Canada; Centre for Biomedical Research (CBR), University of Victoria, 3800 Finnerty Rd., Victoria, BC, V8P 5C2, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, 3800 Finnerty Rd., Victoria, BC, V8P 5C2, Canada.
| |
Collapse
|
143
|
Liu C, Guo J, Tian F, Yang N, Yan F, Ding Y, Wei J, Hu G, Nie G, Sun J. Field-Free Isolation of Exosomes from Extracellular Vesicles by Microfluidic Viscoelastic Flows. ACS NANO 2017; 11:6968-6976. [PMID: 28679045 DOI: 10.1021/acsnano.7b02277] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Exosomes, molecular cargos secreted by almost all mammalian cells, are considered as promising biomarkers to identify many diseases including cancers. However, the small size of exosomes (30-200 nm) poses serious challenges in their isolation from complex media containing a variety of extracellular vesicles (EVs) of different sizes, especially in small sample volumes. Here we present a viscoelasticity-based microfluidic system to directly separate exosomes from cell culture media or serum in a continuous, size-dependent, and label-free manner. Using a small amount of biocompatible polymer as the additive in the media to control the viscoelastic forces exerted on EVs, we are able to achieve a high separation purity (>90%) and recovery (>80%) of exosomes. The proposed technique may serve as a versatile platform to facilitate exosome analyses in diverse biochemical applications.
Collapse
Affiliation(s)
- Chao Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
| | - Jiayi Guo
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- College of Pharmaceutical Science, Jilin University , Changchun 130021, China
| | - Fei Tian
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
| | - Na Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
| | - Fusheng Yan
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
| | - Yanping Ding
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
| | - JingYan Wei
- College of Pharmaceutical Science, Jilin University , Changchun 130021, China
| | - Guoqing Hu
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Guangjun Nie
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jiashu Sun
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
144
|
Pariset E, Agache V, Millet A. Extracellular Vesicles: Isolation Methods. ACTA ACUST UNITED AC 2017; 1:e1700040. [DOI: 10.1002/adbi.201700040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/24/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Eloise Pariset
- CEA; LETI; MINATEC Campus 38054 Grenoble France
- Université Grenoble-Alpes; 38000 Grenoble France
| | - Vincent Agache
- CEA; LETI; MINATEC Campus 38054 Grenoble France
- Université Grenoble-Alpes; 38000 Grenoble France
| | - Arnaud Millet
- ATIP/Avenir Team “Mechanobiology, Immunity and Cancer”; Inserm U1205, Brain-Tech Lab 38054 Grenoble France
- Université Grenoble-Alpes; 38000 Grenoble France
| |
Collapse
|
145
|
Guo S, Dai Z, Hua J, Yang Z, Fang Z, Guo K. Microfluidic synthesis of α-ketoesters via oxidative coupling of acetophenones with alcohols under metal-free conditions. REACT CHEM ENG 2017. [DOI: 10.1039/c7re00107j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
An efficient and novel method for the synthesis of α-ketoesters has been developed via oxidative coupling of acetophenones with alcohols under TBHP/I2/DBU conditions in a microfluidic chip reactor, which has a wide substrate scope, uses a lower dosage of iodine and affords higher product yields in only a few seconds.
Collapse
Affiliation(s)
- Shiyu Guo
- College of Biotechnology and Pharmaceutical Engineering
- Nanjing Tech University
- Nanjing
- China
| | - Zhongxue Dai
- College of Biotechnology and Pharmaceutical Engineering
- Nanjing Tech University
- Nanjing
- China
| | - Jiawei Hua
- College of Biotechnology and Pharmaceutical Engineering
- Nanjing Tech University
- Nanjing
- China
| | - Zhao Yang
- College of Engineering
- China Pharmaceutical University
- Nanjing
- China
| | - Zheng Fang
- College of Biotechnology and Pharmaceutical Engineering
- Nanjing Tech University
- Nanjing
- China
| | - Kai Guo
- College of Biotechnology and Pharmaceutical Engineering
- Nanjing Tech University
- Nanjing
- China
- State Key Laboratory of Materials-Oriented Chemical Engineering
| |
Collapse
|