951
|
Li YJ, Wu JY, Wang JM, Hu XB, Cai JX, Xiang DX. Gemcitabine loaded autologous exosomes for effective and safe chemotherapy of pancreatic cancer. Acta Biomater 2020; 101:519-530. [PMID: 31629893 DOI: 10.1016/j.actbio.2019.10.022] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/19/2019] [Accepted: 10/15/2019] [Indexed: 01/05/2023]
Abstract
Pancreatic cancer remains one of the most highly lethal diseases with very poor prognosis. Gemcitabine (GEM) is the first-line chemotherapeutic drug for pancreatic cancer treatment but is associated with significant side effects when administered systemically. Exosomes have emerged as attractive candidates for drug delivery for their high delivery efficiency and biocompatibility. Here, GEM was loaded into autologous exosomes to formulate ExoGEM for targeted chemotherapy of pancreatic cancer. Autologous exosomes facilitate cellular uptake of GEM and contributed to significantly increased cytotoxic effect of GEM, while heterologous cellular uptake showed less efficiency. Autologous exosomes showed targeting ability to pancreatic cancer in biodistribution study, and GEM concentration in tumor site was increased via ExoGEM delivery. ExoGEM treatment, in tumor-bearing mice, significantly suppressed tumor growth, with prolonged survival in a dose-response manner, but caused minimal damage to normal tissues. More importantly, tumors in several mice treated with ExoGEM were disappeared without recurrence. Autologous exosomes are safe and effective vehicles for targeted delivery of GEM against pancreatic cancer. This delivery strategy may have implications for personalized chemotherapy of pancreatic cancer. STATEMENT OF SIGNIFICANCE: Exosomes are efficient delivery vehicles in intracellular communication. Moreover, potential tropism of autologous exosomes to the tumor microenvironment make them competitive delivery vehicles. The use of cancer-derived exosomes for drug delivery and superior targeting efficacy and enhanced anticancer efficacy of therapeutics have been evidenced. Gemcitabine is a mainstay for pancreatic treatment. However, poor cellular uptake and low targeting effects of gemcitabine often lead to severe systemic toxicity. Therefore, to overcome this limitation, we herein loaded gemcitabine into autologous pancreatic cancer-derived exosomes for the targeted chemotherapy of pancreatic cancer.
Collapse
|
952
|
Sato Y, Kuwahara K, Mogami K, Takahashi K, Nishizawa S. Amphipathic helical peptide-based fluorogenic probes for a marker-free analysis of exosomes based on membrane-curvature sensing. RSC Adv 2020; 10:38323-38327. [PMID: 35517518 PMCID: PMC9057301 DOI: 10.1039/d0ra07763a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/11/2020] [Indexed: 01/08/2023] Open
Abstract
With increasing knowledge about the diverse roles of exosomes in the biological process, much attention has been paid to develop analytical methods for detection and quantification of exosomes. Immunoassays based on the recognition of exosomal protein markers by antibodies were widely used. However, considering that exosomal protein composition varies with the cell type, the protein markers should be carefully selected for a sensitive and selective analysis of target exosomes. Herein, we developed a new class of exosome-binding fluorogenic probes based on membrane curvature (MC) sensing of amphipathic helical (AH) peptides for exosome analysis without the need to use protein markers on the exosomal membranes. The C-terminal region of apolipoprotein A-I labeled with Nile red (ApoC-NR) exhibited a significant fluorescence enhancement upon selective binding to the highly curved membranes of synthetic vesicles. Circular dichroism (CD) measurements involving 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC)/1-2-dioleoyl-sn-glycerol (DOG) vesicles suggested that ApoC-NR recognizes the lipid packing defects in the surface of highly curved membranes via the hydrophobic insertion of the α-helix structure of the ApoC unit. ApoC-NR exhibited a stronger binding affinity for exosome-sized vesicles and a higher MC selectivity compared to all other previously reported peptide probes. ApoC-NR can be used in a simple and rapid “mix and read” analysis of various kinds of exosomes derived from different cell types (limit of detection: –105 particles/μL) without being influenced by the variation in the expression of the surface proteins of the exosomes, which stands in sharp contrast to immunoassays. Fluorogenic probes based on membrane curvature sensing-amphipathic helical peptides have been developed for a marker-free exosome analysis.![]()
Collapse
Affiliation(s)
- Yusuke Sato
- Department of Chemistry
- Graduate School of Science
- Tohoku University
- Sendai 980-8578
- Japan
| | - Kazuki Kuwahara
- Department of Chemistry
- Graduate School of Science
- Tohoku University
- Sendai 980-8578
- Japan
| | - Kenta Mogami
- Department of Chemistry
- Graduate School of Science
- Tohoku University
- Sendai 980-8578
- Japan
| | - Kenta Takahashi
- Department of Chemistry
- Graduate School of Science
- Tohoku University
- Sendai 980-8578
- Japan
| | - Seiichi Nishizawa
- Department of Chemistry
- Graduate School of Science
- Tohoku University
- Sendai 980-8578
- Japan
| |
Collapse
|
953
|
Tian J, Casella G, Zhang Y, Rostami A, Li X. Potential roles of extracellular vesicles in the pathophysiology, diagnosis, and treatment of autoimmune diseases. Int J Biol Sci 2020; 16:620-632. [PMID: 32025210 PMCID: PMC6990925 DOI: 10.7150/ijbs.39629] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022] Open
Abstract
Since extracellular vesicles (EVs) were discovered in 1983 in sheep reticulocytes samples, they have gradually attracted scientific attention and become a topic of great interest in the life sciences field. EVs are small membrane particles, released by virtually every cell that carries a variety of functional molecules. Their main function is to deliver messages to the surrounding area in both physiological and pathological conditions. Initially, they were thought to be either cell debris, signs of cell death, or unspecific structures. However, accumulating evidence support a theory that EVs are a universal mechanism of communication. Thanks to their biological characteristics and functions, EVs are likely to represent a promising strategy for obtaining pathogen information, identifying therapeutic targets and selecting specific biomarkers for a variety of diseases, such as autoimmune diseases. In this review, we provide a brief overview of recent progress in the study of the biology and functions of EVs. We also discuss their roles in diagnosis and therapy, with particular emphasis on autoimmune diseases.
Collapse
Affiliation(s)
- Jing Tian
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Giacomo Casella
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| |
Collapse
|
954
|
Božič D, Hočevar M, Kononenko V, Jeran M, Štibler U, Fiume I, Pajnič M, Pađen L, Kogej K, Drobne D, Iglič A, Pocsfalvi G, Kralj-Iglič V. Pursuing mechanisms of extracellular vesicle formation. Effects of sample processing. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2020. [DOI: 10.1016/bs.abl.2020.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
955
|
Luo L, Wang L, Zeng L, Wang Y, Weng Y, Liao Y, Chen T, Xia Y, Zhang J, Chen J. A ratiometric electrochemical DNA biosensor for detection of exosomal MicroRNA. Talanta 2020; 207:120298. [DOI: 10.1016/j.talanta.2019.120298] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/20/2019] [Accepted: 08/24/2019] [Indexed: 10/26/2022]
|
956
|
|
957
|
Nazarenko I. Extracellular Vesicles: Recent Developments in Technology and Perspectives for Cancer Liquid Biopsy. Recent Results Cancer Res 2020; 215:319-344. [PMID: 31605237 DOI: 10.1007/978-3-030-26439-0_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular micro- and nanoscale membrane vesicles produced by different cells progressively attract the attention of the scientific community. They function as mediators of intercellular communication and transport genetic material and signaling molecules between the cells. In the context of keeping homeostasis, the extracellular vesicles contribute to the regulation of various systemic and local processes. Vesicles released by the tumor and activated stromal cells exhibit multiple functions including support of tumor growth, preparation of the pre-metastatic niches, and immune suppression. Considerable progress has been made regarding the criteria of classification of the vesicles according to their origin, content, and function: Exosomes, microvesicles, also referred to as microparticles or ectosomes, and large oncosomes were defined as actively released vesicles. Additionally, apoptotic bodies represented by a highly heterogeneous population of particles produced during apoptosis, the programmed cell death, should be considered. Because the majority of isolation techniques do not allow the separation of different types of vesicles, a joined term "extracellular vesicles" (EVs) was recommended by the ISEV community for the definition of vesicles isolated from either the cell culture supernatants or the body fluids. Because EV content reflects the content of the cell of origin, multiple studies on EVs from body fluids in the context of cancer diagnosis, prediction, and prognosis were performed, actively supporting their high potential as a biomarker source. Here, we review the leading achievements in EV analysis from body fluids, defined as EV-based liquid biopsy, and provide an overview of the main EV constituents: EV surface proteins, intravesicular soluble proteins, EV RNA including mRNA and miRNA, and EV DNA as potential biomarkers. Furthermore, we discuss recent developments in technology for quantitative EV analysis in the clinical setting and future perspectives toward miniaturized high-precision liquid biopsy approaches.
Collapse
Affiliation(s)
- Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
958
|
Advances in the study of exosomal lncRNAs in tumors and the selection of research methods. Biomed Pharmacother 2019; 123:109716. [PMID: 31896067 DOI: 10.1016/j.biopha.2019.109716] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/19/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
Exosomes are endosome-derived extracellular vesicles that are released upon the fusion of multivesicular bodies with the plasma membrane. These vesicles contain proteins, lipids, and nucleic acids and are found in various human body fluids. Exosomes can transfer bioactive molecules to nearby or distant recipient cells, thereby affecting their function. Recently, exosomes have gained importance as a medium of communication between tumor cells. An increasing number of studies have found that non-coding RNAs in tumor cell-derived exosomes can regulate tumor microenvironments, inhibit immune cell function, promote the growth and invasion of tumor cells, and impart resistance to chemicals in tumor cells. In this review, we focus on the effects of exosomal long non-coding RNAs (lncRNAs) on tumors. As exosomes and their parent cells have similar biological characteristics and coated lncRNAs can exist stably in vivo without being degraded by RNases, exosomal lncRNAs have emerged as novel non-invasive tumor biomarkers for use in the early diagnosis and evaluation of prognosis of tumors. Advancements in the field have led to the development of a variety of techniques in exosomal non-coding RNA research. Currently, most methods include the separation and purification of exosomes, followed by RNA extraction, reverse transcription, and subsequent analyses; thus, these processes are very tedious and vulnerable to contamination and could lead to inaccurate and inconsistent results. Thus, there has been an increase in the development of detection methods for exosomal RNAs. Here, we discuss the existing research methods, their advantages and disadvantages, and a few new techniques.
Collapse
|
959
|
Andaluz Aguilar H, Iliuk AB, Chen IH, Tao WA. Sequential phosphoproteomics and N-glycoproteomics of plasma-derived extracellular vesicles. Nat Protoc 2019; 15:161-180. [PMID: 31863077 DOI: 10.1038/s41596-019-0260-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/16/2019] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) are increasingly being recognized as important vehicles for intercellular communication and as promising sources for biomarker discovery. Because the state of protein post-translational modifications (PTMs) such as phosphorylation and glycosylation can be a key determinant of cellular physiology, comprehensive characterization of protein PTMs in EVs can be particularly valuable for early-stage diagnostics and monitoring of disease status. However, the analysis of PTMs in EVs has been complicated by limited amounts of purified EVs, low-abundance PTM proteins, and interference from proteins and metabolites in biofluids. Recently, we developed an approach to isolate phosphoproteins and glycoproteins in EVs from small volumes of human plasma that enabled us to identify nearly 10,000 unique phosphopeptides and 1,500 unique N-glycopeptides. The approach demonstrated the feasibility of using these data to identify potential markers to differentiate disease from healthy states. Here we present an updated workflow to sequentially isolate phosphopeptides and N-glycopeptides, enabling multiple PTM analyses of the same clinical samples. In this updated workflow, we have improved the reproducibility and efficiency of EV isolation, protein extraction, and phosphopeptide/N-glycopeptide enrichment to achieve sensitive analyses of low-abundance PTMs in EVs isolated from 1 mL of plasma. The modularity of the workflow also allows for the characterization of phospho- or glycopeptides only and enables additional analysis of total proteomes and other PTMs of interest. After blood collection, the protocol takes 2 d, including EV isolation, PTM/peptide enrichment, mass spectrometry analysis, and data quantification.
Collapse
Affiliation(s)
| | - Anton B Iliuk
- Tymora Analytical Operations, West Lafayette, IN, USA.,Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - I-Hsuan Chen
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - W Andy Tao
- Department of Chemistry, Purdue University, West Lafayette, IN, USA. .,Tymora Analytical Operations, West Lafayette, IN, USA. .,Department of Biochemistry, Purdue University, West Lafayette, IN, USA. .,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
960
|
Jiayu H, Hanke Z, Ying G. The Role of Exosomes in Diseases Related to Infertility. Curr Stem Cell Res Ther 2019; 14:437-441. [PMID: 30674267 DOI: 10.2174/1574888x14666190123162842] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/07/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022]
Abstract
Exosomes, small extracellular vesicles with diameters of 40-100nm, are generated through the fusion of multivessel with plasma membrane and secreted by a variety of living cells. Exosomes contain lipid bilayer membrane and releasable functionally active proteins, mRNA and microRNAs (miRNAs). This article reviews the latest progress of researches on exosomes in diseases that lead to infertility.
Collapse
Affiliation(s)
- Huang Jiayu
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Hanke
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gao Ying
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
961
|
Wang B, Li T, Han X, Li Y, Cheng W, Wang L, Lu Z, Yang J, Zhao M. The Level of Circulating Microparticles in Patients with Coronary Heart Disease: a Systematic Review and Meta-Analysis. J Cardiovasc Transl Res 2019; 13:702-712. [PMID: 31834597 DOI: 10.1007/s12265-019-09945-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIMS To assess the correlation between microparticles (MPs) and subgroups of coronary heart disease (CHD), including stable angina (SA), unstable angina (UA), and myocardial infarction (MI). METHODS A literature search was carried out systematically to identify available case-control studies. The level of MPs was compared and MPs' merged standardized mean differences (SMDs) were pooled for the meta-analysis. RESULTS Six studies met the inclusion criteria and were used for systematic review and meta-analysis. The level of MPs was higher in patients with CHD than that in the NS (normal subjects) group (SMD 2.28; 95% confidence interval (CI) 1.70-2.85; P = 0.000), and was also significantly different in subgroups of CHD (UA vs SA: SMD 2.35, 95% CI 1.56-3.14, P = 0.000; MI vs SA: SMD 3.08, 95% CI 2.07-4.09, P = 0.000; MI vs UA: SMD 0.83, 95% CI 0.41-1.26, P = 0.000). The similar results were also found in subgroups analyses of CD31+CD42- endothelium-derived microparticles (EMPs) and CD144+EMPs. CONCLUSION The level of MPs, especially CD31+CD42-EMPs and CD144+EMPs, had an increasing trend with the degree of CHD: NS<SA<UA<MI, suggesting that MPs might be a potential biomarker to identify SA, UA, and MI.
Collapse
Affiliation(s)
- Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Xiaowan Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Wenkun Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Jingjing Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China.
| |
Collapse
|
962
|
Baldari S, Di Rocco G, Magenta A, Picozza M, Toietta G. Extracellular Vesicles-Encapsulated MicroRNA-125b Produced in Genetically Modified Mesenchymal Stromal Cells Inhibits Hepatocellular Carcinoma Cell Proliferation. Cells 2019; 8:cells8121560. [PMID: 31816923 PMCID: PMC6952965 DOI: 10.3390/cells8121560] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer and one of the prominent causes of cancer mortality, leading to approximately 780,000 deaths per year worldwide. Down-regulation of microRNA-125b (miR-125b) is a prognostic indicator in HCC patients. Conversely, over-expression of miR-125b in HCC cells induces cell cycle arrest, inhibits proliferation, migration and invasion. Extracellular vesicles (EVs) function as intercellular messengers transferring proteins, RNAs, DNAs, carbohydrates, and lipids. Since EVs protect their cargo from degradation, delivery of therapeutic bioactive molecules, in particular miRNAs, through EVs represents an innovative avenue for cancer therapy. In this study, we evaluated a replacement strategy for the treatment of HCC via delivery of EVs secreted from human adipose tissue-derived mesenchymal stromal/medicinal signaling cells (ASCs) genetically modified with a lentiviral vector expressing miR-125b with a specific ExoMotif sequence tag to enhance the loading into extracellular vesicles. In particular, we determined that the delivery of miR-125b-loaded EVs produced in engineered ASCs specifically reduces HCC cell proliferation in vitro modulating a series of miR-125b targets, which belong to the p53 signaling pathway. This proof-of-concept study supports the development of innovative therapeutic strategies for HCC via EV-mediated miRNA delivery.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
- Department of Medical Surgical Sciences and Biotechnologies, University of Rome “La Sapienza”, C.so della Repubblica 79, 04100 Latina, Italy
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
| | - Alessandra Magenta
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy;
| | - Mario Picozza
- Laboratory of Neuroimmunology, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
- Correspondence: ; Tel.: +39-06-5266-2604
| |
Collapse
|
963
|
Luo J, Xiong Y, Fu PF, Li EC, Qu L, Fan X, Cai ZJ, Lin AF. Exosomal long non-coding RNAs: biological properties and therapeutic potential in cancer treatment. J Zhejiang Univ Sci B 2019; 20:488-495. [PMID: 31090274 DOI: 10.1631/jzus.b1900039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exosomes and long non-coding RNAs (lncRNAs) are emerging as important elements contributing to a more comprehensive understanding of cancer development and progression. The discovery of lncRNAs in exosomes further indicates their bona fide biological functional roles in cancer development and drug resistance. In this review, we describe the biogenesis of exosomes and summarize the function of exosomal lncRNAs in the field of cancer research. These findings strikingly advance current knowledge of exosomal lncRNAs and suggest that they may be promising diagnostic biomarkers and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Jie Luo
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yan Xiong
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Pei-Fen Fu
- The Breast Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - En-Chun Li
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Lei Qu
- MOE Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao Fan
- MOE Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhi-Jian Cai
- Institute of Immunology, and Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ai-Fu Lin
- The Breast Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,MOE Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
964
|
Schulz E, Karagianni A, Koch M, Fuhrmann G. Hot EVs - How temperature affects extracellular vesicles. Eur J Pharm Biopharm 2019; 146:55-63. [PMID: 31805356 DOI: 10.1016/j.ejpb.2019.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/15/2019] [Accepted: 11/27/2019] [Indexed: 12/25/2022]
Abstract
In recent years, extracellular vesicles (EVs) and outer membrane vesicles (OMVs) have become an extensive and diverse field of research. They hold potential as diagnostic markers, therapeutics and for fundamental biological understanding. Despite ongoing studies, numerous information regarding function, content and stability of EVs remains unclear. If EVs and OMVs ought to be used as therapeutics and in clinical environments, their stability is one of the most important factors to be considered. Especially for formulation development, EVs and OMVs need to be stable at higher temperatures. To the best of our knowledge, very little work has been published regarding heat stability of neither EVs nor OMVs. In the present study, we investigated B lymphoblastoid cell-derived EVs and OMVs derived from myxobacterial species Sorangiineae as model vesicles. We exposed the vesicles to 37 °C, 50 °C, 70 °C and 100 °C for 1 h, 6 h and 24 h, and also autoclaved them. Interestingly, physico-chemical analyses such as size, particle concentration and protein concentration showed minor alterations, particularly at 37 °C. Flow cytometry analysis emphasised these results suggesting that after heat impact, EVs and OMVs were still able to be taken up by macrophage-like dTHP-1 cells. These data indicate that both mammalian and bacterial vesicles show intrinsic stability at physiological temperature. Our findings are important to consider for vesicle formulation and for advanced bioengineering approaches.
Collapse
Affiliation(s)
- Eilien Schulz
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken 66123, Germany
| | - Anna Karagianni
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2.2, Saarbrücken 66123, Germany
| | - Gregor Fuhrmann
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken 66123, Germany.
| |
Collapse
|
965
|
Wang J, Koo KM, Wang Y, Trau M. Engineering State-of-the-Art Plasmonic Nanomaterials for SERS-Based Clinical Liquid Biopsy Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900730. [PMID: 31832306 PMCID: PMC6891916 DOI: 10.1002/advs.201900730] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/26/2019] [Indexed: 05/23/2023]
Abstract
Precision oncology, defined as the use of the molecular understanding of cancer to implement personalized patient treatment, is currently at the heart of revolutionizing oncology practice. Due to the need for repeated molecular tumor analyses in facilitating precision oncology, liquid biopsies, which involve the detection of noninvasive cancer biomarkers in circulation, may be a critical key. Yet, existing liquid biopsy analysis technologies are still undergoing an evolution to address the challenges of analyzing trace quantities of circulating tumor biomarkers reliably and cost effectively. Consequently, the recent emergence of cutting-edge plasmonic nanomaterials represents a paradigm shift in harnessing the unique merits of surface-enhanced Raman scattering (SERS) biosensing platforms for clinical liquid biopsy applications. Herein, an expansive review on the design/synthesis of a new generation of diverse plasmonic nanomaterials, and an updated evaluation of their demonstrated SERS-based uses in liquid biopsies, such as circulating tumor cells, tumor-derived extracellular vesicles, as well as circulating cancer proteins, and tumor nucleic acids is presented. Existing challenges impeding the clinical translation of plasmonic nanomaterials for SERS-based liquid biopsy applications are also identified, and outlooks and insights into advancing this rapidly growing field for practical patient use are provided.
Collapse
Affiliation(s)
- Jing Wang
- Centre for Personalized NanomedicineAustralian Institute for Bioengineering and Nanotechnology (AIBN)The University of QueenslandBrisbaneQLD4072Australia
| | - Kevin M. Koo
- Centre for Personalized NanomedicineAustralian Institute for Bioengineering and Nanotechnology (AIBN)The University of QueenslandBrisbaneQLD4072Australia
| | - Yuling Wang
- Department of Molecular SciencesARC Excellence Centre for Nanoscale BioPhotonicsFaculty of Science and EngineeringMacquarie UniversitySydneyNSW2109Australia
| | - Matt Trau
- Centre for Personalized NanomedicineAustralian Institute for Bioengineering and Nanotechnology (AIBN)The University of QueenslandBrisbaneQLD4072Australia
- School of Chemistry and Molecular BiosciencesThe University of QueenslandBrisbaneQLD4072Australia
| |
Collapse
|
966
|
Chen R, Xu X, Qian Z, Zhang C, Niu Y, Wang Z, Sun J, Zhang X, Yu Y. The biological functions and clinical applications of exosomes in lung cancer. Cell Mol Life Sci 2019; 76:4613-4633. [PMID: 31352532 PMCID: PMC11105651 DOI: 10.1007/s00018-019-03233-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 06/24/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022]
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide, and the high incidence rates are worrisome. Exosomes are a class of extracellular vesicles secreted by most cells, including RNAs, proteins and lipids. Exosomes can mediate cell-to-cell communication in both physiologic and pathologic processes. Accumulated evidences show that cancer-derived exosomes aid in the recruitment and reprogramming of constituents correlated with tumor microenvironment. Furthermore, exosome-based clinical trials have been completed in advanced lung cancer patients. In this review, we discuss the roles of exosomes in a lung cancer microenvironment, such as its participation in lung cancer initiation, progression and metastasis as well as being involved in angiogenesis, epithelial-mesenchymal transition (EMT), immune escape, and drug resistance. In addition, we focus on the potential of exosomes as diagnostic and prognostic biomarkers in lung cancer, as well as the challenges faced by and advantages of exosomes as drug delivery vehicles and in exosome-based immunotherapy.
Collapse
Affiliation(s)
- Rui Chen
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xin Xu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zijun Qian
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Congcong Zhang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yongjie Niu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhixian Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Jianli Sun
- Department of Oncology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Xiao Zhang
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yongchun Yu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
967
|
Tian Y, Gong M, Hu Y, Liu H, Zhang W, Zhang M, Hu X, Aubert D, Zhu S, Wu L, Yan X. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles 2019; 9:1697028. [PMID: 31839906 PMCID: PMC6896440 DOI: 10.1080/20013078.2019.1697028] [Citation(s) in RCA: 365] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 10/08/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) have sparked tremendous interest owing to their prominent potential in diagnostics and therapeutics. Isolation of EVs from complex biological fluids with high purity is essential to the accurate analysis of EV cargo. Unfortunately, generally used isolation techniques do not offer good separation of EVs from non-EV contaminants. Hence, it is important to have a standardized method to characterise the properties of EV preparations, including size distribution, particle concentration, purity and phenotype. Employing a laboratory-built nano-flow cytometer (nFCM) that enables multiparameter analysis of single EVs as small as 40 nm, here we report a new benchmark to the quality and efficiency assessment of EVs isolated from plasma, one of the most difficult body fluids to work with. The performance of five widely used commercial isolation kits was examined and compared with the traditional differential ultracentrifugation (UC). Two to four orders of magnitude higher particle concentrations were observed for EV preparations from platelet-free plasma (PFP) by kits when compared with the EV preparation by UC, yet the purity was much lower. Meanwhile, the particle size distribution profiles of EV preparations by kits closely resembled those of PFP whereas the EV preparation by UC showed a broader size distribution at relatively large particle size. When these kits were used to isolate EVs from vesicle-depleted PFP (VD-PFP), comparable particle counts were obtained with their corresponding EV preparations from PFP, which confirmed again the isolation of a large quantity of non-vesicular contaminants. As CD9, CD63 and CD81 also exist in the plasma matrix, single-particle phenotyping of EVs offers distinct advantage in the validation of EVs compared with ensemble-averaged approaches, such as Western blot analysis. nFCM allows us to compare different isolation techniques without prejudice.
Collapse
Affiliation(s)
- Ye Tian
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Manfei Gong
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Yunyun Hu
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Haisheng Liu
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Wenqiang Zhang
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Miaomiao Zhang
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Xiuxiu Hu
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | | | - Shaobin Zhu
- NanoFCM Inc., Xiamen Pioneering Park for Overseas Chinese Scholars, Xiamen, People's Republic of China
| | - Lina Wu
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Xiaomei Yan
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
968
|
Label-free visualization and characterization of extracellular vesicles in breast cancer. Proc Natl Acad Sci U S A 2019; 116:24012-24018. [PMID: 31732668 DOI: 10.1073/pnas.1909243116] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Despite extensive interest, extracellular vesicle (EV) research remains technically challenging. One of the unexplored gaps in EV research has been the inability to characterize the spatially and functionally heterogeneous populations of EVs based on their metabolic profile. In this paper, we utilize the intrinsic optical metabolic and structural contrast of EVs and demonstrate in vivo/in situ characterization of EVs in a variety of unprocessed (pre)clinical samples. With a pixel-level segmentation mask provided by the deep neural network, individual EVs can be analyzed in terms of their optical signature in the context of their spatial distribution. Quantitative analysis of living tumor-bearing animals and fresh excised human breast tissue revealed abundance of NAD(P)H-rich EVs within the tumor, near the tumor boundary, and around vessel structures. Furthermore, the percentage of NAD(P)H-rich EVs is highly correlated with human breast cancer diagnosis, which emphasizes the important role of metabolic imaging for EV characterization as well as its potential for clinical applications. In addition to the characterization of EV properties, we also demonstrate label-free monitoring of EV dynamics (uptake, release, and movement) in live cells and animals. The in situ metabolic profiling capacity of the proposed method together with the finding of increasing NAD(P)H-rich EV subpopulations in breast cancer have the potential for empowering applications in basic science and enhancing our understanding of the active metabolic roles that EVs play in cancer progression.
Collapse
|
969
|
Dong Z, Tang C, Zhang Z, Zhou W, Zhao R, Wang L, Xu J, Wu Y, Wu J, Zhang X, Xu L, Zhao L, Fang X. Simultaneous Detection of Exosomal Membrane Protein and RNA by Highly Sensitive Aptamer Assisted Multiplex–PCR. ACS APPLIED BIO MATERIALS 2019; 3:2560-2567. [DOI: 10.1021/acsabm.9b00825] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Zaizai Dong
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Chuanhao Tang
- Department of Medical Oncology, Peking University International Hospital, Beijing 102206, P. R. China
| | - Zhen Zhang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Wei Zhou
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Rong Zhao
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Lina Wang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jiachao Xu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yayun Wu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jiang Wu
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xing Zhang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Li Xu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Libo Zhao
- Echo Biotech Co., Ltd, Beijing 102206, P. R. China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
970
|
Shi L, Kuhnell D, Borra VJ, Langevin SM, Nakamura T, Esfandiari L. Rapid and label-free isolation of small extracellular vesicles from biofluids utilizing a novel insulator based dielectrophoretic device. LAB ON A CHIP 2019; 19:3726-3734. [PMID: 31588942 PMCID: PMC7477750 DOI: 10.1039/c9lc00902g] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Exosomes are nano-scale membrane-encapsulated vesicles produced by the majority of cells and have emerged as a rich source of biomarkers for a wide variety of diseases. Although many approaches have been developed for exosome isolation from biofluids, most of them have substantial shortcomings including long processing time, inefficiency, high cost, lack of specificity and/or surface marker-dependency. To address these issues, here we report a novel insulator-based dielectrophoretic (iDEP) device predicated on an array of borosilicate micropipettes to rapidly isolate exosomes from conditioned cell culture media and biofluids, such as plasma, serum, and saliva. The device is capable of exosome isolation from small sample volumes of 200 μL within 20 minutes under a relatively low (10 V cm-1) direct current (DC). This device is easy to fabricate thus, no cleanroom facility and expensive equipment are needed. Therefore, the iDEP device offers a rapid and cost-effective strategy for exosome isolation from biofluids in timely manner while maintaining the yield and purity.
Collapse
Affiliation(s)
- Leilei Shi
- Department of Electrical Engineering and Computer Science, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Damaris Kuhnell
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Vishnupriya J Borra
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Scott M Langevin
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, USA and Cincinnati Cancer Center, Cincinnati, OH, USA
| | - Takahisa Nakamura
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA and Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA and Department of Metabolic Bioregulation, Institute of Development, Aging and Cancer, Tohoku University, Japan
| | - Leyla Esfandiari
- Department of Electrical Engineering and Computer Science, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA and Cincinnati Cancer Center, Cincinnati, OH, USA and Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
971
|
Brittain GC, Chen YQ, Martinez E, Tang VA, Renner TM, Langlois MA, Gulnik S. A Novel Semiconductor-Based Flow Cytometer with Enhanced Light-Scatter Sensitivity for the Analysis of Biological Nanoparticles. Sci Rep 2019; 9:16039. [PMID: 31690751 PMCID: PMC6831566 DOI: 10.1038/s41598-019-52366-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 10/15/2019] [Indexed: 01/20/2023] Open
Abstract
The CytoFLEX is a novel semiconductor-based flow cytometer that utilizes avalanche photodiodes, wavelength-division multiplexing, enhanced optics, and diode lasers to maximize light capture and minimize optical and electronic noise. Due to an increasing interest in the use of extracellular vesicles (EVs) as disease biomarkers, and the growing desire to use flow cytometry for the analyses of biological nanoparticles, we assessed the light-scatter sensitivity of the CytoFLEX for small-particle detection. We found that the CytoFLEX can fully resolve 70 nm polystyrene and 98.6 nm silica beads by violet side scatter (VSSC). We further analyzed the detection limit for biological nanoparticles, including viruses and EVs, and show that the CytoFLEX can detect viruses down to 81 nm and EVs at least as small as 65 nm. Moreover, we could immunophenotype EV surface antigens, including directly in blood and plasma, demonstrating the double labeling of platelet EVs with CD61 and CD9, as well as triple labeling with CD81 for an EV subpopulation in one donor. In order to assess the refractive indices (RIs) of the viruses and EVs, we devised a new method to inversely calculate the RIs using the intensity vs. size data together with Mie-theory scatter efficiencies scaled to reference-particle measurements. Each of the viruses tested had an equivalent RI, approximately 1.47 at 405 nm, which suggests that flow cytometry can be more broadly used to easily determine virus sizes. We also found that the RIs of EVs increase as the particle diameters decrease below 150 nm, increasing from 1.37 for 200 nm EVs up to 1.61 for 65 nm EVs, expanding the lower range of EVs that can be detected by light scatter. Overall, we demonstrate that the CytoFLEX has an unprecedented level of sensitivity compared to conventional flow cytometers. Accordingly, the CytoFLEX can be of great benefit to virology and EV research, and will help to expand the use of flow cytometry for minimally invasive liquid biopsies by allowing for the direct analysis of antigen expression on biological nanoparticles within patient samples, including blood, plasma, urine and bronchoalveolar lavages.
Collapse
Affiliation(s)
- George C Brittain
- Beckman Coulter Life Sciences, Life Science Research, Miami, FL, USA
| | - Yong Q Chen
- Beckman Coulter Life Sciences, Life Science Research, Miami, FL, USA
| | - Edgar Martinez
- Beckman Coulter Life Sciences, Particle Characterization, Miami, FL, USA
| | - Vera A Tang
- University of Ottawa Flow Cytometry and Virometry Core Facility, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Tyler M Renner
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Marc-André Langlois
- University of Ottawa Flow Cytometry and Virometry Core Facility, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- uOttawa Center for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, Canada
| | - Sergei Gulnik
- Beckman Coulter Life Sciences, Life Science Research, Miami, FL, USA.
| |
Collapse
|
972
|
Pramanik A, Gates K, Patibandla S, Davis D, Begum S, Iftekhar R, Alamgir S, Paige S, Porter MM, Ray PC. Water-Soluble and Bright Luminescent Cesium–Lead–Bromide Perovskite Quantum Dot–Polymer Composites for Tumor-Derived Exosome Imaging. ACS APPLIED BIO MATERIALS 2019; 2:5872-5879. [DOI: 10.1021/acsabm.9b00837] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Avijit Pramanik
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Kaelin Gates
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Shamily Patibandla
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Dalephine Davis
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Salma Begum
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Riwad Iftekhar
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Saadman Alamgir
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Shekyra Paige
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Maurice M. Porter
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| | - Paresh Chandra Ray
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi, United States
| |
Collapse
|
973
|
Liu C, Feng Q, Sun J. Lipid Nanovesicles by Microfluidics: Manipulation, Synthesis, and Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804788. [PMID: 30570773 DOI: 10.1002/adma.201804788] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/11/2018] [Indexed: 05/20/2023]
Abstract
Lipid nanovesicles, including endogenous exosomes and synthetic lipid nanoparticles, have shown great potential in disease diagnostics, drug delivery, and cancer biology. Naturally secreted nanovesicles are promising biomarkers for early detection of cancers in vitro. Synthetic nanovesicles serve as robust drug delivery systems with enhanced tumor targeting in vivo. Microfluidic platforms with features of excellent flow control and rapid mixing are exploited as versatile tools for studying lipid nanovesicles of small sizes and delicate structures. Here, an overview of microfluidics for precise manipulation and synthesis of lipid nanovesicles is provided. The mechanisms of isolation and detection of exosomes in microfluidics, as well as the clinical utility of exosomes for cancer diagnosis, are discussed. Several microfluidic designs for controlled assembly of a variety of lipid nanovesicles are highlighted. Opportunities and outstanding challenges of microfluidics-based investigation of lipid nanovesicles are discussed.
Collapse
Affiliation(s)
- Chao Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Feng
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiashu Sun
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
974
|
Li W, Wang H, Zhao Z, Gao H, Liu C, Zhu L, Wang C, Yang Y. Emerging Nanotechnologies for Liquid Biopsy: The Detection of Circulating Tumor Cells and Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805344. [PMID: 30589111 DOI: 10.1002/adma.201805344] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/29/2018] [Indexed: 05/18/2023]
Abstract
Liquid biopsy enables noninvasive and dynamic analysis of molecular or cellular biomarkers, and therefore holds great potential for the diagnosis, prognosis, monitoring of disease progress and treatment efficacy, understanding of disease mechanisms, and identification of therapeutic targets for drug development. In this review, the recent progress in nanomaterials, nanostructures, nanodevices, and nanosensors for liquid biopsy is summarized, with a focus on the detection and molecular characterization of circulating tumor cells (CTCs) and extracellular vesicles (EVs). The developments and advances of nanomaterials and nanostructures in enhancing the sensitivity, specificity, and purity for the detection of CTCs and EVs are discussed. Sensing techniques for signal transduction and amplification as well as visualization strategies are also discussed. New technologies for the reversible release of the isolated CTCs and EVs and for single-CTC/EV analysis are summarized. Emerging microfluidic platforms for the integral on-chip isolation, detection, and molecular analysis are also included. The opportunities, challenges, and prospects of these innovative materials and technologies, especially with regard to their feasibility in clinical applications, are discussed. The applications of nanotechnology-based liquid biopsy will bring new insight into the clinical practice in monitoring and treatment of tumor and other significant diseases.
Collapse
Affiliation(s)
- Wenzhe Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zijian Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Houqian Gao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Changliang Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
975
|
Liu J, Mosavati B, Oleinikov AV, Du E. Biosensors for Detection of Human Placental Pathologies: A Review of Emerging Technologies and Current Trends. Transl Res 2019; 213:23-49. [PMID: 31170377 PMCID: PMC6783355 DOI: 10.1016/j.trsl.2019.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 02/06/2023]
Abstract
Substantial growth in the biosensor research has enabled novel, sensitive and point-of-care diagnosis of human diseases in the last decade. This paper presents an overview of the research in the field of biosensors that can potentially predict and diagnosis of common placental pathologies. A survey of biomarkers in maternal circulation and their characterization methods is presented, including markers of oxidative stress, angiogenic factors, placental debris, and inflammatory biomarkers that are associated with various pathophysiological processes in the context of pregnancy complications. Novel biosensors enabled by microfluidics technology and nanomaterials is then reviewed. Representative designs of plasmonic and electrochemical biosensors for highly sensitive and multiplexed detection of biomarkers, as well as on-chip sample preparation and sensing for automatic biomarker detection are illustrated. New trends in organ-on-a-chip based placental disease models are highlighted to illustrate the capability of these in vitro disease models in better understanding the complex pathophysiological processes, including mass transfer across the placental barrier, oxidative stress, inflammation, and malaria infection. Biosensor technologies that can be potentially embedded in the placental models for real time, label-free monitoring of these processes and events are suggested. Merger of cell culture in microfluidics and biosensing can provide significant potential for new developments in advanced placental models, and tools for diagnosis, drug screening and efficacy testing.
Collapse
Affiliation(s)
- Jia Liu
- College of Engineering and Computer Science, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida
| | - Babak Mosavati
- College of Engineering and Computer Science, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida
| | - Andrew V Oleinikov
- Charles E. Schmidt College of Medicine, Department of Biomedical Science, Florida Atlantic University, Boca Raton, Florida
| | - E Du
- College of Engineering and Computer Science, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida; Charles E. Schmidt College of Science, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida.
| |
Collapse
|
976
|
Wu Z, Zhang Z, Xia W, Cai J, Li Y, Wu S. Extracellular vesicles in urologic malignancies-Implementations for future cancer care. Cell Prolif 2019; 52:e12659. [PMID: 31469460 PMCID: PMC6869217 DOI: 10.1111/cpr.12659] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs), a heterogeneous group of vesicles differing in size and shape, cargo content and function, are membrane-bound and nano-sized vesicles that could be released by nearly all variations of cells. EVs have gained considerable attention in the past decades for their functions in modulating intercellular signalling and roles as potential pools for the novel diagnostic and prognostic biomarkers, as well as therapeutic targets in several cancers including urological neoplasms. In general, human and animal cells both can release distinct types of EVs, including exosomes, microvesicles, oncosomes and large oncosomes, and apoptotic bodies, while the content of EVs can be divided into proteins, lipids and nucleic acids. However, the lack of standard methods for isolation and detection platforms rein the widespread usage in clinical applications warranted furthermore investigations in the development of reliable, specific and sensitive isolation techniques. Whether and how the EVs work has become pertinent issues. With the aid of high-throughput proteomics or genomics methods, a fully understanding of contents contained in EVs from urogenital tumours, beyond all doubt, will improve our ability to identify the complex genomic alterations in the process of cancer and, in turn, contribute to detect potential therapeutic target and then provide personalization strategy for patient.
Collapse
Affiliation(s)
- Zhangsong Wu
- Medical CollegeShenzhen UniversityShenzhenChina
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
| | - Zhiqiang Zhang
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
| | - Wuchao Xia
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Medical CollegeAnhui University of Science and TechnologyHuainanChina
| | - Jiajia Cai
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Medical CollegeAnhui University of Science and TechnologyHuainanChina
| | - Yuqing Li
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
| | - Song Wu
- Medical CollegeShenzhen UniversityShenzhenChina
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen UniversityShenzhen UniversityShenzhenChina
- Medical CollegeAnhui University of Science and TechnologyHuainanChina
- Department of Urological Surgery, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
977
|
Abraham A, Krasnodembskaya A. Mesenchymal stem cell-derived extracellular vesicles for the treatment of acute respiratory distress syndrome. Stem Cells Transl Med 2019; 9:28-38. [PMID: 31647191 PMCID: PMC6954712 DOI: 10.1002/sctm.19-0205] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/26/2019] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a serious and potentially fatal acute inflammatory lung condition which currently has no specific treatments targeting its pathophysiology. However, mesenchymal stem cells have been shown to have very promising therapeutic potential, and recently, it has been established that their effect is largely due to the transfer of extracellular vesicles (EVs). EVs have been shown to transfer a variety of substances such as mRNA, miRNA, and even organelles such as mitochondria in order to ameliorate ARDS in preclinical models. In addition, the fact that they have been proven to have the same effect as their parent cells combined with their numerous advantages over whole cell administration means that they are a promising candidate for clinical application that merits further research.
Collapse
Affiliation(s)
- Aswin Abraham
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| |
Collapse
|
978
|
Shang S, Wang J, Chen S, Tian R, Zeng H, Wang L, Xia M, Zhu H, Zuo C. Exosomal miRNA-1231 derived from bone marrow mesenchymal stem cells inhibits the activity of pancreatic cancer. Cancer Med 2019; 8:7728-7740. [PMID: 31642612 PMCID: PMC6912060 DOI: 10.1002/cam4.2633] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/28/2019] [Accepted: 10/06/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a highly malignant tumor with increased morbidity and mortality, which is difficult to diagnose and cure in the clinic. Through secreting exosomes containing biological molecules, including diverse RNAs and proteins, bone marrow mesenchymal stem cells (BM-MSCs) influence the immunity, inflammation, tumor environment, and cancer metastasis. In this study, low expression of miRNA-1231 (miR-1231) in exosomes derived from the peripheral blood was significantly correlated with the TNM stage of PC, suggesting the potential inhibitory effect of exosomal miR-1231 on PC occurrence and development. The proliferation, migration, invasion, and adhesion to the matrix of PC cells BxPC-3 and PANC-1 were negatively regulated by exosomes derived from the supernatants of BM-MSCs that transfected with miR-1231 oligonucleotides. Simultaneously, tumor growth in vivo was seriously restrained in BALB/C nude mice by tail vein injection with exosomes originated from BM-MSCs that transfected with miR-1231 mimics. The exosomes extracted from BM-MSCs with high level of miR-1231 inhibit the activity of PC, providing the potential application for developing new and efficient medicine for cancer therapy, especially for PC treatment. The exosomal miR-1231 of peripheral blood may also be a potential indicator for PC diagnosis in the future.
Collapse
Affiliation(s)
- Song Shang
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Jinfeng Wang
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Shilin Chen
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Renyun Tian
- Department of Molecular Medicine, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Hui Zeng
- Graduates School, University of South China, Hengyang, China
| | - Liang Wang
- Graduates School, University of South China, Hengyang, China
| | - Man Xia
- Department of Gynecological Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Haizhen Zhu
- Department of Molecular Medicine, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| |
Collapse
|
979
|
Rodell CB, Koch PD, Weissleder R. Screening for new macrophage therapeutics. Theranostics 2019; 9:7714-7729. [PMID: 31695796 PMCID: PMC6831478 DOI: 10.7150/thno.34421] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid derived macrophages play a key role in many human diseases, and their therapeutic modulation via pharmacological means is receiving considerable attention. Of particular interest is the fact that these cells are i) dynamic phenotypes well suited to therapeutic manipulation and ii) phagocytic, allowing them to be efficiently targeted with nanoformulations. However, it is important to consider that macrophages represent heterogeneous populations of subtypes with often competing biological behaviors and functions. In order to develop next generation therapeutics, it is therefore essential to screen for biological effects through a combination of in vitro and in vivo assays. Here, we review the state-of-the-art techniques, including both cell based screens and in vivo imaging tools that have been developed for assessment of macrophage phenotype. We conclude with a forward-looking perspective on the growing need for noninvasive macrophage assessment and laboratory assays to be put into clinical practice and the potential broader impact of myeloid-targeted therapeutics.
Collapse
|
980
|
Mao W, Wen Y, Lei H, Lu R, Wang S, Wang Y, Chen R, Gu Y, Zhu L, Abhange KK, Quinn ZJ, Chen Y, Xue F, Zheng M, Wan Y. Isolation and Retrieval of Extracellular Vesicles for Liquid Biopsy of Malignant Ground-Glass Opacity. Anal Chem 2019; 91:13729-13736. [DOI: 10.1021/acs.analchem.9b03064] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Wenjun Mao
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Yi Wen
- The Pq Laboratory
of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University—SUNY, Binghamton, New York 13902, United States
| | - Haozhi Lei
- PerMed Biomedicine Institute, Shanghai, 201203, China
| | - Rongguo Lu
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Shengfei Wang
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Yuheng Wang
- PerMed Biomedicine Institute, Shanghai, 201203, China
| | - Ruo Chen
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Yuanyuan Gu
- PerMed Biomedicine Institute, Shanghai, 201203, China
| | - Lin Zhu
- PerMed Biomedicine Institute, Shanghai, 201203, China
| | - Komal K. Abhange
- The Pq Laboratory
of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University—SUNY, Binghamton, New York 13902, United States
| | - Zachary J. Quinn
- The Pq Laboratory
of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University—SUNY, Binghamton, New York 13902, United States
| | - Yundi Chen
- The Pq Laboratory
of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University—SUNY, Binghamton, New York 13902, United States
| | - Fei Xue
- The Pq Laboratory
of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University—SUNY, Binghamton, New York 13902, United States
| | - Mingfeng Zheng
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Yuan Wan
- The Pq Laboratory
of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University—SUNY, Binghamton, New York 13902, United States
| |
Collapse
|
981
|
Shu Z, Tan J, Miao Y, Zhang Q. The role of microvesicles containing microRNAs in vascular endothelial dysfunction. J Cell Mol Med 2019; 23:7933-7945. [PMID: 31576661 PMCID: PMC6850938 DOI: 10.1111/jcmm.14716] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/28/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022] Open
Abstract
Many studies have shown that endothelial dysfunction is associated with a variety of cardiovascular diseases. The endothelium is one of the primary targets of circulating microvesicles. Besides, microRNAs emerge as important regulators of endothelial cell function. As a delivery system of microRNAs, microvesicles play an active and important role in regulating vascular endothelial function. In recent years, some studies have shown that microvesicles containing microRNAs regulate the pathophysiological changes in vascular endothelium, such as cell apoptosis, proliferation, migration and inflammation. These studies have provided some clues for the possible roles of microvesicles and microRNAs in vascular endothelial dysfunction‐associated diseases, and opened the door towards discovering potential novel therapeutic targets. In this review, we provide an overview of the main characteristics of microvesicles and microRNAs, summarizing their potential role and mechanism in endothelial dysfunction, and discussing the clinical application and existing problems of microvesicles for better translational applications.
Collapse
Affiliation(s)
- Zeyu Shu
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
982
|
Thippabhotla S, Zhong C, He M. 3D cell culture stimulates the secretion of in vivo like extracellular vesicles. Sci Rep 2019; 9:13012. [PMID: 31506601 PMCID: PMC6736862 DOI: 10.1038/s41598-019-49671-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022] Open
Abstract
For studying cellular communications ex-vivo, a two-dimensional (2D) cell culture model is currently used as the “gold standard”. 2D culture models are also widely used in the study of RNA expression profiles from tumor cells secreted extracellular vesicles (EVs) for tumor biomarker discovery. Although the 2D culture system is simple and easily accessible, the culture environment is unable to represent in vivo extracellular matrix (ECM) microenvironment. Our study observed that 2D- culture derived EVs showed significantly different profiles in terms of secretion dynamics and essential signaling molecular contents (RNAs and DNAs), when compared to the three-dimensional (3D) culture derived EVs. By performing small RNA next-generation sequencing (NGS) analysis of cervical cancer cells and their EVs compared with cervical cancer patient plasma EV-derived small RNAs, we observed that 3D- culture derived EV small RNAs differ from their parent cell small RNA profile which may indicate a specific sorting process. Most importantly, the 3D- culture derived EV small RNA profile exhibited a much higher similarity (~96%) to in vivo circulating EVs derived from cervical cancer patient plasma. However, 2D- culture derived EV small RNA profile correlated better with only their parent cells cultured in 2D. On the other hand, DNA sequencing analysis suggests that culture and growth conditions do not affect the genomic information carried by EV secretion. This work also suggests that tackling EV molecular alterations secreted into interstitial fluids can provide an alternative, non-invasive approach for investigating 3D tissue behaviors at the molecular precision. This work could serve as a foundation for building precise models employed in mimicking in vivo tissue system with EVs as the molecular indicators or transporters. Such models could be used for investigating tumor biomarkers, drug screening, and understanding tumor progression and metastasis.
Collapse
Affiliation(s)
- Sirisha Thippabhotla
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, Kansas, 66045, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, Kansas, 66045, USA.,Bioengineering Research Center, University of Kansas, Lawrence, Kansas, 66045, USA
| | - Mei He
- Bioengineering Research Center, University of Kansas, Lawrence, Kansas, 66045, USA. .,Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, 66045, USA. .,Department of Chemistry, University of Kansas, Lawrence, Kansas, 66045, USA.
| |
Collapse
|
983
|
Brambilla D, Chiari M, Gori A, Cretich M. Towards precision medicine: the role and potential of protein and peptide microarrays. Analyst 2019; 144:5353-5367. [PMID: 31384857 DOI: 10.1039/c9an01142k] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Although the traditional strategy of developing general medical treatments for heterogeneous patient populations has a well-established track record, the acknowledgment that one-size-does-not-fit-all is pushing health-care to enter a new era of tailored interventions. The advent of precision medicine is fueled by the high-throughput analysis of individual DNA variants and mRNA expression profiles. However, due to the role of proteins in providing a more direct view of disease states than genomics alone, the ability to comprehensively analyze protein alterations and post translational modifications (PTMs) is a necessary step to unravel disease mechanisms, develop novel biomarkers and targeted therapies. Protein and peptide microarrays can play a major role in this frame, due to high-throughput, low sample consumption and wide applicability. Here, their current role and potentialities are discussed through the review of some promising applications in the fields of PTMs analysis, enzyme screening, high-content immune-profiling and the phenotyping of extracellular vesicles.
Collapse
Affiliation(s)
- Dario Brambilla
- Consiglio Nazionale delle Ricerche, Istituto di Chimica del Riconoscimento Molecolare (ICRM), Via Mario Bianco, 9, 20131, Milano, Italy.
| | | | | | | |
Collapse
|
984
|
Su W, Li H, Chen W, Qin J. Microfluidic strategies for label-free exosomes isolation and analysis. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
985
|
Lee JH, Choi JH, Chueng STD, Pongkulapa T, Yang L, Cho HY, Choi JW, Lee KB. Nondestructive Characterization of Stem Cell Neurogenesis by a Magneto-Plasmonic Nanomaterial-Based Exosomal miRNA Detection. ACS NANO 2019; 13:8793-8803. [PMID: 31361458 DOI: 10.1021/acsnano.9b01875] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The full realization of stem cell-based treatments for neurodegenerative diseases requires precise control and characterization of stem cell fate. Herein, we report a multifunctional magneto-plasmonic nanorod (NR)-based detection platform to address the limitations associated with the current destructive characterization methods of stem cell neurogenesis. Exosomes and their inner contents have been discovered to play critical roles in cell-cell interactions and intrinsic cellular regulations and have received wide attention as next-generation biomarkers. Moreover, exosomal microRNAs (miRNA) also offer an essential avenue for nondestructive molecular analyses of cell cytoplasm components. To this end, our developed nondestructive, selective, and sensitive detection platform has (i) an immunomagnetic active component for exosome isolation and (ii) a plasmonic/metal-enhanced fluorescence component for sensitive exosomal miRNA detection to characterize stem cell differentiation. In a proof-of-concept demonstration, our multifunctional magneto-plasmonic NR successfully detected the expression level of miRNA-124 and characterized neurogenesis of human-induced pluripotent stem cell-derived neural stem cells in a nondestructive and efficient manner. Furthermore, we demonstrated the versatility and feasibility of our multifunctional magneto-plasmonic NRs by characterizing a heterogeneous population of neural cells in an ex vivo rodent model. Collectively, we believe our multifunctional magneto-plasmonic NR-based exosomal miRNA detection platform has a great potential to investigate the function of cell-cell interactions and intrinsic cellular regulators for controlling stem cell differentiation.
Collapse
Affiliation(s)
- Jin-Ho Lee
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
- Department of Chemical and Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Jin-Ha Choi
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
- Department of Chemical and Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Letao Yang
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Hyeon-Yeol Cho
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
- Department of Chemical and Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
- Department of Life and Nanopharmaceutical Science, College of Pharmacy , Kyung Hee University , Seoul 02447 , Republic of Korea
| |
Collapse
|
986
|
Vinaiphat A, Sze SK. Clinical implications of extracellular vesicles in neurodegenerative diseases. Expert Rev Mol Diagn 2019; 19:813-824. [PMID: 31429341 DOI: 10.1080/14737159.2019.1657407] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction: Extracellular vesicles (EVs) released by neural cells play a crucial role in intracellular communication in both physiological and pathological states. Recent studies have shown that the neuropathogenic manifestation of many progressive nervous system diseases including Parkinson's disease (PD), Alzheimer's diseases (AD), and amyotrophic lateral sclerosis (ALS). These diseases are frequently found to be associated with the accumulation of misfolded proteins, exploit EVs for the spread of aggregates to naive cells in a prion-like mechanism. Therefore, characterization of EVs and understanding their mechanism of action could open a window of opportunity to discover biomarkers and therapeutic targets in a disease-specific manner. Areas covered: In this review, we discuss the role of neural cells-derived EVs in normal and disease states. We also highlight their biomedical potential in modern medicine, including the use of circulating EVs as biomarkers for diagnosis with a special focus on newly-identified potential biomarkers in neurodegenerative disease, and novel methodologies in EVs isolation. Expert opinion: Systematic and comprehensive analysis of EVs in different biofluid sources is needed. Considering the potential for tremendous clinical benefits of EVs research in neurodegenerative disease, there is also an urgent need to standardize neural cells-derived EV enrichment protocols for consensus results.
Collapse
Affiliation(s)
- Arada Vinaiphat
- School of Biological Sciences, Nanyang Technological University , Singapore Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University , Singapore Singapore
| |
Collapse
|
987
|
Zhang P, Wang L, Fang Y, Zheng D, Lin T, Wang H. Label-Free Exosomal Detection and Classification in Rapid Discriminating Different Cancer Types Based on Specific Raman Phenotypes and Multivariate Statistical Analysis. Molecules 2019; 24:molecules24162947. [PMID: 31416240 PMCID: PMC6720265 DOI: 10.3390/molecules24162947] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 01/04/2023] Open
Abstract
Exosomes contain different functional bimolecular characteristics related to physiological or pathological processes and are now recognized as new biomarkers in different human cancers. Rapid detection and classification of cancer-related exosomes might be helpful in the rapid screening of patients that may have cancer. Here, we report a surface enhanced Raman scattering technology for rapid and label-free exosomal detection (Exo-SERS) to aid in the discrimination of different cancer cells based on specific Raman phenotypes and multivariate statistical analysis. The results demonstrated that exosomes derived from both tumor cells and normal cells exhibit special, unique Raman phenotypes. Using the Exo-SERS method, the cancer cells were accurately discriminated from normal cells, and subtle molecular changes between the different cell types could be detected with high sensitive. This research provides a rapid, label-free and non-destructive manner for detecting and discriminating between cancer types.
Collapse
Affiliation(s)
- Ping Zhang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Limin Wang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Yaping Fang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Dawei Zheng
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Taifeng Lin
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Huiqin Wang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
988
|
Functions of Exosomes in the Triangular Relationship between the Tumor, Inflammation, and Immunity in the Tumor Microenvironment. J Immunol Res 2019; 2019:4197829. [PMID: 31467934 PMCID: PMC6701352 DOI: 10.1155/2019/4197829] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Exosomes are extracellular vesicles that contain diverse components such as genetic materials, proteins, and lipids. Owing to their distinct derivation and tissue specificity, exosomes act as double-edged swords during the development of neoplasms. On the one hand, tumor-derived exosomes can modulate the immune system during tumorigenesis by regulating inflammatory cell infiltration and oxidative stress and by promoting epithelial-to-mesenchymal transition and immune-induced tumor dormancy. On the other hand, components of specific immune cell-derived exosomes may contribute to the efficacy of antitumor immunotherapy. In this review, we demonstrate the pivotal role of exosomes in the triangular relationship in the tumor microenvironment between the tumor, inflammation, and immunity, which may provide potential strategies for tumor immunotherapy at genetic and cellular levels.
Collapse
|
989
|
Exosomal MMP2 derived from mature osteoblasts promotes angiogenesis of endothelial cells via VEGF/Erk1/2 signaling pathway. Exp Cell Res 2019; 383:111541. [PMID: 31369752 DOI: 10.1016/j.yexcr.2019.111541] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 11/24/2022]
Abstract
The skeletal system is a dynamic organ that continuously undergoes coupled trabeculae and blood vessels remodeling, indicating the possible existence of molecular crosstalk between endothelial and osteoblastic cells. Since the cross-talk between bone-forming osteoblasts (OBs) and vessel-forming endothelial cells (ECs) have progressively gained investigators' attention, few studies focused on the regulatory function of extracellular vesicles derived from OBs on ECs. In this study, the effect of the exosomes derived from mature osteoblasts (MOBs) on the ECs was investigated. Firstly, exosomes derived from mature osteoblasts (MOB-Exos) were isolated and identified by NanoSight light scatter technology, electron microscopy and Western bolting. Fluorescent labeling of MOB-Exos revealed its internalization by ECs. RNA interference technique was used to knock down matrix metalloproteinase-2 (MMP2) in MOB-Exos. Then ECs were co-cultured with MOB-Exos and MMP2 knockdown MOB-Exos. Wound healing migration assay, transwell migration assay, CCK-8 assay and tube formation assay of ECs were conducted to determine the angiogenic capability of ECs. Then the VEGF/Erk1/2 pathway markers were detected by Western blot. Our results showed that MOB-Exos could promote the proliferation, migration and tube formation of ECs. Meanwhile, the promoted angiogenetic capacities of ECs were impaired when MMP2 in MOB-Exos was knocked down. In addition, immunoblotting indicated that MOB-Exos could promote the activation of the VEGF/Erk1/2 pathway of ECs; whereas the activation of the VEGF/Erk1/2 pathway was attenuated when the ECs were co-cultured with the MMP2 knockdown MOB-Exos. In conclusion, the MMP-2 existing in exosomes derived from MOBs could promote the angiogenesis of ECs in vitro, which might be realized through VEGF/Erk1/2 signaling pathway.
Collapse
|
990
|
Cell Intrinsic and Extrinsic Mechanisms of Caveolin-1-Enhanced Metastasis. Biomolecules 2019; 9:biom9080314. [PMID: 31362353 PMCID: PMC6723107 DOI: 10.3390/biom9080314] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/17/2019] [Accepted: 07/25/2019] [Indexed: 12/19/2022] Open
Abstract
Caveolin-1 (CAV1) is a scaffolding protein with a controversial role in cancer. This review will initially discuss earlier studies focused on the role as a tumor suppressor before elaborating subsequently on those relating to function of the protein as a promoter of metastasis. Different mechanisms are summarized illustrating how CAV1 promotes such traits upon expression in cancer cells (intrinsic mechanisms). More recently, it has become apparent that CAV1 is also a secreted protein that can be included into exosomes where it plays a significant role in determining cargo composition. Thus, we will also discuss how CAV1 containing exosomes from metastatic cells promote malignant traits in more benign recipient cells (extrinsic mechanisms). This ability appears, at least in part, attributable to the transfer of specific cargos present due to CAV1 rather than the transfer of CAV1 itself. The evolution of how our perception of CAV1 function has changed since its discovery is summarized graphically in a time line figure.
Collapse
|
991
|
Deciphering Fungal Extracellular Vesicles: From Cell Biology to Pathogenesis. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019. [DOI: 10.1007/s40588-019-00128-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
992
|
Yang XX, Sun C, Wang L, Guo XL. New insight into isolation, identification techniques and medical applications of exosomes. J Control Release 2019; 308:119-129. [PMID: 31325471 DOI: 10.1016/j.jconrel.2019.07.021] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/29/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022]
Abstract
Exosomes, which are nano-vesicles produced by most cell types, play an irreplaceable role in cell-cell communication. They are extracellular small vesicles that can delivery various cargos of DNA, RNAs, proteins, and lipids. Because exosomes have different secretory components under physiological conditions and pathological conditions, it has been extensively studied in the field of diseases as a therapeutic target, as a drug/gene delivery vector and as a novel cancer marker. Despite the great development in recent decades, there are still many obstacles to be overcome, for example, the separation method is not standardized with low yield and poor stability, which limit its medical application. This review mainly summarizes the main progresses of isolation and identification techniques, diversity function and medical application of exosomes in recent years.
Collapse
Affiliation(s)
- Xiao-Xia Yang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), Drug Screening Unit Platform, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Chao Sun
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), Drug Screening Unit Platform, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Lei Wang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), Drug Screening Unit Platform, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xiu-Li Guo
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), Drug Screening Unit Platform, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
993
|
An Y, Jin T, Zhu Y, Zhang F, He P. An ultrasensitive electrochemical aptasensor for the determination of tumor exosomes based on click chemistry. Biosens Bioelectron 2019; 142:111503. [PMID: 31376716 DOI: 10.1016/j.bios.2019.111503] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Exosomes, lipid bilayer membrane vesicles, can guide various pathological and physiological processes. However, reliable, convenient and sensitive methods for exosome determination for early cancer diagnosis are still technically challenging. Herein, an electrochemical aptasensor based on click chemistry and the DNA hybridization chain reaction (HCR) for signal amplification has been developed for the ultrasensitive detection of tumor exosomes. CD63 aptamer was first immobilized on a glassy carbon electrode for capturing exosomes, and 4-oxo-2-nonenal alkyne (alkynyl-4-ONE) molecules, functionalized lipid electrophiles, were conjugated to the exosomes via the reaction of amino and aldehyde groups. Azide-labeled DNA probe as an anchor was then connected to the exosomes by copper (I)-catalyzed click chemistry. Signal amplification was achieved by HCR, and the numerous linked horseradish peroxidase (HRP) molecules could catalyze the reaction of o-phenylenediamine (OPD) and H2O2. The concentration of exosomes could be quantified by monitoring the electrochemical reduction current of 2,3-diaminophenazine (DAP). Under the optimal conditions, this method allowed the sensitive detection of exosomes in the range of 1.12 × 102 to 1.12 × 108 particles/μL with a limit of detection (LOD) of 96 particles/μL. Furthermore, the present assay enabled sensitive and accurate quantification of exosomes in human serum, and it has high potential for exosome analysis in clinical samples.
Collapse
Affiliation(s)
- Yu An
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Tongyu Jin
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Yuyuan Zhu
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Fan Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China.
| | - Pingang He
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| |
Collapse
|
994
|
Tian W, Liu S, Li B. Potential Role of Exosomes in Cancer Metastasis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4649705. [PMID: 31355262 PMCID: PMC6634128 DOI: 10.1155/2019/4649705] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
High cancer mortality is attributed to metastasis to a large extent. However, cancer metastasis remains devoid of dynamic monitoring and early prevention in terms of current advances in diagnostic means and therapeutic modalities. Meanwhile, studies have shown that reciprocal crosstalk among cells via exosomes plays a critical role in maintaining normal physiological state or triggering disease progression, including cancer metastasis. Therefore, in this review, we focus on the latest literature (primarily from 2018) to summarize action mechanisms and experimental studies of exosomes in cancer metastasis and put forward some problems as well as new outlooks of these studies.
Collapse
Affiliation(s)
- Wenjuan Tian
- Department of Clinical Laboratory, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shanshan Liu
- Department of Clinical Laboratory, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Burong Li
- Department of Clinical Laboratory, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| |
Collapse
|
995
|
Extracellular Microvesicles as New Industrial Therapeutic Frontiers. Trends Biotechnol 2019; 37:707-729. [DOI: 10.1016/j.tibtech.2018.11.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
|
996
|
Liu F, Hu S, Yang H, Li Z, Huang K, Su T, Wang S, Cheng K. Hyaluronic Acid Hydrogel Integrated with Mesenchymal Stem Cell-Secretome to Treat Endometrial Injury in a Rat Model of Asherman's Syndrome. Adv Healthc Mater 2019; 8:e1900411. [PMID: 31148407 PMCID: PMC7045702 DOI: 10.1002/adhm.201900411] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/25/2019] [Indexed: 12/31/2022]
Abstract
Stem cell therapies have made strides toward the efficacious treatment of injured endometrium and the prevention of intrauterine adhesions, or Asherman's syndrome (AS). Despite this progress, they are limited by their risk of tumor formation, low engraftment rates, as well as storage and transportation logistics. While attempts have been made to curb these issues, there remains a need for simple and effective solutions. A growing body of evidence supports the theory that delivering media, conditioned with mesenchymal stem cells, might be a promising alternative to live cell therapy. Mesenchymal stem cell-secretome (MSC-Sec) has a superior safety profile and can be stored without losing its regenerative properties. It is versatile enough to be added to a number of delivery vehicles that improve engraftment and control the release of the therapeutic. Thus, it holds great potential for the treatment of AS. Here, a new strategy for loading crosslinked hyaluronic acid gel (HA gel) with MSC-Sec is reported. The HA gel/MSC-Sec treatment paradigm creates a sustained release system that repairs endometrial injury in rats and promotes viable pregnancy.
Collapse
Affiliation(s)
- Feiran Liu
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Hua Yang
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Gynecology and Obstetrics, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Teng Su
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Shaowei Wang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| |
Collapse
|
997
|
Gao X, Li S, Ding F, Fan H, Shi L, Zhu L, Li J, Feng J, Zhu X, Zhang C. Rapid Detection of Exosomal MicroRNAs Using Virus‐Mimicking Fusogenic Vesicles. Angew Chem Int Ed Engl 2019; 58:8719-8723. [PMID: 31095853 DOI: 10.1002/anie.201901997] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/15/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Xihui Gao
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South CampusShanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South CampusSouthern Medical University Affiliated Fengxian Hospital 6600th Nanfeng Road, Fenxian District Shanghai 201499 China
| | - Sha Li
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South CampusShanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South CampusSouthern Medical University Affiliated Fengxian Hospital 6600th Nanfeng Road, Fenxian District Shanghai 201499 China
- Medical CollegeAnhui University of Science and Technology 168th Taifeng Road Huainan 232001 China
| | - Fei Ding
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Hongjia Fan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South CampusShanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South CampusSouthern Medical University Affiliated Fengxian Hospital 6600th Nanfeng Road, Fenxian District Shanghai 201499 China
| | - Leilei Shi
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Lijuan Zhu
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Jing Li
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South CampusShanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South CampusSouthern Medical University Affiliated Fengxian Hospital 6600th Nanfeng Road, Fenxian District Shanghai 201499 China
| | - Jing Feng
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South CampusShanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South CampusSouthern Medical University Affiliated Fengxian Hospital 6600th Nanfeng Road, Fenxian District Shanghai 201499 China
| | - Xinyuan Zhu
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Chuan Zhang
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| |
Collapse
|
998
|
Xiao Y, Zheng L, Zou X, Wang J, Zhong J, Zhong T. Extracellular vesicles in type 2 diabetes mellitus: key roles in pathogenesis, complications, and therapy. J Extracell Vesicles 2019; 8:1625677. [PMID: 31258879 PMCID: PMC6586118 DOI: 10.1080/20013078.2019.1625677] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 05/15/2019] [Accepted: 05/28/2019] [Indexed: 01/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), a chronic disease, is widely prevalent all over the world. In recent years, the roles of some extracellular vesicles (EVs) in T2DM have attracted much attention. EVs are bilayer membrane vesicles secreted from most cells and can participate in regulating various physiological and pathological processes in vivo by being transported between cells. Recently, it was discovered that some abnormal EVs can contribute to the occurrence of T2DM by inducing insulin resistance and can also participate in the complications of T2DM. In addition, some stem/progenitor cells-derived EVs have a potential application in the therapy of T2DM. This review introduces basic concepts of EVs and summarizes the roles of EVs in the pathogenesis, complications, and therapy of T2DM.
Collapse
Affiliation(s)
- Yongwei Xiao
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Lei Zheng
- Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofeng Zou
- Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jigang Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianing Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
999
|
Zhu G, Pei L, Lin F, Yin H, Li X, He W, Liu N, Gou X. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. J Cell Physiol 2019; 234:23736-23749. [PMID: 31180587 DOI: 10.1002/jcp.28941] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 02/05/2023]
Abstract
Renal ischemia/reperfusion (I/R) injury is the main reason for acute kidney injury (AKI) and is closely related to high morbidity and mortality. In this study, we found that exosomes from human-bone-marrow-derived mesenchymal stem cells (hBMSC-Exos) play a protective role in hypoxia/reoxygenation (H/R) injury. hBMSC-Exos were enriched in miR-199a-3p, and hBMSC-Exo treatment increased the expression level of miR-199a-3p in renal cells. We further explored the function of miR-199a-3p on H/R injury. miR-199a-3p was knocked down in hBMSCs with a miR-199a-3p inhibitor. HK-2 cells cocultured with miR-199a-3p-knockdown hBMSCs were more susceptible to H/R injury and showed more apoptosis than those cocultured with hBMSCs or miR-199a-3p-overexpressing hBMSCs. Meanwhile, we found that HK-2 cells exposed to H/R treatment incubated with hBMSC-Exos decreased semaphorin 3A (Sema3A) and activated the protein kinase B (AKT) and extracellular-signal-regulated kinase (ERK) pathways. However, HK-2 cells cocultured with miR-199a-3p-knockdown hBMSCs restored Sema3A expression and blocked the activation of the AKT and ERK pathways. Moreover, knocking down Sema3A could reactivate the AKT and ERK pathways suppressed by a miR-199a-3p inhibitor. In vivo, we injected hBMSC-Exos into mice suffering from I/R injury; this treatment induced functional recovery and histologic protection and reduced cleaved caspase-3 and Sema3A expression levels, as shown by immunohistochemistry. On the whole, this study demonstrated an antiapoptotic effect of hBMSC-Exos, which protected against I/R injury, via delivering miR-199a-3p to renal cells, downregulating Sema3A expression and thereby activating the AKT and ERK pathways. These findings reveal a novel mechanism of AKI treated with hBMSC-Exos and provide a therapeutic method for kidney diseases.
Collapse
Affiliation(s)
- Gongmin Zhu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijiao Pei
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Lin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hubin Yin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyuan Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nian Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
1000
|
Cao Y, Li L, Han B, Wang Y, Dai Y, Zhao J. A catalytic molecule machine-driven biosensing method for amplified electrochemical detection of exosomes. Biosens Bioelectron 2019; 141:111397. [PMID: 31200334 DOI: 10.1016/j.bios.2019.111397] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/22/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022]
Abstract
Nowadays, exosomes that carry abundant information have attracted increasing attention as potent biomarkers of liquid biopsy and ideal candidates for early diagnosis and treatment of cancers. In this work, we propose a "principle-of-proof" biosensing method for amplified electrochemical detection of exosomes by using HepG2-derived exosomes as models. Specifically, target exosomes are enriched on anti-CD63-functionalized immunobeads and then recognized by a DNA chain containing CD63 aptamer region, which subsequently initiates a catalytic molecule machine that relies on cascade toehold-mediated strand displacement reaction. Benefiting from high efficiency of the molecule machine, the method shows a linear range from 1 × 105 to 5 × 107 particles/mL and a detection limit of 1.72 × 104 particles/mL toward target exosomes, better than most existing detection methods. Moreover, the method demonstrates a high specificity even in serum samples and suggests a potential use in clinic, which may provide sufficient information for disease diagnosis, especially early detection and prognosis monitoring of tumors.
Collapse
Affiliation(s)
- Ya Cao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, 200444, PR China
| | - Lingling Li
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, 200444, PR China; Shanghai Key Laboratory of Bio-Energy Crops, Shanghai University, 200444, PR China
| | - Bing Han
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, 200444, PR China
| | - Ying Wang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, 200444, PR China
| | - Yuhao Dai
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, 200444, PR China
| | - Jing Zhao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, 200444, PR China.
| |
Collapse
|