1
|
Sharma A, Yadav A, Nandy A, Ghatak S. Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine. Pharmaceutics 2024; 16:709. [PMID: 38931833 PMCID: PMC11206934 DOI: 10.3390/pharmaceutics16060709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Of all the numerous nanosized extracellular vesicles released by a cell, the endosomal-originated exosomes are increasingly recognized as potential therapeutics, owing to their inherent stability, low immunogenicity, and targeted delivery capabilities. This review critically evaluates the transformative potential of exosome-based modalities across pharmaceutical and precision medicine landscapes. Because of their precise targeted biomolecular cargo delivery, exosomes are posited as ideal candidates in drug delivery, enhancing regenerative medicine strategies, and advancing diagnostic technologies. Despite the significant market growth projections of exosome therapy, its utilization is encumbered by substantial scientific and regulatory challenges. These include the lack of universally accepted protocols for exosome isolation and the complexities associated with navigating the regulatory environment, particularly the guidelines set forth by the U.S. Food and Drug Administration (FDA). This review presents a comprehensive overview of current research trajectories aimed at addressing these impediments and discusses prospective advancements that could substantiate the clinical translation of exosomal therapies. By providing a comprehensive analysis of both the capabilities and hurdles inherent to exosome therapeutic applications, this article aims to inform and direct future research paradigms, thereby fostering the integration of exosomal systems into mainstream clinical practice.
Collapse
Affiliation(s)
| | | | | | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.S.); (A.Y.); (A.N.)
| |
Collapse
|
2
|
Weber B, Ritter A, Han J, Schaible I, Sturm R, Relja B, Huber-Lang M, Hildebrand F, Pallas C, Widera M, Henrich D, Marzi I, Leppik L. Development of a Sampling and Storage Protocol of Extracellular Vesicles (EVs)-Establishment of the First EV Biobank for Polytraumatized Patients. Int J Mol Sci 2024; 25:5645. [PMID: 38891833 PMCID: PMC11172154 DOI: 10.3390/ijms25115645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
In the last few years, several studies have emphasized the existence of injury-specific EV "barcodes" that could have significant importance for the precise diagnosis of different organ injuries in polytrauma patients. To expand the research potential of the NTF (network trauma research) biobank of polytraumatized patients, the NTF research group decided to further establish a biobank for EVs. However, until now, the protocols for the isolation, characterization, and storage of EVs for biobank purposes have not been conceptualized. Plasma and serum samples from healthy volunteers (n = 10) were used. Three EV isolation methods of high relevance for the work with patients' samples (ultracentrifugation, size exclusion chromatography, and immune magnetic bead-based isolation) were compared. EVs were quantified using nanoparticle tracking analysis, EV proteins, and miRNAs. The effects of different isolation solutions; the long storage of samples (up to 3 years); and the sensibility of EVs to serial freezing-thawing cycles and different storage conditions (RT, 4/-20/-80 °C, dry ice) were evaluated. The SEC isolation method was considered the most suitable for EV biobanking. We did not find any difference in the quantity of EVs between serum and plasma-EVs. The importance of particle-free PBS as an isolation solution was confirmed. Plasma that has been frozen for a long time can also be used as a source of EVs. Serial freezing-thawing cycles were found to affect the mean size of EVs but not their amount. The storage of EV samples for 5 days on dry ice significantly reduced the EV protein concentration.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Aileen Ritter
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Jiaoyan Han
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Inna Schaible
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Ramona Sturm
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Borna Relja
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, 89081 Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Christiane Pallas
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Ingo Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany (L.L.)
| |
Collapse
|
3
|
Onkar A, Khan F, Goenka A, Rajendran RL, Dmello C, Hong CM, Mubin N, Gangadaran P, Ahn BC. Smart Nanoscale Extracellular Vesicles in the Brain: Unveiling their Biology, Diagnostic Potential, and Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6709-6742. [PMID: 38315446 DOI: 10.1021/acsami.3c16839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Information exchange is essential for the brain, where it communicates the physiological and pathological signals to the periphery and vice versa. Extracellular vesicles (EVs) are a heterogeneous group of membrane-bound cellular informants actively transferring informative calls to and from the brain via lipids, proteins, and nucleic acid cargos. In recent years, EVs have also been widely used to understand brain function, given their "cell-like" properties. On the one hand, the presence of neuron and astrocyte-derived EVs in biological fluids have been exploited as biomarkers to understand the mechanisms and progression of multiple neurological disorders; on the other, EVs have been used in designing targeted therapies due to their potential to cross the blood-brain-barrier (BBB). Despite the expanding literature on EVs in the context of central nervous system (CNS) physiology and related disorders, a comprehensive compilation of the existing knowledge still needs to be made available. In the current review, we provide a detailed insight into the multifaceted role of brain-derived extracellular vesicles (BDEVs) in the intricate regulation of brain physiology. Our focus extends to the significance of these EVs in a spectrum of disorders, including brain tumors, neurodegenerative conditions, neuropsychiatric diseases, autoimmune disorders, and others. Throughout the review, parallels are drawn for using EVs as biomarkers for various disorders, evaluating their utility in early detection and monitoring. Additionally, we discuss the promising prospects of utilizing EVs in targeted therapy while acknowledging the existing limitations and challenges associated with their applications in clinical scenarios. A foundational comprehension of the current state-of-the-art in EV research is essential for informing the design of future studies.
Collapse
Affiliation(s)
- Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California 94143, United States
| | - Fatima Khan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Crismita Dmello
- Department of Neurological Surgery and Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Nida Mubin
- Department of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
4
|
Zhang F, Zhang L, Yu H. Potential Druggability of Mesenchymal Stem/Stromal Cell-derived Exosomes. Curr Stem Cell Res Ther 2024; 19:1195-1209. [PMID: 38523514 DOI: 10.2174/011574888x311270240319084835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024]
Abstract
Exosomes secreted by mesenchymal stem/stromal cells (MSC-Exos) are advantageous candidate sources for novel acellular therapy. Despite the current standards of good manufacturing practice (GMP), the deficiency of suitable quality-control methods and the difficulties in large-scale preparation largely restrict the development of therapeutic products and their clinical applications worldwide. Herein, we mainly focus on three dominating issues commonly encountered in exosomal GMP, including issues upstream of the cell culture process, downstream of the purification process, exosomes quality control, and the drug properties of exosomes and their druggability from a corporate perspective. Collectively, in this review article, we put forward the issues of preparing clinical exosome drugs for the treatment of diverse diseases and provide new references for the clinical application of GMP-grade MSC-Exos.
Collapse
Affiliation(s)
- Fan Zhang
- Faculty of Life Sciences and Medicine, Kunming University of Science and Technology, Kunming, 650500, China
| | - Leisheng Zhang
- Science and Technology Innovation Center, The Fourth People's Hospital of Jinan (The Third Affiliated Hospital of Shandong First Medical University), Jinan, 250031, China
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hao Yu
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
5
|
Kang C, He H, Liu P, Liu Y, Li X, Zhang J, Ran H, Zeng X, Zhao H, Liu J, Qiu S. Role of dendritic cell‑derived exosomes in allergic rhinitis (Review). Int J Mol Med 2023; 52:117. [PMID: 37888754 PMCID: PMC10635688 DOI: 10.3892/ijmm.2023.5320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Allergic rhinitis (AR) is a common pathological condition in otorhinolaryngology. Its prevalence has been increasing worldwide and is becoming a major burden to the world population. Dendritic cells (DCs) are typically activated and matured after capturing, phagocytosing, and processing allergens during the immunopathogenesis of AR. In addition, the process of DC activation and maturation is accompanied by the production of exosomes, which are cell‑derived extracellular vesicles (EVs) that can carry proteins, lipids, nucleic acids, and other cargoes involved in intercellular communication and material transfer. In particular, DC‑derived exosomes (Dex) can participate in allergic immune responses, where the biological substances carried by them can have potentially important implications for both the pathogenesis and treatment of AR. Dex can also be exploited to carry anti‑allergy agents to effectively treat AR. This provides a novel method to explore the pathogenesis of and treatment strategies for AR further. Therefore, the present review focuses on the origin, composition, function, and biological characteristics of DCs, exosomes, and Dex, in addition to the possible relationship between Dex and AR.
Collapse
Affiliation(s)
- Chenglin Kang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
- Department of Otolaryngology, Second People's Hospital of Gansu Province, Lanzhou, Gansu 730000, P.R. China
| | - Haipeng He
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Yue Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Xiaomei Li
- Department of Otolaryngology, Second People's Hospital of Gansu Province, Lanzhou, Gansu 730000, P.R. China
| | - Jin Zhang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
- Department of Otorhinolaryngology, The Second People's Hospital of Yibin, Yibin, Sichuan 644000, P.R. China
| | - Hong Ran
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Xianhai Zeng
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Hailiang Zhao
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Jiangqi Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Shuqi Qiu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519041, P.R. China
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
6
|
Marassi V, Giordani S, Placci A, Punzo A, Caliceti C, Zattoni A, Reschiglian P, Roda B, Roda A. Emerging Microfluidic Tools for Simultaneous Exosomes and Cargo Biosensing in Liquid Biopsy: New Integrated Miniaturized FFF-Assisted Approach for Colon Cancer Diagnosis. SENSORS (BASEL, SWITZERLAND) 2023; 23:9432. [PMID: 38067805 PMCID: PMC10708636 DOI: 10.3390/s23239432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023]
Abstract
The early-stage diagnosis of cancer is a crucial clinical need. The inadequacies of surgery tissue biopsy have prompted a transition to a less invasive profiling of molecular biomarkers from biofluids, known as liquid biopsy. Exosomes are phospholipid bilayer vesicles present in many biofluids with a biologically active cargo, being responsible for cell-to-cell communication in biological systems. An increase in their excretion and changes in their cargo are potential diagnostic biomarkers for an array of diseases, including cancer, and they constitute a promising analyte for liquid biopsy. The number of exosomes released, the morphological properties, the membrane composition, and their content are highly related to the physiological and pathological states. The main analytical challenge to establishing liquid biopsy in clinical practice is the development of biosensors able to detect intact exosomes concentration and simultaneously analyze specific membrane biomarkers and those contained in their cargo. Before analysis, exosomes also need to be isolated from biological fluids. Microfluidic systems can address several issues present in conventional methods (i.e., ultracentrifugation, size-exclusion chromatography, ultrafiltration, and immunoaffinity capture), which are time-consuming and require a relatively high amount of sample; in addition, they can be easily integrated with biosensing systems. A critical review of emerging microfluidic-based devices for integrated biosensing approaches and following the major analytical need for accurate diagnostics is presented here. The design of a new miniaturized biosensing system is also reported. A device based on hollow-fiber flow field-flow fractionation followed by luminescence-based immunoassay is applied to isolate intact exosomes and characterize their cargo as a proof of concept for colon cancer diagnosis.
Collapse
Affiliation(s)
- Valentina Marassi
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
- byFlow srl, 40129 Bologna, Italy
| | - Stefano Giordani
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
| | - Anna Placci
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
| | - Angela Punzo
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
| | - Cristiana Caliceti
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
- Interdepartmental Centre for Renewable Sources, Environment, Sea and Energy—CIRI FRAME, University of Bologna, 40131 Bologna, Italy
- Interdepartmental Centre for Industrial Agrofood Research—CIRI Agrofood, University of Bologna, 47521 Cesena, Italy
| | - Andrea Zattoni
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
- byFlow srl, 40129 Bologna, Italy
| | - Pierluigi Reschiglian
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
- byFlow srl, 40129 Bologna, Italy
| | - Barbara Roda
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
- byFlow srl, 40129 Bologna, Italy
| | - Aldo Roda
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (V.M.); (S.G.); (A.P.); (A.Z.); (P.R.)
- National Institute of Biostructure and Biosystems (INBB), 00136 Rome, Italy; (A.P.); (C.C.)
| |
Collapse
|
7
|
Man K, Eisenstein NM, Hoey DA, Cox SC. Bioengineering extracellular vesicles: smart nanomaterials for bone regeneration. J Nanobiotechnology 2023; 21:137. [PMID: 37106449 PMCID: PMC10134574 DOI: 10.1186/s12951-023-01895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In the past decade, extracellular vesicles (EVs) have emerged as key regulators of bone development, homeostasis and repair. EV-based therapies have the potential to circumnavigate key issues hindering the translation of cell-based therapies including functional tissue engraftment, uncontrolled differentiation and immunogenicity issues. Due to EVs' innate biocompatibility, low immunogenicity, and high physiochemical stability, these naturally-derived nanoparticles have garnered growing interest as potential acellular nanoscale therapeutics for a variety of diseases. Our increasing knowledge of the roles these cell-derived nanoparticles play, has made them an exciting focus in the development of novel pro-regenerative therapies for bone repair. Although these nano-sized vesicles have shown promise, their clinical translation is hindered due to several challenges in the EV supply chain, ultimately impacting therapeutic efficacy and yield. From the biochemical and biophysical stimulation of parental cells to the transition to scalable manufacture or maximising vesicles therapeutic response in vivo, a multitude of techniques have been employed to improve the clinical efficacy of EVs. This review explores state of the art bioengineering strategies to promote the therapeutic utility of vesicles beyond their native capacity, thus maximising the clinical potential of these pro-regenerative nanoscale therapeutics for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Neil M Eisenstein
- Research and Clinical Innovation, Royal Centre for Defence Medicine, ICT Centre, Vincent Drive, Birmingham, B15 2SQ, UK
- Institute of Translational Medicine, University of Birmingham, Heritage Building, Mindelsohn Way, Birmingham, B15 2TH, UK
| | - David A Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, D02 R590, Ireland
- Dept. of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College, Dublin 2, D02 DK07, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, D02 VN51, Dublin, Ireland
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
8
|
Lu S, Cui Q, Zheng H, Ma Y, Kang Y, Tang K. Challenges and Opportunities for Extracellular Vesicles in Clinical Oncology Therapy. Bioengineering (Basel) 2023; 10:bioengineering10030325. [PMID: 36978715 PMCID: PMC10045216 DOI: 10.3390/bioengineering10030325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles that can be released by all cell types. They may have different biogenesis, physical features, and cargo. EVs are important biomarkers for the diagnosis and prediction of many diseases due to their essential role in intercellular communication, their highly variable cargoes, and their accumulation in various body fluids. These natural particles have been investigated as potential therapeutic materials for many diseases. In our previous studies, the clinical usage of tumor-cell-derived microparticles (T-MPs) as a novel medication delivery system was examined. This review summarizes the clinical translation of EVs and related clinical trials, aiming to provide suggestions for safer and more effective oncology therapeutic systems, particularly in biotherapeutic and immunotherapeutic systems.
Collapse
Affiliation(s)
- Shuya Lu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qingfa Cui
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huan Zheng
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuan Ma
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanchun Kang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence:
| |
Collapse
|
9
|
Zhang Y, Liang F, Zhang D, Qi S, Liu Y. Metabolites as extracellular vesicle cargo in health, cancer, pleural effusion, and cardiovascular diseases: An emerging field of study to diagnostic and therapeutic purposes. Biomed Pharmacother 2023; 157:114046. [PMID: 36469967 DOI: 10.1016/j.biopha.2022.114046] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/19/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Extracellular vesicles (EVs) are highly diverse nanoscale membrane-bound structures released from different cell types into the extracellular environment. They play essential functions in cell signaling by transporting their cargo, such as proteins, RNA, DNA, lipids, metabolites, and small molecules, to recipient cells. It has recently been shown that EVs might modulate carcinogenesis by delivering cargo to recipient cells. Furthermore, recent discoveries revealed that changes in plasma-derived EV levels and cargo in subjects with metabolic diseases were documented by many researchers, suggesting that EVs might be a promising source of disease biomarkers. One of the cargos of EVs that has recently attracted the most attention is metabolites. The metabolome of these vesicles introduces a plethora of disease indicators; hence, examining the metabolomics of EVs detected in human biofluids would be an effective approach. On the other hand, metabolites have various roles in biological systems, including the production of energies, synthesizing macromolecules, and serving as signaling molecules and hormones. Metabolome rewiring in cancer and stromal cells is a characteristic of malignancy, but the current understanding of how this affects the metabolite composition and activity of tumor-derived EVs remains in its infancy. Since new findings and studies in the field of exosome biology and metabolism are constantly being published, it is likely that diagnostic and treatment techniques, including the use of exosome metabolites, will be launched in the coming years. Recent years have seen increased interest in the EV metabolome as a possible source for biomarker development. However, our understanding of the role of these molecules in health and disease is still immature. In this work, we have provided the latest findings regarding the role of metabolites as EV cargoes in the pathophysiology of diseases, including cancer, pleural effusion (PE), and cardiovascular disease (CVD). We also discussed the significance of metabolites as EV cargoes of microbiota and their role in host-microbe interaction. In addition, the latest findings on metabolites in the form of EV cargoes as biomarkers for disease diagnosis and treatment are presented in this study.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Feng Liang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - DuoDuo Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin Province 130021, People's Republic of China
| | - Shuang Qi
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.
| | - Yan Liu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China.
| |
Collapse
|
10
|
Bettio V, Mazzucco E, Antona A, Cracas S, Varalda M, Venetucci J, Bruno S, Chiabotto G, Venegoni C, Vasile A, Chiocchetti A, Quaglia M, Camussi G, Cantaluppi V, Panella M, Rolla R, Manfredi M, Capello D. Extracellular vesicles from human plasma for biomarkers discovery: Impact of anticoagulants and isolation techniques. PLoS One 2023; 18:e0285440. [PMID: 37163560 PMCID: PMC10171685 DOI: 10.1371/journal.pone.0285440] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023] Open
Abstract
Extracellular vesicles (EVs) isolated from plasma are increasingly recognized as promising circulating biomarkers for disease discovery and progression, as well as for therapeutic drug delivery. The scientific community underlined the necessity of standard operative procedures for the isolation and storage of the EVs to ensure robust results. The understanding of the impact of the pre-analytical variables is still limited and some considerations about plasma anticoagulants and isolation methods are necessary. Therefore, we performed a comparison study between EVs isolated by ultracentrifugation and by affinity substrate separation from plasma EDTA and sodium citrate. The EVs were characterized by Nano Tracking Analysis, Western Blot, cytofluorimetric analysis of surface markers, and lipidomic analysis. While anticoagulants did not significantly alter any of the analyzed parameters, the isolation methods influenced EVs size, purity, surface markers expression and lipidomic profile. Compared to ultracentrifugation, affinity substrate separation yielded bigger particles highly enriched in tetraspanins (CD9, CD63, CD81), fatty acids and glycerolipids, with a predominant LDL- and vLDL-like contamination. Herein, we highlighted that the isolation method should be carefully evaluated prior to study design and the need of standardized operative procedures for EVs isolation and application to biomarkers discovery.
Collapse
Affiliation(s)
- Valentina Bettio
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
- UPO Biobank, University of Piemonte Orientale, Novara, Italy
| | - Eleonora Mazzucco
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
- UPO Biobank, University of Piemonte Orientale, Novara, Italy
| | - Annamaria Antona
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Silvia Cracas
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Marco Varalda
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Jacopo Venetucci
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Giulia Chiabotto
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Chiara Venegoni
- Interdisciplinary Research Center of Autoimmune Diseases, Center on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
- Department of Health Science, "Maggiore della Carità" University Hospital, Novara, Italy
| | - Alessandra Vasile
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Annalisa Chiocchetti
- Interdisciplinary Research Center of Autoimmune Diseases, Center on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
- Department of Health Science, "Maggiore della Carità" University Hospital, Novara, Italy
| | - Marco Quaglia
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale, "Maggiore della Carità" University Hospital, Novara, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Vincenzo Cantaluppi
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale, "Maggiore della Carità" University Hospital, Novara, Italy
| | - Massimiliano Panella
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Roberta Rolla
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
- Clinical Chemistry, Azienda Ospedaliera-Universitaria "Maggiore della Carità", Università del Piemonte Orientale, Novara, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Daniela Capello
- Department of Translational Medicine, Center of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
- UPO Biobank, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
11
|
Benderitter M, Herrera-Reyes E, Tamarat R. Mesenchymal stromal cells in the regeneration of radiation-induced organ sequelae: will they make the difference? JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2022; 42:024001. [PMID: 35532367 DOI: 10.1088/1361-6498/ac6dd8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/04/2022] [Indexed: 06/14/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a stem cell product with good safety that demonstrate significant clinical efficacy in the treatment of different pathologies, including radiation diseases (e.g. radiological burns, pelvic radiation disease). While the first results for some first human applications for the treatment of radiation disease suggest benefit, larger trials with clinically important endpoints are needed before definitive conclusions can be drawn. However, the supply and cost of MSCs remain the two main limitations for this innovative therapeutic product. Exosomes (EXOs), a stem cell product associated with MSC therapy, have shown promising efficacy and safety in humans. MSC-EXO therapeutics represent a promising next-generation approach for treating radiation diseases involving a primary (major) inflammatory component. Provided that conditions for MSC-EXO production and bio-banking are agreed in the near future, the transition to industrial production of MSC-EXOs will be possible, and this is required to initiate well-controlled clinical trials for approval by the European Medicines Agency (EMA) and US Food and Drug Administration (FDA).
Collapse
Affiliation(s)
- M Benderitter
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Pôle Santé, 31 Avenue de la Division Leclerc, BP17, Fontenay-aux-Roses 92262, France
| | - E Herrera-Reyes
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Pôle Santé, 31 Avenue de la Division Leclerc, BP17, Fontenay-aux-Roses 92262, France
| | - R Tamarat
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Pôle Santé, 31 Avenue de la Division Leclerc, BP17, Fontenay-aux-Roses 92262, France
| |
Collapse
|
12
|
Inci F. Benchmarking a Microfluidic-Based Filtration for Isolating Biological Particles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:1897-1909. [PMID: 35041413 DOI: 10.1021/acs.langmuir.1c03119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Isolating particles from complex fluids is a crucial approach in multiple fields including biomedicine. In particular, biological matrices contain a myriad of distinct particles with different sizes and structures. Extracellular vesicles (EVs), for instance, are nanosized particles carrying vital information from donor to recipient cells, and they have garnered significant impact on disease diagnostics, drug delivery, and theranostics applications. Among all the EV types, exosome particles are one of the smallest entities, sizing from 30 to 100 nm. Separating such small substances from a complex media such as tissue culture and serum is still one of the most challenging steps in this field. Membrane filtration is one of the convenient approaches for these operations; yet clogging, low-recovery, and high fouling are still major obstacles. In this study, we design a two-filter-integrated microfluidic device focusing on dead-end and cross-flow processes at the same time, thereby minimizing any interfering factors on the recovery. The design of this platform is also numerically assessed to understand pressure-drop and flow rate effects over the procedure. As a model, we isolate exosome particles from human embryonic kidney cells cultured in different conditions, which also mimic complex fluids such as serum. Moreover, by altering the flow direction, we refresh the membranes for minimizing clogging issues and benchmark the platform performance for multitime use. By comprehensively analyzing the design and operation parameters of this platform, we address the aforementioned existing barriers in the recovery, clogging, and fouling factors, thereby achieving the use of a microfluidic device multiple times for bio-nanoparticle isolation without any notable issues.
Collapse
Affiliation(s)
- Fatih Inci
- UNAM - National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey
- Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| |
Collapse
|
13
|
Bernardino RMM, Leão R, Henrique R, Pinheiro LC, Kumar P, Suravajhala P, Beck HC, Carvalho AS, Matthiesen R. Extracellular Vesicle Proteome in Prostate Cancer: A Comparative Analysis of Mass Spectrometry Studies. Int J Mol Sci 2021; 22:ijms222413605. [PMID: 34948404 PMCID: PMC8707426 DOI: 10.3390/ijms222413605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 12/24/2022] Open
Abstract
Molecular diagnostics based on discovery research holds the promise of improving screening methods for prostate cancer (PCa). Furthermore, the congregated information prompts the question whether the urinary extracellular vesicles (uEV) proteome has been thoroughly explored, especially at the proteome level. In fact, most extracellular vesicles (EV) based biomarker studies have mainly targeted plasma or serum. Therefore, in this study, we aim to inquire about possible strategies for urinary biomarker discovery particularly focused on the proteome of urine EVs. Proteomics data deposited in the PRIDE archive were reanalyzed to target identifications of potential PCa markers. Network analysis of the markers proposed by different prostate cancer studies revealed moderate overlap. The recent throughput improvements in mass spectrometry together with the network analysis performed in this study, suggest that a larger standardized cohort may provide potential biomarkers that are able to fully characterize the heterogeneity of PCa. According to our analysis PCa studies based on urinary EV proteome presents higher protein coverage compared to plasma, plasma EV, and voided urine proteome. This together with a direct interaction of the prostate gland and urethra makes uEVs an attractive option for protein biomarker studies. In addition, urinary proteome based PCa studies must also evaluate samples from bladder and renal cancers to assess specificity for PCa.
Collapse
Affiliation(s)
- Rui Miguel Marques Bernardino
- Computational and Experimental Biology Group, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal;
- Urology Department, Centro Hospitalar e Universitário de Lisboa Central, 1169-050 Lisbon, Portugal;
- Correspondence: (R.M.M.B.); (R.M.); Tel.: +351-939218696 (R.M.M.B. & R.M.)
| | - Ricardo Leão
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal;
| | - Rui Henrique
- Pathology Department, Instituto Português de Oncologia, 4200-072 Porto, Portugal;
| | - Luis Campos Pinheiro
- Urology Department, Centro Hospitalar e Universitário de Lisboa Central, 1169-050 Lisbon, Portugal;
| | - Prashant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India;
- Somaiya Institute of Research and Consultancy (SIRAC), Somaiya Vidyavihar University (SVU), Vidyavihar, Mumbai 400077, India
| | - Prashanth Suravajhala
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri Campus, Clappana P.O., Kollam 690525, India;
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark;
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal;
| | - Rune Matthiesen
- Computational and Experimental Biology Group, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal;
- Correspondence: (R.M.M.B.); (R.M.); Tel.: +351-939218696 (R.M.M.B. & R.M.)
| |
Collapse
|
14
|
Bahram Sangani N, Gomes AR, Curfs LMG, Reutelingsperger CP. The role of Extracellular Vesicles during CNS development. Prog Neurobiol 2021; 205:102124. [PMID: 34314775 DOI: 10.1016/j.pneurobio.2021.102124] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/16/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022]
Abstract
With a diverse set of neuronal and glial cell populations, Central Nervous System (CNS) has one of the most complex structures in the body. Intercellular communication is therefore highly important to coordinate cell-to-cell interactions. Besides electrical and chemical messengers, CNS cells also benefit from another communication route, what is known as extracellular vesicles, to harmonize their interactions. Extracellular Vesicles (EVs) and their subtype exosomes are membranous particles secreted by cells and contain information packaged in the form of biomolecules such as small fragments of DNA, lipids, miRNAs, mRNAs, and proteins. They are able to efficiently drive changes upon their arrival to recipient cells. EVs actively participate in all stages of CNS development by stimulating neural cell proliferation, differentiation, synaptic formation, and mediating reciprocal interactions between neurons and oligodendrocyte for myelination process. The aim of the present review is to enlighten the presence and contribution of EVs at each CNS developmental milestone.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; GKC-Rett Expertise Centre, Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - Ana Rita Gomes
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal; Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Portugal.
| | - Leopold M G Curfs
- GKC-Rett Expertise Centre, Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - Chris P Reutelingsperger
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; GKC-Rett Expertise Centre, Maastricht University Medical Centre, Maastricht, the Netherlands.
| |
Collapse
|
15
|
Annaratone L, De Palma G, Bonizzi G, Sapino A, Botti G, Berrino E, Mannelli C, Arcella P, Di Martino S, Steffan A, Daidone MG, Canzonieri V, Parodi B, Paradiso AV, Barberis M, Marchiò C. Basic principles of biobanking: from biological samples to precision medicine for patients. Virchows Arch 2021; 479:233-246. [PMID: 34255145 PMCID: PMC8275637 DOI: 10.1007/s00428-021-03151-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/24/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
The term "biobanking" is often misapplied to any collection of human biological materials (biospecimens) regardless of requirements related to ethical and legal issues or the standardization of different processes involved in tissue collection. A proper definition of biobanks is large collections of biospecimens linked to relevant personal and health information (health records, family history, lifestyle, genetic information) that are held predominantly for use in health and medical research. In addition, the International Organization for Standardization, in illustrating the requirements for biobanking (ISO 20387:2018), stresses the concept of biobanks being legal entities driving the process of acquisition and storage together with some or all of the activities related to collection, preparation, preservation, testing, analysing and distributing defined biological material as well as related information and data. In this review article, we aim to discuss the basic principles of biobanking, spanning from definitions to classification systems, standardization processes and documents, sustainability and ethical and legal requirements. We also deal with emerging specimens that are currently being generated and shaping the so-called next-generation biobanking, and we provide pragmatic examples of cancer-associated biobanking by discussing the process behind the construction of a biobank and the infrastructures supporting the implementation of biobanking in scientific research.
Collapse
Affiliation(s)
- Laura Annaratone
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giuseppe De Palma
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Giuseppina Bonizzi
- Unit of Histopathology and Molecular Diagnostics, Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | - Gerardo Botti
- Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Naples, Italy
| | - Enrico Berrino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Pamela Arcella
- Department of Oncology, University of Turin, Turin, Italy
| | - Simona Di Martino
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy
| | | | - Vincenzo Canzonieri
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.,Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy
| | | | - Angelo Virgilio Paradiso
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Massimo Barberis
- Unit of Histopathology and Molecular Diagnostics, Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy. .,Department of Medical Sciences, University of Turin, Turin, Italy.
| | | |
Collapse
|
16
|
A predictive biomarker panel for bone metastases: Liquid biopsy approach. J Bone Oncol 2021; 29:100374. [PMID: 34189028 PMCID: PMC8220227 DOI: 10.1016/j.jbo.2021.100374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 01/12/2023] Open
Abstract
Data mining of published microarray datasets directed us to the identification of a multi gene panel involving of 15 genes that are particular to bone metastases. Serum exosomal markers HSP90AA1, SPP1, IL3, and PTK2 found in the present study might be useful in detecting the early spread of bone metastases leading to better clinical outcomes. This multi-gene panel and their related pathways may assist as promising conclusion predictors using novel approaches of exosome as liquid biopsy and their application in therapeutic targets in breast and lung cancer patients with bone metastases.
Bone metastases is one of the common metastatic site and leading cause of cancer-related mortality in progressive cancer patients. The purpose of the present study is to establish a liquid biopsy based multi-gene classifier and associated signalling pathways for early diagnosis of bone metastases. We used publically available microarray datasets and analysed them in a platform/chip-specific manner using GeneSpring software. Analyses of gene expression datasets identified 15 consistently over-expressed genes with statistical significance. Further, expression profile of same set of 15 genes were compared in breast and lung cancer exosome derived mRNA with (n = 10) and without (n = 10) bone metastases against healthy controls. ROC curve analysis performed individually for all the 15 genes shortlisted the 5 most relevant genes with significant sensitivity and specificity in both cancers. This liquid biopsy-based bone metastases predictor using multi-gene panel is a unique approach with potential clinical applications for effective management of aggressive cancers.
Collapse
|
17
|
Perocheau D, Touramanidou L, Gurung S, Gissen P, Baruteau J. Clinical applications for exosomes: Are we there yet? Br J Pharmacol 2021; 178:2375-2392. [PMID: 33751579 PMCID: PMC8432553 DOI: 10.1111/bph.15432] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/18/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a subset of extracellular vesicles essential for cell-cell communication in health and disease with the ability to transport nucleic acids, functional proteins and other metabolites. Their clinical use as diagnostic biomarkers and therapeutic carriers has become a major field of research over recent years, generating rapidly expanding scientific interest and financial investment. Their reduced immunogenicity compared to liposomes or viral vectors and their ability to cross major physiological barriers like the blood-brain barrier make them an appealing and innovative option as biomarkers and therapeutic agents. Here, we review the latest clinical developments of exosome biotechnology for diagnostic and therapeutic purposes, including the most recent COVID-19-related exosome-based clinical trials. We present current exosome engineering strategies for optimal clinical safety and efficacy, and assess the technology developed for good manufacturing practice compliant scaling up and storage approaches along with their limitations in pharmaceutical industry.
Collapse
Affiliation(s)
- Dany Perocheau
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Loukia Touramanidou
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sonam Gurung
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paul Gissen
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK.,Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Julien Baruteau
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK.,Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
18
|
Bagheri Hashkavayi A, Cha BS, Lee ES, Kim S, Park KS. Advances in Exosome Analysis Methods with an Emphasis on Electrochemistry. Anal Chem 2020; 92:12733-12740. [PMID: 32902258 DOI: 10.1021/acs.analchem.0c02745] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exosomes, small extracellular vesicles, are released by various cell types. They are found in bodily fluids, including blood, urine, serum, and saliva, and play essential roles in intercellular communication. Exosomes contain various biomarkers, such as nucleic acids and proteins, that reflect the status of their parent cells. Since they influence tumorigenesis and metastasis in cancer patients, exosomes are excellent noninvasive potential indicators for early cancer detection. Aptamers with specific binding properties have distinct advantages over antibodies, making them effective versatile bioreceptors for the detection of exosome biomarkers. Here, we review various aptamer-based exosome detection approaches based on signaling methods, such as fluorescence, colorimetry, and chemiluminescence, focusing on electrochemical strategies that are easier, cost-effective, and more sensitive than others. Further, we discuss the clinical applications of electrochemical exosome analysis strategies as well as future research directions in this field.
Collapse
Affiliation(s)
- Ayemeh Bagheri Hashkavayi
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Byung Seok Cha
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Eun Sung Lee
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Seokjoon Kim
- 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
|
19
|
Wei P, Wu F, Kang B, Sun X, Heskia F, Pachot A, Liang J, Li D. Plasma extracellular vesicles detected by Single Molecule array technology as a liquid biopsy for colorectal cancer. J Extracell Vesicles 2020; 9:1809765. [PMID: 32944195 PMCID: PMC7480466 DOI: 10.1080/20013078.2020.1809765] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Circulating extracellular vesicles (EVs) were recognized as a promising source of diagnostic biomarker. However, there are limited studies published in this area, partly due to the limited number of detection platforms capable of detecting extracellular vesicles. In this study, extracellular vesicle immunoassays were developed using the Single Molecule array technology (SiMoa) and their clinical applications to cancer diagnosis were evaluated. Two extracellular vesicle detection assays, CD9-CD63 and Epcam-CD63, were designed to detect universal extracellular vesicles and tumour-derived extracellular vesicles, respectively. Our results show that CD9-CD63 and Epcam-CD63 SiMoa assays specifically detect extracellular vesicles but not free proteins with high sensitivities. The Epcam-CD63 levels detected in cancer cell culture media were consistent with levels of Epcam-expressing EVs isolated from the same cancer cell lines and detected by Western blot. Furthermore, the assays distinguish cancerous from non-cancerous plasma samples. The highest CD9-CD63 and Epcam-CD63 signals were observed in colorectal cancer patients comparing to healthy and benign controls. Both assays showed superior diagnostic performance for colorectal cancer. In addition, our results show that CD9-CD63 detection is an independent prognosis factor for both progression free survival and overall survival, while Epcam-CD63 detectionis an independent prognosis factor for OS.
Collapse
Affiliation(s)
- Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fei Wu
- Fudan University Shanghai Cancer Center - InstitutMérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Open Innovation & Partnerships Department, bioMerieux (Shanghai) Company Limited, Shanghai, China
| | - Bin Kang
- Fudan University Shanghai Cancer Center - InstitutMérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Open Innovation & Partnerships Department, bioMerieux (Shanghai) Company Limited, Shanghai, China
| | - Xiaohua Sun
- Fudan University Shanghai Cancer Center - InstitutMérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Open Innovation & Partnerships Department, bioMerieux (Shanghai) Company Limited, Shanghai, China
| | - Fabienne Heskia
- Global Medical Affairs Deptartment, bioMérieux SA, Marcy l'Etoile, France
| | - Alexandre Pachot
- Open Innovation & Partnerships Department, bioMérieux SA, Marcy l'Etoile, France
| | - Ji Liang
- Fudan University Shanghai Cancer Center - InstitutMérieux Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Open Innovation & Partnerships Department, bioMerieux (Shanghai) Company Limited, Shanghai, China
| | - Dawei Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
20
|
Royo F, Théry C, Falcón-Pérez JM, Nieuwland R, Witwer KW. Methods for Separation and Characterization of Extracellular Vesicles: Results of a Worldwide Survey Performed by the ISEV Rigor and Standardization Subcommittee. Cells 2020; 9:cells9091955. [PMID: 32854228 PMCID: PMC7563174 DOI: 10.3390/cells9091955] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/23/2020] [Indexed: 02/05/2023] Open
Abstract
Research on extracellular vesicles (EVs) is growing exponentially due to an increasing appreciation of EVs as disease biomarkers and therapeutics, an expanding number of EV-containing materials under study, and application of new preparation, detection, and cargo analysis methods. Diversity of both sources and methodologies imposes challenges on the comparison of measurement results between studies and laboratories. While reference guidelines and minimal requirements for EV research have achieved the important objective of assembling community consensus, it is also essential to understand which methodologies and quality controls are currently being applied, and how usage trends are evolving. As an initial response to this need, the International Society for Extracellular Vesicles (ISEV) performed a worldwide survey in 2015 on "Techniques used for the isolation and characterization of extracellular vesicles" and published the results from this survey in 2016. In 2019, a new survey was performed to assess the changing state of the field. The questionnaire received more than 600 full or partial responses, and the present manuscript summarizes the results of this second worldwide survey. The results emphasize that separation methods such as ultracentrifugation and density gradients are still the most commonly used methods, the use of size exclusion chromatography has increased, and techniques based on tangential flow and microfluidics are now being used by more than 10% of respondents. The survey also reveals that most EV researchers still do not perform sample quality controls before or after isolation of EVs. Finally, the majority of EV researchers emphasize that separation and characterization of EVs should receive more attention.
Collapse
Affiliation(s)
- Felix Royo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160 Derio, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Clotilde Théry
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France;
| | - Juan M. Falcón-Pérez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160 Derio, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
- Correspondence: (J.M.F.-P.); (R.N.); (K.W.W.)
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, Amsterdam UMC, Location AMC, University of Amsterdam, 19268 Amsterdam, The Netherlands
- Vesicle Observation Centre, Amsterdam UMC, Location AMC, University of Amsterdam, 19268 Amsterdam, The Netherlands
- Correspondence: (J.M.F.-P.); (R.N.); (K.W.W.)
| | - Kenneth W. Witwer
- School of Medicine, Departments of Molecular and Comparative Pathobiology and Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Correspondence: (J.M.F.-P.); (R.N.); (K.W.W.)
| |
Collapse
|
21
|
Azam Z, Quillien V, Wang G, To SST. The potential diagnostic and prognostic role of extracellular vesicles in glioma: current status and future perspectives. Acta Oncol 2019; 58:353-362. [PMID: 30632857 DOI: 10.1080/0284186x.2018.1551621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lack of appropriate diagnostic/prognostic tools for glioblastoma (GB) is considered one of the major setbacks in the early diagnosis and treatment of this deadly brain tumor. The current gold standard for its diagnosis and staging still relies on invasive biopsy followed by histological examination as well as molecular profiling. Nevertheless, noninvasive approaches are being explored and one example is through the investigation of extracellular vesicles (EVs) in the biofluids of GB patients. EVs are known to carry molecular cargoes such as DNA, mRNA, miRNA, proteins and lipids in almost every type of body fluids. Thus, molecular signature of GB may be present in the EVs derived from these patients. This review focuses on the diagnostic/prognostic potential of EVs in GB, through presenting recent studies on (i) molecular components of EVs, (ii) links between EVs and GB tumor microenvironment, and (iii) clinical potential of EV biomarkers, together with the technical shortcomings researchers need to consider for future studies.
Collapse
Affiliation(s)
- Zulfikar Azam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Véronique Quillien
- Department of Biology, Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Shing-Shun Tony To
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
22
|
Noorwali A, Faidah M, Ahmed N, Bima A. Tracking iron oxide labelled mesenchymal stem cells(MSCs) using magnetic resonance imaging (MRI) in a rat model of hepatic cirrhosis. Bioinformation 2019; 15:1-10. [PMID: 31359992 PMCID: PMC6651036 DOI: 10.6026/97320630015001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/24/2018] [Indexed: 01/07/2023] Open
Abstract
Homing and tumor attenuation potential of BM-MSCs labelled with superparamagnetic iron-oxide nanoparticles (SPIONs) in a rat model of hepatic cirrhosis was evaluated. Rat BM-MSCs were derived, characterized and labelled with SPIONs (200 nm; 25 mg Fe/ml). Hepatic cirrhosis was induced in Wistar rats (n=30; 10/group) with carbon tetrachloride (CCl4; 0.3 mL/kg body weight) injected twice a week for 12 weeks. Group-I was administered vehicle (castor-oil) alone; Group-II received two doses of unlabelled BM-MSCs (3x106 cells) and Group-III received two doses of SPIONs labelled BM-MSCs (3x106 cells) via tail vein injection (0.5 ml) at weekly intervals. All animals were sacrificed after two weeks for histological, radiological and biochemical analysis. Derived BM-MSCs demonstrated MSCs related CD markers. Histology confirmed induction of hepatic cirrhosis with CCL4. Levels of alanine-aminotransferase, aspartate-aminotransferase,alkaline-phosphatase and gamma glutamyl-transferase returned to normal levels following treatment with BM-MSCs. Uptake and homing of SPIONs labelled BM-MSCs, and reduction in the size of cirrhotic nodules were confirmed using transmission electron microscopy and magnetic resonance imaging respectively. BM-MSCs reduced the pathological effects of CCL4 induced hepatic cirrhosis and labelling BMMSCs with SPIONs were non-toxic and enabled efficient tracking using non-invasive methods.
Collapse
Affiliation(s)
- Abdulwahab Noorwali
- Stem Cell Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mamdooh Faidah
- Department of Medical Laboratory,College of Health Sciences,King Abdulaziz University,Jeddah 21589 Saudi Arabia
| | - Naushad Ahmed
- Department of Radiology,King Abdulaziz University Hospital,King Abdulaziz University,Jeddah 21589, Saudi Arabia
| | - Abdulhadi Bima
- Department of Clinical Biochemistry,King Abdulaziz University Hospital,King Abdulaziz University,Jeddah 21 89,Saudi Arabia
| |
Collapse
|
23
|
Shi M, Sheng L, Stewart T, Zabetian CP, Zhang J. New windows into the brain: Central nervous system-derived extracellular vesicles in blood. Prog Neurobiol 2019; 175:96-106. [PMID: 30685501 DOI: 10.1016/j.pneurobio.2019.01.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/18/2018] [Accepted: 01/23/2019] [Indexed: 12/28/2022]
Abstract
Extracellular vesicles (EVs), including exosomes and (shedding) microvesicles, are released by nearly all cell types and carry a cargo of proteins and nucleic acids that varies by the cell of origin. They are thought to play critical roles in normal central nervous system (CNS) function and neurological disorders. A recently revealed key characteristic of EVs is that they may travel between the CNS and peripheral circulation. This property has led to intense interest in how EVs might serve as a vehicle for toxic protein clearance and as a readily accessible source of biomarkers for CNS disorders. Furthermore, by bypassing the blood-brain barrier, modified EVs could serve as a unique drug delivery system that targets specific neuronal populations. Further work is necessary to develop and optimize techniques that enable high-yield capture of relevant EV populations, analyze individual EVs and their cargos, and validate preliminary results of EV-derived biomarkers in independent cohorts.
Collapse
Affiliation(s)
- Min Shi
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Lifu Sheng
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Tessandra Stewart
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Cyrus P Zabetian
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Parkinson's Disease Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jing Zhang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98104, USA; Beijing Key Laboratory of Research and Transformation on Neurodegenerative Diseases Biomarkers, Department of Pathology, Peking University Third Hospital/Institute of Basic Science, Peking University Health Science Center, Beijing 100083, China.
| |
Collapse
|
24
|
Venturella M, Carpi FM, Zocco D. Standardization of Blood Collection and Processing for the Diagnostic Use of Extracellular Vesicles. CURRENT PATHOBIOLOGY REPORTS 2019. [DOI: 10.1007/s40139-019-00189-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
25
|
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: 11.3] [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
|
26
|
Lane RE, Korbie D, Hill MM, Trau M. Extracellular vesicles as circulating cancer biomarkers: opportunities and challenges. Clin Transl Med 2018; 7:14. [PMID: 29855735 PMCID: PMC5981152 DOI: 10.1186/s40169-018-0192-7] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are small, lipid-bound particles containing nucleic acid and protein cargo which are excreted from cells under a variety of normal and pathological conditions. EVs have garnered substantial research interest in recent years, due to their potential utility as circulating biomarkers for a variety of diseases, including numerous types of cancer. The following review will discuss the current understanding of the form and function of EVs, their specific role in cancer pathogenesis and their potential for non-invasive disease diagnosis and/or monitoring. This review will also highlight several key issues for this field, including the importance of implementing robust and reproducible sample handling protocols, and the challenge of extracting an EV-specific biomarker signal from a complex biological background.
Collapse
Affiliation(s)
- R E Lane
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - D Korbie
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - M M Hill
- The University of Queensland Diamantina Institute, Faculty of Medicine, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.,QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - M Trau
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia. .,School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
27
|
Daughton CG. Monitoring wastewater for assessing community health: Sewage Chemical-Information Mining (SCIM). THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 619-620:748-764. [PMID: 29161600 PMCID: PMC6091531 DOI: 10.1016/j.scitotenv.2017.11.102] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/08/2017] [Accepted: 11/08/2017] [Indexed: 04/13/2023]
Abstract
Timely assessment of the aggregate health of small-area human populations is essential for guiding the optimal investment of resources needed for preventing, avoiding, controlling, or mitigating human exposure risks, as well as for maintaining or promoting health. Seeking those interventions yielding the greatest benefit with respect to the allocation of resources is critical for making progress toward community sustainability, reducing health disparities, promoting social justice, and maintaining or improving collective health and well-being. More informative, faster, and less-costly approaches are needed for guiding investigation of cause-effect linkages involving communities and stressors originating from both the built and natural environments. One such emerging approach involves the continuous monitoring of sewage for chemicals that serve as indicators of the collective status of human health (or stress/disease) or any other facet relevant to gauging time-trends in community-wide health. This nascent approach can be referred to as Sewage Chemical-Information Mining (SCIM) and involves the monitoring of sewage for the information that resides in the form of natural and anthropogenic chemicals that enter sewers as a result of the everyday actions, activities, and behaviors of humans. Of particular interest is a specific embodiment of SCIM that would entail the targeted monitoring of a broad suite of endogenous biomarkers of key physiologic processes (as opposed to xenobiotics or their metabolites). This application is termed BioSCIM-an approach roughly analogous to a hypothetical community-wide collective clinical urinalysis, or to a hypothetical en masse human biomonitoring program. BioSCIM would be used for gauging the status or time-trends in community-wide health on a continuous basis. This paper presents an update on the progress made with the development of the BioSCIM concept in the period of time since its original publication in 2012, as well as the next steps required for its continued development.
Collapse
Affiliation(s)
- Christian G Daughton
- Environmental Futures Analysis Branch, Systems Exposure Division, National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, 944 East Harmon Avenue, Las Vegas 89119, NV, USA.
| |
Collapse
|
28
|
Abstract
Historically, small molecules, including steroid hormones and cytokines, have been attributed a role in paracrine and endocrine signaling, and now include a new player: biological nanoparticles, or 'exosomes'. Generated intracellularly, and defined simply as nanoparticulate packages of signaling moieties, exosomes have emerged as vehicles for highly specialized local and distant intercellular communication. Exosomes are increasingly being recognized as contributing factors in many diseases, and their potential as biomarkers and in therapeutics is rapidly emerging. This review highlights recent advances in the exploitation of exosomes in diagnostic and therapeutic applications. We discuss various facets of nanoparticles, namely the isolation and manipulation of exosomes, the construction of synthetic exosome-like particles in vivo, and their potential use in the treatment of various diseases.
Collapse
|
29
|
Ramirez MI, Amorim MG, Gadelha C, Milic I, Welsh JA, Freitas VM, Nawaz M, Akbar N, Couch Y, Makin L, Cooke F, Vettore AL, Batista PX, Freezor R, Pezuk JA, Rosa-Fernandes L, Carreira ACO, Devitt A, Jacobs L, Silva IT, Coakley G, Nunes DN, Carter D, Palmisano G, Dias-Neto E. Technical challenges of working with extracellular vesicles. NANOSCALE 2018; 10:881-906. [PMID: 29265147 DOI: 10.1039/c7nr08360b] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular Vesicles (EVs) are gaining interest as central players in liquid biopsies, with potential applications in diagnosis, prognosis and therapeutic guidance in most pathological conditions. These nanosized particles transmit signals determined by their protein, lipid, nucleic acid and sugar content, and the unique molecular pattern of EVs dictates the type of signal to be transmitted to recipient cells. However, their small sizes and the limited quantities that can usually be obtained from patient-derived samples pose a number of challenges to their isolation, study and characterization. These challenges and some possible options to overcome them are discussed in this review.
Collapse
Affiliation(s)
- Marcel I Ramirez
- Fundação Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil and Universidade Federal do Paraná, Curitiba, PR, Brazil
| | | | - Catarina Gadelha
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ivana Milic
- School of Life and Health Sciences, Aston University, England, UK
| | | | | | - Muhammad Nawaz
- Universidade de São Paulo, São Paulo, SP, Brazil and University of Gothenburg, Sweden
| | - Naveed Akbar
- Division of Cardiovascular Medicine, University of Oxford, Oxford, England, UK
| | - Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, University of Oxford, Oxford, England, UK
| | - Laura Makin
- Sir William Dunn School of Pathology, University of Oxford, Oxford, England, UK
| | - Fiona Cooke
- University of St Andrews, St Andrews, Fife, Scotland, UK
| | - Andre L Vettore
- Federal University of São Paulo campus Diadema, Diadema, Brazil
| | | | | | - Julia A Pezuk
- Universidade Anhanguera de São Paulo, São Paulo, Brazil
| | - Lívia Rosa-Fernandes
- Universidade de São Paulo, São Paulo, SP, Brazil and University of Southern Denmark, Odense, Denmark
| | | | - Andrew Devitt
- School of Life and Health Sciences, Aston University, England, UK
| | | | | | - Gillian Coakley
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Diana N Nunes
- CIPE, A.C.Camargo Cancer Center, São Paulo, SP, Brazil.
| | - Dave Carter
- Oxford Brookes University, Oxford, England, UK
| | - Giuseppe Palmisano
- Universidade de São Paulo, São Paulo, SP, Brazil and IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Emmanuel Dias-Neto
- CIPE, A.C.Camargo Cancer Center, São Paulo, SP, Brazil. and Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
30
|
Cui S, Cheng Z, Qin W, Jiang L. Exosomes as a liquid biopsy for lung cancer. Lung Cancer 2017; 116:46-54. [PMID: 29413050 DOI: 10.1016/j.lungcan.2017.12.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/04/2017] [Accepted: 12/17/2017] [Indexed: 12/16/2022]
Abstract
In lung cancer and other malignancies, the so-called "liquid biopsy" is quickly moving into clinical practice. Its full potential has not yet been fully identified, but the "liquid biopsy" is no longer a promise but has become a reality that allows for better treatment selection and monitoring of lung cancer. This emerging field has significant potential to make up for the limitations of the traditional tissue-derived biomaterials. Exosomes are spherical nano-sized vesicles with a diameter of 40-100 nm and a density of 1.13-1.19 g/ml. In both physiological and pathological conditions, exosomes can be released by different cell types, including immune cells, stem cells and tumor cells. These small molecules may serve as promising biomarkers in lung cancer "liquid biopsy" as they can be easily obtained from most body fluids. In addition, the lipid bilayer of exosomes allows for stable cargoes which are relatively hard to degrade. Furthermore, the composition of exosomes reflects that of their parental cells, suggesting that exosomes are potential surrogates of the original cells and, therefore, are useful for understanding cell biology. Previous studies have demonstrated that exosomes play important roles in cell-to-cell communication. Moreover, tumor-derived exosomes are evolved in tumor-specific biological process, including tumor proliferation and progression. Recently, a growing number of studies has focused on exosomal cargo and their use in lung cancer genesis and progression. In addition, their utility as lung cancer diagnostic, prognostic and predictive biomarkers have also been studied. The current review primarily summaries lung cancer-related exosomal biomarkers that have recently been identified and discusses their potential in clinical practice.
Collapse
Affiliation(s)
- Shaohua Cui
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Zhuoan Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China.
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, China.
| |
Collapse
|
31
|
Lim YJ, Lee SJ. Are exosomes the vehicle for protein aggregate propagation in neurodegenerative diseases? Acta Neuropathol Commun 2017; 5:64. [PMID: 28851422 PMCID: PMC5576311 DOI: 10.1186/s40478-017-0467-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 02/08/2023] Open
Abstract
Abnormal protein aggregation has been implicated in neurodegenerative processes in human neurological disorders, such as Alzheimer’s disease and Parkinson’s disease. Recently, studies have established a novel concept that protein aggregates are transmitted among neuronal cells. By extension, such interneuronal aggregate transmission has been hypothesized to be the underlying mechanism for the pathological and clinical disease progression. However, the precise mechanism of the interneuronal aggregate transmission remains ill-defined. Recent reports have suggested that exosomes, a specific group of extracellular vesicles that are involved in intercellular transfer of cellular macromolecules such as proteins and RNAs, could play an important role in the aggregate transmission among neurons. Here, we review various types of extracellular vesicles and critically evaluate the evidence supporting the role of exosomes in interneuronal aggregate transmission and neurodegeneration. We also discuss the competing mechanisms other than the exosome-mediated transmission. By doing so, we aim to assess the current state of knowledge on the mechanism of interneuronal aggregate transmission and suggest the future directions of research towards understanding the mechanism.
Collapse
|
32
|
Raik S, Kumar A, Bhattacharyya S. Insights into cell-free therapeutic approach: Role of stem cell "soup-ernatant". Biotechnol Appl Biochem 2017; 65:104-118. [PMID: 28321921 DOI: 10.1002/bab.1561] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/02/2017] [Indexed: 12/16/2022]
Abstract
Current advances in medicine have revolutionized the field of regenerative medicine dramatically with newly evolved therapies for repair or replacement of degenerating or injured tissues. Stem cells (SCs) can be harvested from different sources for clinical therapeutics, which include fetal tissues, umbilical cord blood, embryos, and adult tissues. SCs can be isolated and differentiated into desired lineages for tissue regeneration and cell replacement therapy. However, several loopholes need to be addressed properly before this can be extended for large-scale therapeutic application. These include a careful approach for patient safety during SC treatments and tolerance of recipients. SC treatments are associated with a number of risk factors and require successful integration and survival of transplanted cells in the desired microenvironment with concurrent tissue regeneration. Recent studies have focused on developing alternatives that can replace the cell-based therapy using paracrine factors. The development of stem "cell free" therapies can be devoted mainly to the use of soluble factors (secretome), extracellular vesicles, and mitochondrial transfer. The present review emphasizes on the paradigms related to the use of SC-based therapeutics and the potential applications of a cell-free approach as an alternative to cell-based therapy in the area of regenerative medicine.
Collapse
Affiliation(s)
- Shalini Raik
- Department of Biophysics, PGIMER, Chandigarh, India
| | - Ajay Kumar
- Department of Biophysics, PGIMER, Chandigarh, India
| | | |
Collapse
|
33
|
Foster BP, Balassa T, Benen TD, Dominovic M, Elmadjian GK, Florova V, Fransolet MD, Kestlerova A, Kmiecik G, Kostadinova IA, Kyvelidou C, Meggyes M, Mincheva MN, Moro L, Pastuschek J, Spoldi V, Wandernoth P, Weber M, Toth B, Markert UR. Extracellular vesicles in blood, milk and body fluids of the female and male urogenital tract and with special regard to reproduction. Crit Rev Clin Lab Sci 2016; 53:379-95. [PMID: 27191915 DOI: 10.1080/10408363.2016.1190682] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are released from almost all cells and tissues. They are able to transport substances (e.g. proteins, RNA or DNA) at higher concentrations than in their environment and may adhere in a receptor-controlled manner to specific cells or tissues in order to release their content into the respective target structure. Blood contains high concentrations of EVs mainly derived from platelets, and, at a smaller amount, from erythrocytes. The female and male reproductive tracts produce EVs which may be associated with fertility or infertility and are released into body fluids and mucosas of the urogenital organs. In this review, the currently relevant detection methods are presented and critically compared. During pregnancy, placenta-derived EVs are dynamically detectable in peripheral blood with changing profiles depending upon progress of pregnancy and different pregnancy-associated pathologies, such as preeclampsia. EVs offer novel non-invasive diagnostic tools which may reflect the situation of the placenta and the foetus. EVs in urine have the potential of reflecting urogenital diseases including cancers of the neighbouring organs. Several methods for detection, quantification and phenotyping of EVs have been established, which include electron microscopy, flow cytometry, ELISA-like methods, Western blotting and analyses based on Brownian motion. This review article summarises the current knowledge about EVs in blood and cord blood, in the different compartments of the male and female reproductive tracts, in trophoblast cells from normal and pre-eclamptic pregnancies, in placenta ex vivo perfusate, in the amniotic fluid, and in breast milk, as well as their potential effects on natural killer cells as possible targets.
Collapse
Affiliation(s)
- B P Foster
- a Maternal and Fetal Health Research Centre, School of Biomedicine, University of Manchester, and Manchester Academic Health Sciences Centre, University Research , Manchester , UK
| | - T Balassa
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - T D Benen
- c Microtrac GmbH , Krefeld , Germany
| | - M Dominovic
- d Department of Physiology and Immunology , Medical Faculty, University of Rijeka , Rijeka , Croatia
| | - G K Elmadjian
- e Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - V Florova
- f Department of Obstetrics , Gynecology and Perinatology, First Moscow State Medical University , Moscow , Russia
| | - M D Fransolet
- g Laboratory of Tumor and Development Biology , GIGA-R, University of Liège , Liège , Belgium
| | - A Kestlerova
- h Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine , Charles University Prague , Czech Republic
- i Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University , Prague , Czech Republic
| | - G Kmiecik
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - I A Kostadinova
- k Department of Immunoneuroendocrinology , Institute of Biology and Immunology of Reproduction , Sofia , Bulgaria
| | - C Kyvelidou
- l Department of Biology , University of Crete , Crete , Greece
| | - M Meggyes
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - M N Mincheva
- m Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - L Moro
- n ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic- Universitat de Barcelona , Barcelona , Spain
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - J Pastuschek
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - V Spoldi
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - P Wandernoth
- p Institute of Anatomy, University Hospital, University Duisburg-Essen , Essen , Germany
| | - M Weber
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - B Toth
- q Department of Gynecological Endocrinology and Fertility Disorders , Ruprecht-Karls University of Heidelberg , Heidelberg , Germany
| | - U R Markert
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| |
Collapse
|
34
|
Cretoiu D, Xu J, Xiao J, Cretoiu SM. Telocytes and Their Extracellular Vesicles-Evidence and Hypotheses. Int J Mol Sci 2016; 17:E1322. [PMID: 27529228 PMCID: PMC5000719 DOI: 10.3390/ijms17081322] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 12/18/2022] Open
Abstract
Entering the new millennium, nobody believed that there was the possibility of discovering a new cellular type. Nevertheless, telocytes (TCs) were described as a novel kind of interstitial cell. Ubiquitously distributed in the extracellular matrix of any tissue, TCs are regarded as cells with telopodes involved in intercellular communication by direct homo- and heterocellular junctions or by extracellular vesicle (EVs) release. Their discovery has aroused the interest of many research groups worldwide, and many researchers regard them as potentially regenerative cells. Given the experience of our laboratory, where these cells were first described, we review the evidence supporting the fact that TCs release EVs, and discuss alternative hypotheses about their future implications.
Collapse
Affiliation(s)
- Dragos Cretoiu
- Division of Cellular and Molecular Biology and Histology, Department of Morphological Sciences, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania.
- Victor Babeş National Institute of Pathology, Bucharest 050096, Romania.
| | - Jiahong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Sanda M Cretoiu
- Division of Cellular and Molecular Biology and Histology, Department of Morphological Sciences, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania.
- Victor Babeş National Institute of Pathology, Bucharest 050096, Romania.
| |
Collapse
|
35
|
Stremersch S, De Smedt SC, Raemdonck K. Therapeutic and diagnostic applications of extracellular vesicles. J Control Release 2016; 244:167-183. [PMID: 27491882 DOI: 10.1016/j.jconrel.2016.07.054] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/24/2016] [Accepted: 07/29/2016] [Indexed: 02/07/2023]
Abstract
During the past two decades, extracellular vesicles (EVs) have been identified as important mediators of intercellular communication, enabling the functional transfer of bioactive molecules from one cell to another. Consequently, it is becoming increasingly clear that these vesicles are involved in many (patho)physiological processes, providing opportunities for therapeutic applications. Moreover, it is known that the molecular composition of EVs reflects the physiological status of the producing cell and tissue, rationalizing their exploitation as biomarkers in various diseases. In this review the composition, biogenesis and diversity of EVs is discussed in a therapeutic and diagnostic context. We describe emerging therapeutic applications, including the use of EVs as drug delivery vehicles and as cell-free vaccines, and reflect on future challenges for clinical translation. Finally, we discuss the use of EVs as a biomarker source and highlight recent studies and clinical successes.
Collapse
Affiliation(s)
- Stephan Stremersch
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
36
|
He M, Zeng Y. Microfluidic Exosome Analysis toward Liquid Biopsy for Cancer. ACTA ACUST UNITED AC 2016; 21:599-608. [PMID: 27215792 DOI: 10.1177/2211068216651035] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Indexed: 12/20/2022]
Abstract
Assessment of a tumor's molecular makeup using biofluid samples, known as liquid biopsy, is a prominent research topic in precision medicine for cancer, due to its noninvasive property allowing repeat sampling for monitoring molecular changes of tumors over time. Circulating exosomes recently have been recognized as promising tumor surrogates because they deliver enriched biomarkers, such as proteins, RNAs, and DNA. However, purification and characterization of these exosomes are technically challenging. Microfluidic lab-on-a-chip technology effectively addresses these challenges owing to its inherent advantages in integration and automation of multiple functional modules, enhancing sensing performance, and expediting analysis processes. In this article, we review the state-of-the-art development of microfluidic technologies for exosome isolation and molecular characterization with emphasis on their applications toward liquid biopsy-based analysis of cancer. Finally, we share our perspectives on current challenges and future directions of microfluidic exosome analysis.
Collapse
Affiliation(s)
- Mei He
- Department of Biological and Agricultural Engineering, College of Engineering, Kansas State University, Manhattan, KS, USA
| | - Yong Zeng
- Department of Chemistry, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
37
|
Stem Cell Banking and Its Impact on Cardiac Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 951:163-178. [PMID: 27837563 DOI: 10.1007/978-3-319-45457-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cardiovascular diseases, including heart failure, are the most frequent cause of death annually, even higher than any other pathologies. Specifically, patients who suffer from myocardial infarction may encounter adverse remodeling processes of the heart that can ultimately lead to heart failure. Prognosis of patients affected by heart failure is very poor with 5-year mortality close to 50 %. Despite the impressive progress in the clinical treatment of heart failure in recent years, heart transplantation is still required to avoid death as the result of the inexorable decline in cardiac function. Unfortunately, the availability of donor human hearts for transplantation largely fails to cover the number of potential recipient requests. From this urgent unmet clinical need the interest in stem cell applications for heart regeneration made its start, and has rapidly grown in the last decades. Indeed, the discovery and application of stem and progenitor cells as therapeutic agents has raised substantial interest with the objective of reversing these processes, and ultimately inducing cardiac regeneration. In this scenario, the role of biobanking may play a remarkable role to provide cells at the right time according to the patient's clinical needs, mostly for autologous use in the acute setting of myocardial infarction, largely reducing the time needed for cell preparation and expansion before administration.
Collapse
|