1
|
Essola JM, Zhang M, Yang H, Li F, Xia B, Mavoungou JF, Hussain A, Huang Y. Exosome regulation of immune response mechanism: Pros and cons in immunotherapy. Bioact Mater 2024; 32:124-146. [PMID: 37927901 PMCID: PMC10622742 DOI: 10.1016/j.bioactmat.2023.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Due to its multiple features, including the ability to orchestrate remote communication between different tissues, the exosomes are the extracellular vesicles arousing the highest interest in the scientific community. Their size, established as an average of 30-150 nm, allows them to be easily uptaken by most cells. According to the type of cells-derived exosomes, they may carry specific biomolecular cargoes used to reprogram the cells they are interacting with. In certain circumstances, exosomes stimulate the immune response by facilitating or amplifying the release of foreign antigens-killing cells, inflammatory factors, or antibodies (immune activation). Meanwhile, in other cases, they are efficiently used by malignant elements such as cancer cells to mislead the immune recognition mechanism, carrying and transferring their cancerous cargoes to distant healthy cells, thus contributing to antigenic invasion (immune suppression). Exosome dichotomic patterns upon immune system regulation present broad advantages in immunotherapy. Its perfect comprehension, from its early biogenesis to its specific interaction with recipient cells, will promote a significant enhancement of immunotherapy employing molecular biology, nanomedicine, and nanotechnology.
Collapse
Affiliation(s)
- Julien Milon Essola
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences. Beijing 100049, PR China
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences. Beijing 100049, PR China
| | - Jacques François Mavoungou
- Université Internationale de Libreville, Libreville, 20411, Gabon
- Central and West African Virus Epidemiology, Libreville, 2263, Gabon
- Département de phytotechnologies, Institut National Supérieur d’Agronomie et de Biotechnologie, Université des Sciences et Techniques de Masuku, Franceville, 901, Gabon
- Institut de Recherches Agronomiques et Forestiers, Centre National de la Recherche Scientifique et du développement Technologique, Libreville, 16182, Gabon
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- Rigerna Therapeutics Co. Ltd., China
| |
Collapse
|
2
|
Wang HS, Lin S, Yu HM. Exosome-mediated Repair of Intervertebral Disc Degeneration: The Potential Role of miRNAs. Curr Stem Cell Res Ther 2024; 19:798-808. [PMID: 37150986 DOI: 10.2174/1574888x18666230504094233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 05/09/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a serious condition that manifests as low back pain, intervertebral disc protrusion, and spinal canal stenosis. At present, the main treatment methods for IVDD are surgical interventions such as discectomy, total disc replacement, and spinal fusion. However, these interventions have shown limitations, such as recurrent lumbar disc herniation after discectomy, lesions in adjacent segments, and failure of fixation. To overcome these shortcomings, researchers have been exploring stem cell transplantation therapy, such as mesenchymal stem cell (MSC) transplantation, but the treatment results are still controversial. Therefore, researchers are in search of new methods that are more efficient and have better outcomes. The exosomes from stem cells contain a variety of bioactive molecules that mediate cell interactions, and these components have been investigated for their potential therapeutic role in the repair of various tissue injuries. Recent studies have shown that MSC-derived miRNAs in exosomes and vesicles have therapeutic effects on nucleus pulposus cells, annulus fibrosus, and cartilage endplate. miRNAs play a role in many cell activities, such as cell proliferation, apoptosis, and cytokine release, by acting on mRNA translation, and they may have immense therapeutic potential, especially when combined with stem cell therapy. This article reviews the current status of research on intervertebral disc repair, especially with regard to the latest research findings on the molecular biological mechanisms of miRNAs in MSC-derived exosomes in intervertebral disc repair.
Collapse
Affiliation(s)
- Han-Shi Wang
- Department of Orthopaedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
| | - Hai-Ming Yu
- Department of Orthopaedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
3
|
Sadeghi MS, Lotfi M, Soltani N, Farmani E, Fernandez JHO, Akhlaghitehrani S, Mohammed SH, Yasamineh S, Kalajahi HG, Gholizadeh O. Recent advances on high-efficiency of microRNAs in different types of lung cancer: a comprehensive review. Cancer Cell Int 2023; 23:284. [PMID: 37986065 PMCID: PMC10661689 DOI: 10.1186/s12935-023-03133-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023] Open
Abstract
Carcinoma of the lung is among the most common types of cancer globally. Concerning its histology, it is categorized as a non-small cell carcinoma (NSCLC) and a small cell cancer (SCLC) subtype. MicroRNAs (miRNAs) are a member of non-coding RNA whose nucleotides range from 19 to 25. They are known to be critical regulators of cancer via epigenetic control of oncogenes expression and by regulating tumor suppressor genes. miRNAs have an essential function in a tumorous microenvironment via modulating cancer cell growth, metastasis, angiogenesis, metabolism, and apoptosis. Moreover, a wide range of information produced via several investigations indicates their tumor-suppressing, oncogenic, diagnostic assessment, and predictive marker functions in different types of lung malignancy. miRNA mimics or anti-miRNAs can be transferred into a lung cancer cell, with possible curative implications. As a result, miRNAs hold promise as targets for lung cancer treatment and detection. In this study, we investigate the different functions of various miRNAs in different types of lung malignancy, which have been achieved in recent years that show the lung cancer-associated regulation of miRNAs expression, concerning their function in lung cancer beginning, development, and resistance to chemotherapy, also the probability to utilize miRNAs as predictive biomarkers for therapy reaction.
Collapse
Affiliation(s)
- Mohammad Saleh Sadeghi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Lotfi
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Narges Soltani
- School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Lee CS, Lee M, Na K, Hwang HS. Stem Cell-Derived Extracellular Vesicles for Cancer Therapy and Tissue Engineering Applications. Mol Pharm 2023; 20:5278-5311. [PMID: 37867343 DOI: 10.1021/acs.molpharmaceut.3c00376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Recently, stem cells and their secretomes have attracted great attention in biomedical applications, particularly extracellular vesicles (EVs). EVs are secretomes of cells for cell-to-cell communication. They play a role as intercellular messengers as they carry proteins, nucleic acids, lipids, and therapeutic agents. They have also been utilized as drug-delivery vehicles due to their biocompatibility, low immunogenicity, stability, targetability, and engineerable properties. The therapeutic potential of EVs can be further enhanced by surface engineering and modification using functional molecules such as aptamers, peptides, and antibodies. As a consequence, EVs hold great promise as effective delivery vehicles for enhancing treatment efficacy while avoiding side effects. Among various cell types that secrete EVs, stem cells are ideal sources of EVs because stem cells have unique properties such as self-renewal and regenerative potential for transplantation into damaged tissues that can facilitate their regeneration. However, challenges such as immune rejection and ethical considerations remain significant hurdles. Stem cell-derived EVs have been extensively explored as a cell-free approach that bypasses many challenges associated with cell-based therapy in cancer therapy and tissue regeneration. In this review, we summarize and discuss the current knowledge of various types of stem cells as a source of EVs, their engineering, and applications of EVs, focusing on cancer therapy and tissue engineering.
Collapse
Affiliation(s)
- Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Kun Na
- Department of BioMedical-Chemical Engineering, The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
5
|
Ramesh D, Bakkannavar S, Bhat VR, Sharan K. Extracellular vesicles as novel drug delivery systems to target cancer and other diseases: Recent advancements and future perspectives. F1000Res 2023; 12:329. [PMID: 37868300 PMCID: PMC10589634 DOI: 10.12688/f1000research.132186.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 10/24/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bound vesicles produced into the extracellular space by cells. Apoptotic bodies (ApoBD), microvesicles (MVs), and exosomes are examples of EVs, which act as essential regulators in cell-cell communication in both normal and diseased conditions. Natural cargo molecules such as miRNA, messenger RNA, and proteins are carried by EVs and transferred to nearby cells or distant cells through the process of circulation. Different signalling cascades are then influenced by these functionally active molecules. The information to be delivered to the target cells depends on the substances within the EVs that also includes synthesis method. EVs have attracted interest as potential delivery vehicles for therapies due to their features such as improved circulation stability, biocompatibility, reduced immunogenicity, and toxicity. Therefore, EVs are being regarded as potent carriers of therapeutics that can be used as a therapeutic agent for diseases like cancer. This review focuses on the exosome-mediated drug delivery to cancer cells and the advantages and challenges of using exosomes as a carrier molecule.
Collapse
Affiliation(s)
- Divya Ramesh
- Forensic Medicine and Toxicology, Katsurba Medical College, Manipal, Manipal Academy of Higher Education, MAHE, Manipal, Karnataka, 576104, India
| | - Shankar Bakkannavar
- Forensic Medicine and Toxicology, Katsurba Medical College, Manipal, Manipal Academy of Higher Education, MAHE, Manipal, Karnataka, 576104, India
| | - Vinutha R Bhat
- Biochemistry, Katsurba Medical College, Manipal, Manipal Academy of Higher Education, MAHE, Manipal, Karnataka, 576104, India
| | - Krishna Sharan
- Radiotherapy Oncology, Katsurba Medical College, Manipal, Manipal Academy of Higher Education, MAHE, Manipal, Karnataka, 576104, India
| |
Collapse
|
6
|
Jimenez SA, Piera-Velazquez S. Probable role of exosomes in the extension of fibrotic alterations from affected to normal cells in systemic sclerosis. Rheumatology (Oxford) 2023; 62:999-1008. [PMID: 35944210 PMCID: PMC9977136 DOI: 10.1093/rheumatology/keac451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/14/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
SSc is a systemic autoimmune disease of unknown etiology characterized by frequently progressive cutaneous and internal organ fibrosis causing severe disability, organ failure and high mortality. A remarkable feature of SSc is the extension of the fibrotic alterations to nonaffected tissues. The mechanisms involved in the extension of fibrosis have remained elusive. We propose that this process is mediated by exosome microvesicles released from SSc-affected cells that induce an activated profibrotic phenotype in normal or nonaffected cells. Exosomes are secreted microvesicles involved in an intercellular communication system. Exosomes can transfer their macromolecular content to distant target cells and induce paracrine effects in the recipient cells, changing their molecular pathways and gene expression. Confirmation of this hypothesis may identify the molecular mechanisms responsible for extension of the SSc fibrotic process from affected cells to nonaffected cells and may allow the development of novel therapeutic approaches for the disease.
Collapse
Affiliation(s)
- Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and The Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and The Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Sridharan B, Lim HG. Exosomes and ultrasound: The future of theranostic applications. Mater Today Bio 2023; 19:100556. [PMID: 36756211 PMCID: PMC9900624 DOI: 10.1016/j.mtbio.2023.100556] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Biomaterials and pertaining formulations have been very successful in various diagnostic and therapeutic applications because of its ability to overcome pharmacological limitations. Some of them have gained significant focus in the recent decade for their theranostic properties. Exosomes can be grouped as biomaterials, since they consist of various biological micro/macromolecules and possess all the properties of a stable biomaterial with size in nano range. Significant research has gone into isolation and exploitation of exosomes as potential theranostic agent. However, the limitations in terms of yield, efficacy, and target specificity are continuously being addressed. On the other hand, several nano/microformulations are responsive to physical or chemical alterations and were successfully stimulated by tweaking the physical characteristics of the surrounding environment they are in. Some of them are termed as photodynamic, sonodynamic or thermodynamic therapeutic systems. In this regard, ultrasound and acoustic systems were extensively studied for its ability towards altering the properties of the systems to which they were applied on. In this review, we have detailed about the diagnostic and therapeutic applications of exosomes and ultrasound separately, consisting of their conventional applications, drawbacks, and developments for addressing the challenges. The information were categorized into various sections that provide complete overview of the isolation strategies and theranostic applications of exosomes in various diseases. Then the ultrasound-based disease diagnosis and therapy were elaborated, with special interest towards the use of ultrasound in enhancing the efficacy of nanomedicines and nanodrug delivery systems, Finally, we discussed about the ability of ultrasound in enhancing the diagnostic and therapeutic properties of exosomes, which could be the future of theranostics.
Collapse
Affiliation(s)
| | - Hae Gyun Lim
- Corresponding author. Biomedical Ultrasound Lab, Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
8
|
Piening LM, Wachs RA. Matrix-Bound Nanovesicles: What Are They and What Do They Do? Cells Tissues Organs 2023; 212:111-123. [PMID: 35168230 DOI: 10.1159/000522575] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/07/2022] [Indexed: 11/19/2022] Open
Abstract
Over the past 50 years, several different types of extracellular vesicles have been discovered including exosomes, microvesicles, and matrix vesicles. These vesicles are secreted by cells for specific purposes and contain cargo such as microRNA, cytokines, and lipids. A novel extracellular vesicle, the matrix-bound nanovesicle (MBV), has been recently discovered. The MBV is similar to the microvesicle, however, it is attached to the extracellular matrix, instead of being secreted. This review compares MBVs to other types of extracellular vesicles to try and better understand their origin and function. Further, this review will explain various extracellular vesicle isolation methods and how these can be used for MBVs and summarize characterization of MBV cargo such as microRNA, proteins, and lipids. Lastly, we will summarize the effects of MBVs on cells. MBVs are a novel class of extracellular vesicles that hold great promise as a platform for delivery of targeted gene and drug therapeutics.
Collapse
Affiliation(s)
- Logan M Piening
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Rebecca A Wachs
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
9
|
Lu S, Lu L, Liu Y, Li Z, Fang Y, Chen Z, Zhou J. Native and engineered extracellular vesicles for wound healing. Front Bioeng Biotechnol 2022; 10:1053217. [PMID: 36568307 PMCID: PMC9780283 DOI: 10.3389/fbioe.2022.1053217] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) that act as messengers mediate communication between parent and recipient cells through their contents, including nucleic acids, proteins, and lipids. These endogenous vesicles have emerged as a novel cell-free strategy for the treatment of diseases. EVs can be released by various types of cells with unique biological properties. Recent studies have shown that native EVs are used as therapeutic agents to promote tissue repair by delivering various growth factors and trophic factors including VEGF, EGF, TFN-α, IL-1β, and TGF-β to participate in all physiological processes of wound healing. Furthermore, to improve their specificity, safety, and efficiency for wound healing, the content and surface of EVs can be designed, modified, and engineered. The engineering strategies of EVs are divided into parent cell modification and indirect modification of EVs. The therapeutic potential of current EVs and engineered EVs for wound healing still requires the exploration of their large-scale clinical applications through innovative approaches. Herein, we provide an overview of the current biological knowledge about wound healing and EVs, as well as the application of native EVs in promoting wound healing. We also outline recent advances in engineering EV methodologies to achieve ideal therapeutic potential. Finally, the therapeutic applications of engineered EVs in wound healing are reviewed, and the challenges and prospects for the translation of engineered EVs to clinical applications are discussed.
Collapse
Affiliation(s)
- Shengli Lu
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Liping Lu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Liu
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Dermatology, Leiden University Medical Center, Leiden, Netherland
| | - Zenan Li
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Fang
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhizhao Chen
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jianda Zhou
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Sbarigia C, Vardanyan D, Buccini L, Tacconi S, Dini L. SARS-CoV-2 and extracellular vesicles: An intricate interplay in pathogenesis, diagnosis and treatment. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.987034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) are widely recognized as intercellular communication mediators. Among the different biological processes, EVs play a role in viral infections, supporting virus entrance and spread into host cells and immune response evasion. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection became an urgent public health issue with significant morbidity and mortality worldwide, being responsible for the current COVID-19 pandemic. Since EVs are implicated in SARS-CoV-2 infection in a morphological and functional level, they have gained growing interest for a better understanding of SARS-CoV-2 pathogenesis and represent possible diagnostic tools to track the disease progression. Furthermore, thanks to their biocompatibility and efficient immune activation, the use of EVs may also represent a promising strategy for the development of new therapeutic strategies against COVID-19. In this review, we explore the role of EVs in viral infections with a focus on SARS-CoV-2 biology and pathogenesis, considering recent morphometric studies. The common biogenesis aspects and structural similarities between EVs and SARS-CoV-2 will be examined, offering a panoramic of their multifaceted interplay and presenting EVs as a machinery supporting the viral cycle. On the other hand, EVs may be exploited as early diagnostic biomarkers and efficient carriers for drug delivery and vaccination, and ongoing studies will be reviewed to highlight EVs as potential alternative therapeutic strategies against SARS-CoV-2 infection.
Collapse
|
11
|
Haldavnekar R, Venkatakrishnan K, Tan B. Cancer Stem Cell Derived Extracellular Vesicles with Self-Functionalized 3D Nanosensor for Real-Time Cancer Diagnosis: Eliminating the Roadblocks in Liquid Biopsy. ACS NANO 2022; 16:12226-12243. [PMID: 35968931 DOI: 10.1021/acsnano.2c02971] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Liquid biopsy for determining the presence of cancer and the underlying tissue of origin is crucial to overcome the limitations of existing tissue biopsy and imaging-based techniques by capturing critical information from the dynamic tumor heterogeneity. A newly emerging liquid biopsy with extracellular vesicles (EVs) is gaining momentum, but its clinical relevance is in question due to the biological and technical challenges posed by existing technologies. The biological barriers of existing technologies include the inability to generate fundamental details of molecular structure, chemical composition as well as functional variations in EVs by gathering simultaneous information on multiple intra-EV molecules, unavailability of holistic qualitative analysis, in addition to the inability to identify tissue of origin. Technological barriers include reliance on EV isolation with a few labeled biomarkers, resulting in the inability to generate comprehensive information on the disease. A more favorable approach would be to generate holistic information on the disease without the use of labels. Such a marker-free diagnosis is impossible with the existing liquid biopsy due to the unavailability clinically validated cancer stem cells (CSC)-specific markers and dependence of existing technologies on EV isolation, undermining the clinical relevance of EV-based liquid biopsy. Here, CSC EVs were employed as an independent liquid biopsy modality. We hypothesize that tracking the signals of CSCs in peripheral blood with CSC EVs will provide a reliable solution for accurate cancer diagnosis, as CSC are the originators of tumor contributing to tumor heterogeneity. We report nanoengineered 3D sensors of extremely small nano-scaled probes self-functionalized for SERS, enabling integrative molecular and functional profiling of otherwise undetectable CSC EVs. A substantially enhanced SERS and ultralow limit of detection (10 EVs per 10 μL) were achieved. This was attributed to the efficient probe-EV interaction due to the 3D networks of nanoprobes, ensuring simultaneous detection of multiple EV signals. We experimentally demonstrate the crucial role of CSC EVs in cancer diagnosis. We then completed a pilot validation of this modality for cancer detection as well as for identification of the tissue of origin. An artificial neural network distinguished cancer from noncancer with 100% sensitivity and 100% specificity for three hard to detect cancers (breast, lung, and colorectal cancer). Binary classification to distinguish one tissue of origin against all other achieved 100% accuracy, while simultaneous identification of all three tissues of origin with multiclass classification achieved up to 79% accuracy. This noninvasive tool may complement existing cancer diagnostics, treatment monitoring as well as longitudinal disease monitoring by validation with a large cohort of clinical samples.
Collapse
Affiliation(s)
- Rupa Haldavnekar
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nanocharacterization Laboratory, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface Facility, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Krishnan Venkatakrishnan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nanocharacterization Laboratory, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface Facility, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Bo Tan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
- Nanocharacterization Laboratory, Faculty of Engineering and Architectural Sciences, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| |
Collapse
|
12
|
Padinharayil H, Varghese J, John MC, Rajanikant GK, Wilson CM, Al-Yozbaki M, Renu K, Dewanjee S, Sanyal R, Dey A, Mukherjee AG, Wanjari UR, Gopalakrishnan AV, George A. Non-small cell lung carcinoma (NSCLC): Implications on molecular pathology and advances in early diagnostics and therapeutics. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
13
|
Zhou H, Zhu L, Song J, Wang G, Li P, Li W, Luo P, Sun X, Wu J, Liu Y, Zhu S, Zhang Y. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol Cancer 2022; 21:86. [PMID: 35337361 PMCID: PMC8951719 DOI: 10.1186/s12943-022-01556-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and a leading cause of carcinogenic death. To date, surgical resection is regarded as the gold standard by the operator for clinical decisions. Because conventional tissue biopsy is invasive and only a small sample can sometimes be obtained, it is unable to represent the heterogeneity of tumor or dynamically monitor tumor progression. Therefore, there is an urgent need to find a new minimally invasive or noninvasive diagnostic strategy to detect CRC at an early stage and monitor CRC recurrence. Over the past years, a new diagnostic concept called “liquid biopsy” has gained much attention. Liquid biopsy is noninvasive, allowing repeated analysis and real-time monitoring of tumor recurrence, metastasis or therapeutic responses. With the advanced development of new molecular techniques in CRC, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), exosomes, and tumor-educated platelet (TEP) detection have achieved interesting and inspiring results as the most prominent liquid biopsy markers. In this review, we focused on some clinical applications of CTCs, ctDNA, exosomes and TEPs and discuss promising future applications to solve unmet clinical needs in CRC patients.
Collapse
Affiliation(s)
- Hui Zhou
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jun Song
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Guohui Wang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ping Luo
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jin Wu
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yunze Liu
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yi Zhang
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
14
|
Ashrafizadeh M, Kumar AP, Aref AR, Zarrabi A, Mostafavi E. Exosomes as Promising Nanostructures in Diabetes Mellitus: From Insulin Sensitivity to Ameliorating Diabetic Complications. Int J Nanomedicine 2022; 17:1229-1253. [PMID: 35340823 PMCID: PMC8943613 DOI: 10.2147/ijn.s350250] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus (DM) is among the chronic metabolic disorders that its incidence rate has shown an increase in developed and wealthy countries due to lifestyle and obesity. The treatment of DM has always been of interest, and significant effort has been made in this field. Exosomes belong to extracellular vesicles with nanosized features (30-150 nm) that are involved in cell-to-cell communication and preserving homeostasis. The function of exosomes is different based on their cargo, and they may contain lipids, proteins, and nucleic acids. The present review focuses on the application of exosomes in the treatment of DM; both glucose and lipid levels are significantly affected by exosomes, and these nanostructures enhance lipid metabolism and decrease its deposition. Furthermore, exosomes promote glucose metabolism and affect the level of glycolytic enzymes and glucose transporters in DM. Type I DM results from the destruction of β cells in the pancreas, and exosomes can be employed to ameliorate apoptosis and endoplasmic reticulum (ER) stress in these cells. The exosomes have dual functions in mediating insulin resistance/sensitivity, and M1 macrophage-derived exosomes inhibit insulin secretion. The exosomes may contain miRNAs, and by transferring among cells, they can regulate various molecular pathways such as AMPK, PI3K/Akt, and β-catenin to affect DM progression. Noteworthy, exosomes are present in different body fluids such as blood circulation, and they can be employed as biomarkers for the diagnosis of diabetic patients. Future studies should focus on engineering exosomes derived from sources such as mesenchymal stem cells to treat DM as a novel strategy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, 34956, Istanbul, Turkey
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Translational Sciences, Xsphera Biosciences Inc., Boston, MA, 02210, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
15
|
The Potential Application of Extracellular Vesicles from Liquid Biopsies for Determination of Pharmacogene Expression. Pharmaceuticals (Basel) 2022; 15:ph15020252. [PMID: 35215364 PMCID: PMC8879428 DOI: 10.3390/ph15020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Pharmacogenomics (PGx) entails the study of heritability of drug response. This may include both variability in genes related to pharmacokinetics (drug absorption, distribution, metabolism and excretion) and pharmacodynamics (e.g., drug receptors or signaling pathways). Individualizing drug therapy taking into account the genetic profile of the patient has the potential to make drug therapy safer and more effective. Currently, this approach relies on the determination of genetic variants in pharmacogenes by genotyping. However, it is widely acknowledged that large variability in gene expression is attributed to non-structural genetic variants. Therefore, at least from a theoretical viewpoint individualizing drug therapy based upon expression of pharmacogenes rather than on genotype may be advantageous but has been difficult to implement in the clinical setting. Extracellular vesicles (EVs) are lipid encapsulated structures that contain cargo such as lipids, nucleic acids and proteins. Since their cargo is tissue- and cell-specific they can be used to determine the expression of pharmacogenes in the liver. In this review, we describe methods of EV isolation and the potential of EVs isolated from liquid biopsies as a tool to determine the expression of pharmacogenes for use in personalized medicine.
Collapse
|
16
|
Praja RK, Wongwattanakul M, Tippayawat P, Phoksawat W, Jumnainsong A, Sornkayasit K, Leelayuwat C. Attenuated Total Reflectance-Fourier Transform Infrared (ATR-FTIR) Spectroscopy Discriminates the Elderly with a Low and High Percentage of Pathogenic CD4+ T Cells. Cells 2022; 11:458. [PMID: 35159268 PMCID: PMC8834052 DOI: 10.3390/cells11030458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023] Open
Abstract
In the aging process, the presence of interleukin (IL)-17-producing CD4+CD28-NKG2D+T cells (called pathogenic CD4+ T cells) is strongly associated with inflammation and the development of various diseases. Thus, their presence needs to be monitored. The emergence of attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy empowered with machine learning is a breakthrough in the field of medical diagnostics. This study aimed to discriminate between the elderly with a low percentage (LP; ≤3%) and a high percentage (HP; ≥6%) of pathogenic CD4+CD28-NKG2D+IL17+ T cells by utilizing ATR-FTIR coupled with machine learning algorithms. ATR spectra of serum, exosome, and HDL from both groups were explored in this study. Only exosome spectra in the 1700-1500 cm-1 region exhibited possible discrimination for the LP and HP groups based on principal component analysis (PCA). Furthermore, partial least square-discriminant analysis (PLS-DA) could differentiate both groups using the 1700-1500 cm-1 region of exosome ATR spectra with 64% accuracy, 69% sensitivity, and 61% specificity. To obtain better classification performance, several spectral models were then established using advanced machine learning algorithms, including J48 decision tree, support vector machine (SVM), random forest (RF), and neural network (NN). Herein, NN was considered to be the best model with an accuracy of 100%, sensitivity of 100%, and specificity of 100% using serum spectra in the region of 1800-900 cm-1. Exosome spectra in the 1700-1500 and combined 3000-2800 and 1800-900 cm-1 regions using the NN algorithm gave the same accuracy performance of 95% with a variation in sensitivity and specificity. HDL spectra with the NN algorithm also showed excellent test performance in the 1800-900 cm-1 region with 97% accuracy, 100% sensitivity, and 95% specificity. This study demonstrates that ATR-FTIR coupled with machine learning algorithms can be used to study immunosenescence. Furthermore, this approach can possibly be applied to monitor the presence of pathogenic CD4+ T cells in the elderly. Due to the limited number of samples used in this study, it is necessary to conduct a large-scale study to obtain more robust classification models and to assess the true clinical diagnostic performance.
Collapse
Affiliation(s)
- Rian Ka Praja
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen 40002, Thailand;
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (M.W.); (P.T.); (A.J.); (K.S.)
| | - Molin Wongwattanakul
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (M.W.); (P.T.); (A.J.); (K.S.)
| | - Patcharaporn Tippayawat
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (M.W.); (P.T.); (A.J.); (K.S.)
- Department of Clinical Microbiology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wisitsak Phoksawat
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Research and Diagnostic Center for Emerging Infectious Diseases, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Amonrat Jumnainsong
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (M.W.); (P.T.); (A.J.); (K.S.)
- Department of Clinical Immunology and Transfusion Sciences, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Kanda Sornkayasit
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (M.W.); (P.T.); (A.J.); (K.S.)
| | - Chanvit Leelayuwat
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (M.W.); (P.T.); (A.J.); (K.S.)
- Department of Clinical Immunology and Transfusion Sciences, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
17
|
Alghamdi M, Alamry SA, Bahlas SM, Uversky VN, Redwan EM. Circulating extracellular vesicles and rheumatoid arthritis: a proteomic analysis. Cell Mol Life Sci 2021; 79:25. [PMID: 34971426 PMCID: PMC11072894 DOI: 10.1007/s00018-021-04020-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Circulating extracellular vesicles (EVs) are membrane-bound nanoparticles secreted by most cells for intracellular communication and transportation of biomolecules. EVs carry proteins, lipids, nucleic acids, and receptors that are involved in human physiology and pathology. EV cargo is variable and highly related to the type and state of the cellular origin. Three subtypes of EVs have been identified: exosomes, microvesicles, and apoptotic bodies. Exosomes are the smallest and the most well-studied class of EVs that regulate different biological processes and participate in several diseases, such as cancers and autoimmune diseases. Proteomic analysis of exosomes succeeded in profiling numerous types of proteins involved in disease development and prognosis. In rheumatoid arthritis (RA), exosomes revealed a potential function in joint inflammation. These EVs possess a unique function, as they can transfer specific autoantigens and mediators between distant cells. Current proteomic data demonstrated that exosomes could provide beneficial effects against autoimmunity and exert an immunosuppressive action, particularly in RA. Based on these observations, effective therapeutic strategies have been developed for arthritis and other inflammatory disorders.
Collapse
Affiliation(s)
- Mohammed Alghamdi
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
- Laboratory Department, University Medical Services Center, King Abdulaziz University, P.O. Box 80200, Jeddah, 21589, Saudi Arabia
| | - Sultan Abdulmughni Alamry
- Immunology Diagnostic Laboratory Department, King Abdulaziz University Hospital, P.O Box 80215, Jeddah, 21589, Saudi Arabia
| | - Sami M Bahlas
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, P.O. Box 80215, Jeddah, 21589, Saudi Arabia
| | - Vladimir N Uversky
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Elrashdy M Redwan
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia.
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, 21934, Alexandria, Egypt.
| |
Collapse
|
18
|
Zhang X, Liu D, Gao Y, Lin C, An Q, Feng Y, Liu Y, Liu D, Luo H, Wang D. The Biology and Function of Extracellular Vesicles in Cancer Development. Front Cell Dev Biol 2021; 9:777441. [PMID: 34805181 PMCID: PMC8602830 DOI: 10.3389/fcell.2021.777441] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) exert their biological functions by delivering proteins, metabolites, and nucleic acids to recipient cells. EVs play important roles in cancer development. The anti-tumor effect of EVs is by their cargos carrying proteins, metabolites, and nucleic acids to affect cell-to-cell communication. The characteristics of cell-to-cell communication can potentially be applied for the therapy of cancers, such as gastric cancer. In addition, EVs can be used as an effective cargos to deliver ncRNAs, peptides, and drugs, to target tumor tissues. In addition, EVs have the ability to regulate cell apoptosis, autophagy, proliferation, and migration of cancer cells. The ncRNA and peptides that were engaged with EVs were associated with cell signaling pathways in cancer development. This review focuses on the composition, cargo, function, mechanism, and application of EVs in cancers.
Collapse
Affiliation(s)
- Xinyi Zhang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Dianfeng Liu
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Yongjian Gao
- Department of Hepatobiliary and Pancreas Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chao Lin
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, China.,School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Qingwu An
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Ye Feng
- Department of Hepatobiliary and Pancreas Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yangyang Liu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Da Liu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Haoming Luo
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
19
|
Role of Extracellular Vesicle-Based Cell-to-Cell Communication in Multiple Myeloma Progression. Cells 2021; 10:cells10113185. [PMID: 34831408 PMCID: PMC8625088 DOI: 10.3390/cells10113185] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/04/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma (MM) progression closely depends on the bidirectional crosstalk between tumor cells and the surrounding microenvironment, which leads to the creation of a tumor supportive niche. Extracellular vesicles (EVs) have emerged as key players in the pathological interplay between the malignant clone and near/distal bone marrow (BM) cells through their biologically active cargo. Here, we describe the role of EVs derived from MM and BM cells in reprogramming the tumor microenvironment and in fostering bone disease, angiogenesis, immunosuppression, drug resistance, and, ultimately, tumor progression. We also examine the emerging role of EVs as new therapeutic agents for the treatment of MM, and their potential use as clinical biomarkers for early diagnosis, disease classification, and therapy monitoring.
Collapse
|
20
|
Liu J, Ren L, Li S, Li W, Zheng X, Yang Y, Fu W, Yi J, Wang J, Du G. The biology, function, and applications of exosomes in cancer. Acta Pharm Sin B 2021; 11:2783-2797. [PMID: 34589397 PMCID: PMC8463268 DOI: 10.1016/j.apsb.2021.01.001] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/30/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
Exosomes are cell-derived nanovesicles with diameters from 30 to 150 nm, released upon fusion of multivesicular bodies with the cell surface. They can transport nucleic acids, proteins, and lipids for intercellular communication and activate signaling pathways in target cells. In cancers, exosomes may participate in growth and metastasis of tumors by regulating the immune response, blocking the epithelial-mesenchymal transition, and promoting angiogenesis. They are also involved in the development of resistance to chemotherapeutic drugs. Exosomes in liquid biopsies can be used as non-invasive biomarkers for early detection and diagnosis of cancers. Because of their amphipathic structure, exosomes are natural drug delivery vehicles for cancer therapy.
Collapse
Key Words
- ABCA3, ATP-binding cassette transporter A3
- APCs, antigen-presenting cells
- Biomarkers
- CAFs, cancer-associated fibroblasts
- CCRCC, clear-cell renal cell carcinoma
- CD-UPRT, cytosine deaminase-uracil phosphoribosyltransferase
- CDH3, cadherin 3
- CRC, colorectal cancer
- DC, dendritic cells
- DEXs, DC-derived exosomes
- DLBCL, diffuse large B-cell lymphoma
- DNM3, dynamin 3
- Del-1, developmental endothelial locus-1
- Drug delivery
- Drug resistance
- ECM, extracellular matrix
- EMT, epithelial–mesenchymal transition
- ESCRT, endosomal sorting complex required for transport
- Exosomes
- GPC1, glypican-1
- HA, hyaluronic acid
- HCC, hepatocellular carcinoma
- HIF1, hypoxia-inducible factor 1
- HTR, hormone therapy-resistant
- HUVECs, human umbilical vein endothelial cells
- ILVs, intraluminal vesicles
- MDSCs, myeloid-derived suppressor cells
- MIF, migration inhibitory factor
- MSC, mesenchymal stem cells
- MVB, multivesicular body
- NKEXOs, natural killer cell-derived exosomes
- NNs, nanoparticles
- NSCLC, non-small cell lung cancer
- PA, phosphatidic acid
- PCC, pheochromocytoma
- PD-L1, programmed cell death receptor ligand 1
- PDAC, pancreatic ductal adenocarcinoma
- PGL, paraganglioma
- PI, phosphatidylinositol
- PS, phosphatidylserine
- PTRF, polymerase I and transcript release factor
- RCC, renal cell carcinoma
- SM, sphingomyelin
- SNARE, soluble NSF-attachment protein receptor
- TEX, tumor-derived exosomes
- TSG101, tumor susceptibility gene 101
- Tumor immunity
- Tumor metastasis
- circRNAs, circular RNAs
- dsDNA, double stranded DNA
- hTERT, human telomerase reverse transcriptase
- lamp2b, lysosome-associated membrane glycoprotein 2b
- lncRNAs, long non-coding RNAs
- miRNA, microRNA
- mtDNA, mitochondrial DNA
- ncRNA, non-coding RNAs
Collapse
Affiliation(s)
- Jinyi Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Sha Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiangjin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yihui Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Weiqi Fu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jie Yi
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Beijing 100730, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
21
|
The Fatty Acid and Protein Profiles of Circulating CD81-Positive Small Extracellular Vesicles Are Associated with Disease Stage in Melanoma Patients. Cancers (Basel) 2021; 13:cancers13164157. [PMID: 34439311 PMCID: PMC8392159 DOI: 10.3390/cancers13164157] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Early detection of cutaneous melanoma is the key to increasing survival and proper therapeutic adjustment, especially in stages II–IV. We investigated whether the fatty acid (FA) and protein compositions of small extracellular vesicles (sEV) expressing CD81, derived from the plasma of stage 0–I, II and III–IV melanoma patients, could reflect disease stage. Results showed a higher content of FA and differences in C18:0/C18:1 ratio, a marker of cell membrane fluidity, that distinguished patients’ CD81sEV from those of healthy donors (HD). By proteomic analysis (identifier PXD024434) we identified significant increases in CD14, PON1, PON3 and APOA5 in stage II CD81sEV compared to HD. In stage III–IV, CD81sEV’ RAP1B expression was decreased. These stage-related signatures may support the potential of sEV to provide information for early diagnosis, prediction of metastatic behavior, treatment and follow-up of melanoma patients. Abstract The early detection of cutaneous melanoma, a potentially lethal cancer with rising incidence, is fundamental to increasing survival and therapeutic adjustment. In stages II–IV especially, additional indications for adjuvant therapy purposes after resection and for treatment of metastatic patients are urgently needed. We investigated whether the fatty acid (FA) and protein compositions of small extracellular vesicles (sEV) derived from the plasma of stage 0–I, II and III–IV melanoma patients (n = 38) could reflect disease stage. The subpopulation of sEV expressing CD81 EV marker (CD81sEV) was captured by an ad hoc immune affinity technique from plasma depleted of large EV. Biological macromolecules were investigated by gas chromatography and mass spectrometry in CD81sEV. A higher content of FA was detectable in patients with respect to healthy donors (HD). Moreover, a higher C18:0/C18:1 ratio, as a marker of cell membrane fluidity, distinguished early (stage 0–I) from late (III–IV) stages’ CD81sEV. Proteomics detected increases in CD14, PON1, PON3 and APOA5 exclusively in stage II CD81sEV, and RAP1B was decreased in stage III–IV CD81sEV, in comparison to HD. Our results suggest that stage dependent alterations in CD81sEV’ FA and protein composition may occur early after disease onset, strengthening the potential of circulating sEV as a source of discriminatory information for early diagnosis, prediction of metastatic behavior and following up of melanoma patients.
Collapse
|
22
|
Al-Dossary AA, Tawfik EA, Isichei AC, Sun X, Li J, Alshehri AA, Alomari M, Almughem FA, Aldossary AM, Sabit H, Almalik AM. Engineered EV-Mimetic Nanoparticles as Therapeutic Delivery Vehicles for High-Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13123075. [PMID: 34203051 PMCID: PMC8234974 DOI: 10.3390/cancers13123075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In this review, we begin with the role of natural extracellular vesicles (EVs) in high-grade serous ovarian cancer (HGSOC). Then, we narrow our focus on the advantages of using EV-mimetic nanoparticles as a delivery vehicle for RNAi therapy and other chemotherapeutics. Furthermore, we discuss the challenges of the clinical translation of engineering EV mimetic drug delivery systems and the promising directions of further development. Abstract High-grade serous ovarian cancer (HGSOC) is the most lethal gynecological malignancy among women. Several obstacles impede the early diagnosis and effective treatment options for ovarian cancer (OC) patients, which most importantly include the development of platinum-drug-resistant strains. Currently, extensive efforts are being put into the development of strategies capable of effectively circumventing the physical and biological barriers present in the peritoneal cavity of metastatic OC patients, representing a late stage of gastrointestinal and gynecological cancer with an extremely poor prognosis. Naturally occurring extracellular vesicles (EVs) have been shown to play a pivotal role in progression of OC and are now being harnessed as a delivery vehicle for cancer chemotherapeutics. However, there are limitations to their clinical application due to current challenges in their preparation techniques. Intriguingly, there is a recent drive towards the use of engineered synthetic EVs for the delivery of chemotherapeutics and RNA interference therapy (RNAi), as they show the promise of overcoming the obstacles in the treatment of OC patients. This review discusses the therapeutic application of EVs in OC and elucidates the potential use of engineered EV-mimetic nanoparticles as a delivery vehicle for RNAi therapy and other chemotherapeutics, which would potentially improve clinical outcomes of OC patients.
Collapse
Affiliation(s)
- Amal A. Al-Dossary
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 34212, Saudi Arabia;
- Correspondence: ; Tel.: +966-1-333-31137
| | - Essam A. Tawfik
- National Center for Pharmaceutical Technology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (E.A.T.); (A.A.A.); (F.A.A.); (A.M.A.)
| | - Adaugo C. Isichei
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 34212, Saudi Arabia;
| | - Xin Sun
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA; (X.S.); (J.L.)
| | - Jiahe Li
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA; (X.S.); (J.L.)
| | - Abdullah A. Alshehri
- National Center for Pharmaceutical Technology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (E.A.T.); (A.A.A.); (F.A.A.); (A.M.A.)
| | - Munther Alomari
- Department of Stem Cell Biology, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Fahad A. Almughem
- National Center for Pharmaceutical Technology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (E.A.T.); (A.A.A.); (F.A.A.); (A.M.A.)
| | - Ahmad M. Aldossary
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia;
| | - Hussein Sabit
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Abdulaziz M. Almalik
- National Center for Pharmaceutical Technology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (E.A.T.); (A.A.A.); (F.A.A.); (A.M.A.)
| |
Collapse
|
23
|
Bhat EA, Sajjad N, Thokar FM. Current advancement of exosomes as biomarkers for cancer diagnosis and forecasting. Cancer Treat Res Commun 2021; 28:100417. [PMID: 34126578 DOI: 10.1016/j.ctarc.2021.100417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/27/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Exosomes are normal vesicles produced in the late endosomes of a cell. They are secreted by cells and play a role in cell-to-cell contact. They are an invaluable aid in cancer diagnosis as they include miRNA, proteins and lncRNAs. Depending on the function of these constituents in cancer, the expression of exosome constituents can be upregulated or downregulated in cancer. Exosomes provide high concentration and protective environment for their cargo, thereby making them superior targets for cancer diagnosis. It has recently been documented that exosomes modulate cell-cell connectivity by molecules included in the exosomes, leading to the maintenance of tissue homeostasis. In addition, exosomes released from cancer cells are implicated in the development of cancer. Data on the role of exosomes in cancer will thus enhance the effectiveness of new diagnostic and therapeutic approaches. In particular, exosomes are useful sources for biomarkers due to selective cargo loading and similarity to their parental cells. In this review, we summarize the recent findings to use exosomes as cancer biomarkers for early detection, diagnosis, and therapy selection.
Collapse
Affiliation(s)
- Eijaz Ahmed Bhat
- Life sciences institute, Zhejiang University, Hangzhou, Zhejiang, 310058, P.R. China; Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India.
| | - Nasreena Sajjad
- Department of Biochemistry, University of Kashmir, Hazratbal, Jammu and Kashmir, India
| | - Fahd M Thokar
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, UP 202002, India
| |
Collapse
|
24
|
Hassanzadeh A, Rahman HS, Markov A, Endjun JJ, Zekiy AO, Chartrand MS, Beheshtkhoo N, Kouhbanani MAJ, Marofi F, Nikoo M, Jarahian M. Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res Ther 2021; 12:297. [PMID: 34020704 PMCID: PMC8138094 DOI: 10.1186/s13287-021-02378-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, mesenchymal stem/stromal cells (MSCs) and their widespread biomedical applications have attracted great consideration from the scientific community around the world. However, reports have shown that the main populations of the transplanted MSCs are trapped in the liver, spleen, and lung upon administration, highlighting the importance of the development of cell-free therapies. Concerning rising evidence suggesting that the beneficial effects of MSC therapy are closely linked to MSC-released components, predominantly MSC-derived exosomes, the development of an MSC-based cell-free approach is of paramount importance. The exosomes are nano-sized (30100nm) lipid bilayer membrane vesicles, which are typically released by MSCs and are found in different body fluids. They include various bioactive molecules, such as messenger RNA (mRNA), microRNAs, proteins, and bioactive lipids, thus showing pronounced therapeutic competence for tissues recovery through the maintenance of their endogenous stem cells, the enhancement of regenerative phenotypic traits, inhibition of apoptosis concomitant with immune modulation, and stimulation of the angiogenesis. Conversely, the specific roles of MSC exosomes in the treatment of various tumors remain challenging. The development and clinical application of novel MSC-based cell-free strategies can be supported by better understanding their mechanisms, classifying the subpopulation of exosomes, enhancing the conditions of cell culture and isolation, and increasing the production of exosomes along with engineering exosomes to deliver drugs and therapeutic molecules to the target sites. In the current review, we deliver a brief overview of MSC-derived exosome biogenesis, composition, and isolation methods and discuss recent investigation regarding the therapeutic potential of MSC exosomes in regenerative medicine accompanied by their double-edged sword role in cancer.
Collapse
Affiliation(s)
- Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Sulaymaniyah, Iraq
| | | | - Judi Januadi Endjun
- Medical Faculty, UPN Veteran, Jakarta, Indonesia.,Gatot Soebroto Indonesia Army Hospital, Jakarta, Indonesia
| | | | | | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120, Heidelberg, Germany.
| |
Collapse
|
25
|
Mirzaei R, Babakhani S, Ajorloo P, Ahmadi RH, Hosseini-Fard SR, Keyvani H, Ahmadyousefi Y, Teimoori A, Zamani F, Karampoor S, Yousefimashouf R. The emerging role of exosomal miRNAs as a diagnostic and therapeutic biomarker in Mycobacterium tuberculosis infection. Mol Med 2021; 27:34. [PMID: 33794771 PMCID: PMC8017856 DOI: 10.1186/s10020-021-00296-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), has been the world's driving fatal bacterial contagious disease globally. It continues a public health emergency, and around one-third of the global community has been affected by latent TB infection (LTBI). This is mostly due to the difficulty in diagnosing and treating patients with TB and LTBI. Exosomes are nanovesicles (40-100 nm) released from different cell types, containing proteins, lipids, mRNA, and miRNA, and they allow the transfer of one's cargo to other cells. The functional and diagnostic potential of exosomal miRNAs has been demonstrated in bacterial infections, including TB. Besides, it has been recognized that cells infected by intracellular pathogens such as Mtb can be secreting an exosome, which is implicated in the infection's fate. Exosomes, therefore, open a unique viewpoint on the investigative process of TB pathogenicity. This study explores the possible function of exosomal miRNAs as a diagnostic biomarker. Moreover, we include the latest data on the pathogenic and therapeutic role of exosomal miRNAs in TB.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Sajad Babakhani
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Parisa Ajorloo
- Department of Biology, Sciences and Research Branch, Islamic Azad University, Tehran, Iran
| | - Razieh Heidari Ahmadi
- Department of Genetics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran.,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Teimoori
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
26
|
Progress of exosomes in the diagnosis and treatment of lung cancer. Biomed Pharmacother 2020; 134:111111. [PMID: 33352449 DOI: 10.1016/j.biopha.2020.111111] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
The incidence and mortality of lung cancer account for first place all over the world. Lung cancer lacks early diagnostic biomarkers; lung cancer patients are usually diagnosed in both middle and advanced stages and have poor treatment outcomes. It is more important to find the first diagnostic tools for lung cancer with high specificity and sensitivity. Besides, exosomes are usually nanometer-sized bi-layered lipid vesicles formed and produced by various types of cells. As one of the main modes of intercellular communication, they can deliver multiple functional biomolecules, such as DNA, microRNAs, messenger RNA (mRNA), long non-coding RNA, and proteins, and the events as mentioned above affects different physiological processes of recipient cells. It has been reported that exosomes are involved in different types of cancer, including lung cancer. Various studies proved that exosomes are involved in multiple cancer processes such as cell proliferation, metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, and the tumor microenvironment in lung cancer. Tumor-derived exosomes (TEX) contain a variety of stimulatory and inhibitory factors involved in regulating immune response, which can affect the tumor microenvironment (TME) and thus participate in the formation and progression of lung cancer. This review's primary purpose to review the latest research progress of exosomes in diagnosing and treating lung cancer.
Collapse
|
27
|
Exosomes: Cell-Derived Nanoplatforms for the Delivery of Cancer Therapeutics. Int J Mol Sci 2020; 22:ijms22010014. [PMID: 33374978 PMCID: PMC7792591 DOI: 10.3390/ijms22010014] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022] Open
Abstract
Exosomes are cell-secreted nanovesicles that naturally contain biomolecular cargoes such as lipids, proteins, and nucleic acids. Exosomes mediate intercellular communication, enabling the transfer biological signals from the donor cells to the recipient cells. Recently, exosomes are emerging as promising drug delivery vehicles due to their strong stability in blood circulation, high biocompatibility, low immunogenicity, and natural targeting ability. In particular, exosomes derived from specific types of cells can carry endogenous signaling molecules with therapeutic potential for cancer treatment, thus presenting a significant impact on targeted drug delivery and therapy. Furthermore, exosomes can be engineered to display targeting moieties on their surface or to load additional therapeutic agents. Therefore, a comprehensive understanding of exosome biogenesis and the development of efficient exosome engineering techniques will provide new avenues to establish convincing clinical therapeutic strategies based on exosomes. This review focuses on the therapeutic applications of exosomes derived from various cells and the exosome engineering technologies that enable the accurate delivery of various types of cargoes to target cells for cancer therapy.
Collapse
|
28
|
Abstract
Exosomes are defined as a type of extracellular vesicle released when multivesicular bodies of the endocytic pathway fuse with the plasma membrane. They are characterized by their role in extracellular communication, partly due to their composition, and present the ability to recognize and interact with cells from the immune system, enabling an immune response. Their targeting capability and nanosized dimensions make them great candidates for cancer therapy. As chemotherapy is associated with cytotoxicity and multiple drug resistance, the use of exosomes targeting capabilities, able to deliver anticancer drugs specifically to cancer cells, is a great approach to overcome these disadvantages. The objective is to assess treatment efficiency in reducing tumor cells, as well as overall safety and response by cancer carriers. So far, results show exosomes as a promising therapeutic strategy in the fight against cancer. This review summarizes the characteristics and composition of exosomes, as well as explaining in detail the involved parties in the origin of exosomes. Furthermore, some considerations about exosome application in immunotherapy are addressed. The main isolation and loading methods are described to give an insight into how exosomes can be obtained and manipulated. Finally, some therapeutic applications of exosomes in cancer therapy are described.
Collapse
|
29
|
Peng H, Ji W, Zhao R, Yang J, Lu Z, Li Y, Zhang X. Exosome: a significant nano-scale drug delivery carrier. J Mater Chem B 2020; 8:7591-7608. [PMID: 32697267 DOI: 10.1039/d0tb01499k] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years, due to the limitations of the nature of therapeutic agents, many synthetic nano-delivery systems have emerged to enhance the efficacy of drugs. Extracellular vesicles are currently a class of natural nano-scale drug carriers released by cells. As a tiny vesicle with a lipid bilayer membrane that can be secreted by most cells in the body, exosomes carry and transmit important signal molecules, Therefore, they have been a research hotspot in biomedicine and biomaterials due to their size advantages and huge potential in drug therapy. Many people are optimistic about the clinical application prospects of exosomes and are actively exploring the broad functions of exosomes and developing exosome therapeutic agents to make positive contributions to human health. In this review, we provide basic knowledge and focus on summarizing the advantages of exosomes as drug carriers, methods of loading drugs, targeting strategies, in vivo and in vitro tracing methods, and some of the latest developments in exosomes as drug carriers. In particular, the review provides an outlook for this field.
Collapse
Affiliation(s)
- Huan Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | | | | | | | | | | | | |
Collapse
|
30
|
Xie H, Di K, Huang R, Khan A, Xia Y, Xu H, Liu C, Tan T, Tian X, Shen H, He N, Li Z. Extracellular vesicles based electrochemical biosensors for detection of cancer cells: A review. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.02.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
31
|
Exosomes: Multiple-targeted multifunctional biological nanoparticles in the diagnosis, drug delivery, and imaging of cancer cells. Biomed Pharmacother 2020; 129:110442. [PMID: 32593129 DOI: 10.1016/j.biopha.2020.110442] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
Exosomes are biological nanoparticles (30-150 nm) secreted in the extracellular area from all of cells, that mediate intercellular message. Exosomes act as the carriers for numerous proteins, DNAs, RNAs and cell-signaling molecules. Therefore, exosomes secreted by the tumor cells are useful for diagnostic purposes because of their persistent presence in the blood and their provision of genetic cargo similar to those in tumor. Due to the risks of aggressive activity and ambiguity of biological activity in other tissues, the use of exosomes in drug delivery and imaging has been limited. However, their high loading, stability and longer circulation time, excellent targeting, high cell penetration performance, and optimal biodegradability have made them potential agents in targeted cancer treatment. Therefore, in addition to examining methods for isolating and loading exosomes, this paper discusses the applications of exosomes in biological measurement, imaging, and therapeutic activities. Also, this review describes the challenges of using exosomes compared to conventional methods and shows that it is very useful to use them due to less aggressive activities. Finally, this review attempts to provide an appropriate incentive by showing the performance of exosomes in cancer therapy through targeted drug delivery, gene therapy, imaging and diagnosis.
Collapse
|
32
|
Shao B, Xiao Z. Recent achievements in exosomal biomarkers detection by nanomaterials-based optical biosensors - A review. Anal Chim Acta 2020; 1114:74-84. [PMID: 32359518 DOI: 10.1016/j.aca.2020.02.041] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 01/07/2023]
Abstract
Exosomal biomarkers including tumor-derived exosomes, exosomal surface proteins and exosomal nucleic acids have emerged as one of the most important and general cancer biomarkers in modern biomedical science. These indicators can provide momentous biological information for early diagnosis and treatment of cancer. Recently, numerous studies have been conducted to design biosensors for exosomal biomarkers detection and profiling with high sensitivity and strong applied ability. Among these biosensors, nanomaterial-based optical biosensors are prospective future platforms for rapid and cost-effective detection of exosomal biomarkers. Firstly, we have focused on the progress and advancements in different optical-transducing approaches (Surface-Enhanced Raman Scattering, Surface Plasmon Resonance, Colorimetry, Immunochromatographic assay, Chemiluminescence, Electrochemiluminescence, and fluorescence) for detecting and profiling exosomal biomarkers. Additionally, we have summarized strengths and drawbacks of each strategy. Finally, challenges and future outlooks in developing efficient nanomaterial-based optical biosensor systems for exosomal tumor biomarkers detection have been discussed. The review will exhibit an overview of this field and provide meaningful information for scientific researchers.
Collapse
Affiliation(s)
- Baoyi Shao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, 210096, PR China
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, 210096, PR China.
| |
Collapse
|
33
|
Sokolov AV, Kostin NN, Ovchinnikova LA, Lomakin YA, Kudriaeva AA. Targeted Drug Delivery in Lipid-like Nanocages and Extracellular Vesicles. Acta Naturae 2019; 11:28-41. [PMID: 31413877 PMCID: PMC6643341 DOI: 10.32607/20758251-2019-11-2-28-41] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
The possibility of targeted drug delivery to a specific tissue, organ, or cell has opened new promising avenues in treatment development. The technology of targeted delivery aims to create multifunctional carriers that are capable of long circulation in the patient's organism and possess low toxicity at the same time. The surface of modern synthetic carriers has high structural similarity to the cell membrane, which, when combined with additional modifications, also promotes the transfer of biological properties in order to penetrate physiological barriers effectively. Along with artificial nanocages, further efforts have recently been devoted to research into extracellular vesicles that could serve as natural drug delivery vehicles. This review provides a detailed description of targeted delivery systems that employ lipid and lipid-like nanocages, as well as extracellular vesicles with a high level of biocompatibility, highlighting genetically encoded drug delivery vehicles.
Collapse
Affiliation(s)
- A. V. Sokolov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16 /10, Moscow, 117997, Russia
| | - N. N. Kostin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16 /10, Moscow, 117997, Russia
| | - L. A. Ovchinnikova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16 /10, Moscow, 117997, Russia
| | - Y. A. Lomakin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16 /10, Moscow, 117997, Russia
| | - A. A. Kudriaeva
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Str. 16 /10, Moscow, 117997, Russia
| |
Collapse
|
34
|
Lin J, Ma L, Zhang D, Gao J, Jin Y, Han Z, Lin D. Tumour biomarkers-Tracing the molecular function and clinical implication. Cell Prolif 2019; 52:e12589. [PMID: 30873683 PMCID: PMC6536410 DOI: 10.1111/cpr.12589] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/19/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022] Open
Abstract
In recent years, with the increase in cancer mortality caused by metastasis, and with the development of individualized and precise medical treatment, early diagnosis with precision becomes the key to decrease the death rate. Since detecting tumour biomarkers in body fluids is the most non‐invasive way to identify the status of tumour development, it has been widely investigated for the usage in clinic. These biomarkers include different expression or mutation in microRNAs (miRNAs), circulating tumour DNAs (ctDNAs), proteins, exosomes and circulating tumour cells (CTCs). In the present article, we summarized and discussed some updated research on these biomarkers. We overviewed their biological functions and evaluated their multiple roles in human and small animal clinical treatment, including diagnosis of cancers, classification of cancers, prognostic and predictive values for therapy response, monitors for therapy efficacy, and anti‐cancer therapeutics. Biomarkers including different expression or mutation in miRNAs, ctDNAs, proteins, exosomes and CTCs provide more choice for early diagnosis of tumour detection at early stage before metastasis. Combination detection of these tumour biomarkers may provide higher accuracy at the lowest molecule combination number for tumour early detection. Moreover, tumour biomarkers can provide valuable suggestions for clinical anti‐cancer treatment and execute monitoring of treatment efficiency.
Collapse
Affiliation(s)
- Jiahao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lie Ma
- Department of Respiratory Disease, The Navy General Hospital of PLA, Beijing, China
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiafeng Gao
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yipeng Jin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhihai Han
- Department of Respiratory Disease, The Navy General Hospital of PLA, Beijing, China
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
35
|
Li SL, An N, Liu B, Wang SY, Wang JJ, Ye Y. Exosomes from LNCaP cells promote osteoblast activity through miR-375 transfer. Oncol Lett 2019; 17:4463-4473. [PMID: 30988815 PMCID: PMC6447935 DOI: 10.3892/ol.2019.10110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023] Open
Abstract
Previous studies have revealed that exosomes influence tumour metastasis, diagnosis and treatment. In addition, exosomal microRNAs (miRNAs/miRs) are closely associated with the metastatic microenvironment; however, the regulatory role of exosomal miRNAs from prostate cancer cells on bone metastasis remains poorly understood. In the present study, a series of experiments were performed to determine whether exosomal miR-375 from LNCaP cells promote osteoblast activity. Exosomes were isolated and purified by ultracentrifugation, total RNA from cells and total miRNA from exosomes were then extracted, and miR-375 levels were detected by reverse transcription-quantitative polymerase chain reaction. Exosome libraries from LNCaP and RWPE-1 cells were sequenced and selected using an Illumina HiSeq™ 2500 system. The effects of exosomes on osteoblasts were determined and osteoblast activity was evaluated by measuring the activity of alkaline phosphatase, the extent of extracellular matrix mineralisation and the expression of osteoblast activity-associated marker genes. Morphological observations, particle size analysis and molecular phenotyping confirmed that cell supernatants contained exosomes. Differential expression analysis confirmed high miR-375 expression levels in LNCaP cell-derived exosomes. The ability of exosomes to enter osteoblasts and increase their levels of miR-375 was further analysed. The results demonstrated that exosomal miR-375 significantly promoted osteoblast activity. In conclusion, the present study may lead to further investigation of the function role of exosomal miR-375 in the activation and differentiation of osteoblasts in PCa.
Collapse
Affiliation(s)
- Su-Liang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Na An
- Department of Laboratory Medicine, Shaanxi Jiaotong Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Bing Liu
- Department of Pathology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Sheng-Yu Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Jian-Jun Wang
- Intensive Care Unit, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Yun Ye
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| |
Collapse
|
36
|
Lichtenstein AV. Strategies of the War on Cancer: To Kill or to Neutralize? Front Oncol 2019; 8:667. [PMID: 30687641 PMCID: PMC6335948 DOI: 10.3389/fonc.2018.00667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022] Open
|
37
|
Abstract
Exosomes are a class of extracellular vesicles released by multiple cells types including tumor cells, with a size range of 30-100 nm and a lipid bilayer membrane. Recently, the role of exosomes in cell-to-cell communication has been extensively studied, showed that exosomes can deliver their functional RNAs and proteins to recipient cells, impacting transcription and translation of recipient cells. Emerging evidence suggests that hepatocellular carcinoma (HCC) cell-derived exosomes can construct a fertile environment to support HCC cells proliferation, grow, invasion and metastasis, development of drug resistance. Circulating exosomes can be used as noninvasive biomarkers for early diagnosis, moreover as drug delivery vehicles, provide new insights into the treatment of HCC.
Collapse
|
38
|
Hadifar S, Fateh A, Yousefi MH, Siadat SD, Vaziri F. Exosomes in tuberculosis: Still terra incognita? J Cell Physiol 2018; 234:2104-2111. [PMID: 30317619 DOI: 10.1002/jcp.27555] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/13/2018] [Indexed: 12/20/2022]
Abstract
Today, diagnosis, vaccination, and treatment of tuberculosis (TB) remain major clinical challenges. Therefore, an introduction of new diagnostic measures and biomarkers is necessary to improve infection control. The ideal biomarker for TB infection can be defined as a host or pathogen-derived biomolecule, which is potent for identifying infection and determining its clinical stage. Exosomes, defined as cell-derived nanovesicles released into biological fluids, are involved in cell-cell communication and immune modulation. These vesicles have emerged as a new platform for improving the clinical diagnosis and prognosis of different infectious diseases and cancers. The role of these nanovehicles, as alternative biomarkers for the improvement of TB diagnosis and treatment, has been demonstrated in a significant body of literature. In this review, we summarized recent progress in the clinical application of exosome-based biomarkers in TB infection.
Collapse
Affiliation(s)
- Shima Hadifar
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Hadi Yousefi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
39
|
EV, Microvesicles/MicroRNAs and Stem Cells in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:123-135. [PMID: 29754178 DOI: 10.1007/978-3-319-74470-4_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The role of extracellular vesicles (EV) in carcinogenesis has become the focus of much research. These microscopic messengers have been found to regulate immune system function, particularly in tumorigenesis, as well as conditioning future metastatic sites for the attachment and growth of tumor tissue. Through an interaction with a range of host tissues, EVs are able to generate a pro-tumor environment that is essential for tumorigenesis. These small nanovesicles are an ideal candidate for a non-invasive indicator of pathogenesis and/or disease progression as they can display individualized nucleic acid, protein, and lipid expression profiles that are often reflective of disease state, and can be easily detected in bodily fluids, even after extended cryo-storage. Furthermore, the ability of EVs to securely transport signaling molecules and localize to distant tissues suggests these particles may greatly improve the delivery of therapeutic treatments, particularly in cancer. In this chapter, we discuss the role of EV in the identification of new diagnostic and prognostic cancer biomarkers, as well as the development of novel EV-based cancer therapies.
Collapse
|
40
|
Zhou L, Lv T, Zhang Q, Zhu Q, Zhan P, Zhu S, Zhang J, Song Y. The biology, function and clinical implications of exosomes in lung cancer. Cancer Lett 2017; 407:84-92. [PMID: 28807820 DOI: 10.1016/j.canlet.2017.08.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/29/2017] [Accepted: 08/04/2017] [Indexed: 02/06/2023]
Abstract
Exosomes are 30-100 nm small membrane vesicles of endocytic origin that are secreted by all types of cells, and can also be found in various body fluids. Increasing evidence implicates that exosomes confer stability and can deliver their cargos such as proteins and nucleic acids to specific cell types, which subsequently serve as important messengers and carriers in lung carcinogenesis. Here, we describe the biogenesis and components of exosomes mainly in lung cancer, we summarize their function in lung carcinogenesis (epithelial mesenchymal transition, oncogenic cell transformation, angiogenesis, metastasis and immune response in tumor microenvironment), and importantly we focus on the clinical potential of exosomes as biomarkers and therapeutics in lung cancer. In addition, we also discuss current challenges that might impede the clinical use of exosomes. Further studies on the functional roles of exosomes in lung cancer requires thorough research.
Collapse
Affiliation(s)
- Li Zhou
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Qun Zhang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Qingqing Zhu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Ping Zhan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Suhua Zhu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Jianya Zhang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China.
| |
Collapse
|
41
|
Lv L, Li C, Zhang X, Ding N, Cao T, Jia X, Wang J, Pan L, Jia H, Li Z, Zhang J, Chen F, Zhang Z. RNA Profiling Analysis of the Serum Exosomes Derived from Patients with Active and Latent Mycobacterium tuberculosis Infection. Front Microbiol 2017; 8:1051. [PMID: 28659881 PMCID: PMC5466984 DOI: 10.3389/fmicb.2017.01051] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/26/2017] [Indexed: 11/23/2022] Open
Abstract
Tuberculosis (TB) has exceeded HIV as the most lethal infectious disease globally for two consecutive years. Moreover, one third of the world’s population is estimated to have latent tuberculosis infection (LTBI). This is mainly because of difficulties associated with diagnosis and treatment for both TB and LTBI patients. Exosomes provide a promising research tool for TB diagnosis and treatment because they are released from various cells containing valuable biochemical information related to disease. In this study, we performed RNA-sequencing analysis on exosomes derived from clinical specimens of healthy controls (HC), active tuberculosis (ATB), and LTBI patients. Our results revealed the distinct gene expression profiles of the exosomes from LTBI and ATB patients. (1) We identified many distinct up-regulated and down-regulated differentially expressed genes (DEGs) in LTBI and ATB samples, and further screened the top-20 DEGs which might provide a potential panel for differentiation of HC, LTBI, and ATB. (2) We classified all the DEGs into six expression patterns, screened the top-20 genes in each pattern, and mainly focused on those highly expressed in LTBI and ATB. (3) Some Mycobacterium tuberculosis (Mtb) RNAs were only enriched in the exosomes of LTBI samples. (4) Pathway and function analysis further indicated down-regulated signaling pathways/immune response and up-regulated apoptosis/necrosis. Our findings indicate the selective packaging of RNA cargoes into exosomes under different stages of Mtb infection, while facilitating the development of potential targets for the diagnosis, prevention and treatment of tuberculosis.
Collapse
Affiliation(s)
- Lingna Lv
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijing, China
| | - Cuidan Li
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China.,University of Chinese Academy of SciencesBeijing, China
| | - Xiuli Zhang
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China.,University of Chinese Academy of SciencesBeijing, China
| | - Nan Ding
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China
| | - Tianshu Cao
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China
| | - Xinmiao Jia
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China.,University of Chinese Academy of SciencesBeijing, China
| | - Jinghui Wang
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijing, China
| | - Liping Pan
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijing, China
| | - Hongyan Jia
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijing, China
| | - Zihui Li
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijing, China
| | - Ju Zhang
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China
| | - Fei Chen
- CAS Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of SciencesBeijing, China.,University of Chinese Academy of SciencesBeijing, China.,Collaborative Innovation Center for Genetics and DevelopmentShanghai, China.,Sino-Danish College, University of Chinese Academy of SciencesBeijing, China
| | - Zongde Zhang
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
42
|
Shtam TA, Burdakov VS, Landa SB, Naryzhny SN, Bairamukov VY, Malek AV, Orlov YN, Filatov MV. Aggregation by lectins as an approach for exosome isolation from biological fluids: Validation for proteomic studies. ACTA ACUST UNITED AC 2017. [DOI: 10.1134/s1990519x17020043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Kang H, Kim J, Park J. Methods to isolate extracellular vesicles for diagnosis. MICRO AND NANO SYSTEMS LETTERS 2017. [DOI: 10.1186/s40486-017-0049-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
44
|
Cai S, Cheng X, Pan X, Li J. Emerging role of exosomes in liver physiology and pathology. Hepatol Res 2017; 47:194-203. [PMID: 27539153 DOI: 10.1111/hepr.12794] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/21/2016] [Accepted: 08/10/2016] [Indexed: 12/13/2022]
Abstract
Exosomes can mediate intercellular communication by conveying various bioactive molecules. Plentiful evidence suggests that exosomes are involved in many liver diseases including hepatitis C virus infection, hepatitis B virus infection, hepatocellular carcinoma, liver fibrosis, cirrhosis, non-alcoholic fatty liver disease, and alcoholic liver disease. Moreover, exosomes are present in nearly all human body fluids. Therefore, exosomal miRNA or proteins have the potential to be novel biomarkers of liver diseases. In the treatment of liver diseases, exosomes could participate in adaptive immune response and mesenchymal stem cell-based therapy. Exosomes can also be used as vehicles for genetic materials and drug delivery.
Collapse
Affiliation(s)
- Shuangpeng Cai
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei, China
| | - Xiaoyu Cheng
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei, China
| | - Xueyin Pan
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei, China
| | - Jun Li
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei, China
| |
Collapse
|
45
|
Abstract
Better diagnostic biomarkers and therapeutic options are still necessary for patients with sarcomas due to the current limitations of diagnosis and treatment. Exosomes are small extracellular membrane vesicles that are released by various cells and are found in most body fluids. Tumor-derived exosomes have been proven to mediate tumorigenesis, intercellular communication, microenvironment modulation, and metastasis in different cancers, including in sarcomas. Recently, exosomes have been considered as potential biomarkers for sarcoma diagnosis and prognosis, and as possible targets for sarcoma therapy. Moreover, due to their specific cell tropism and bioavailability, exosomes can also be engineered as vehicles for drug delivery. In this review, we discuss recent advances in the roles of tumor-derived exosomes in sarcoma and their potential clinical applications.
Collapse
|
46
|
Soung YH, Ford S, Zhang V, Chung J. Exosomes in Cancer Diagnostics. Cancers (Basel) 2017; 9:cancers9010008. [PMID: 28085080 PMCID: PMC5295779 DOI: 10.3390/cancers9010008] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/21/2022] Open
Abstract
Exosomes are endosome derived extracellular vesicles of 30–120 nm size ranges. Exosomes have been identified as mediators of cell-to-cell communication by transferring bioactive molecules such as nucleic acids, proteins and lipids into recipient cells. While exosomes are secreted by multiple cell types, cancer derived exosomes not only influence the invasive potentials of proximally located cells, but also affect distantly located tissues. Based on their ability to alter tumor microenvironment by regulating immunity, angiogenesis and metastasis, there has been growing interest in defining the clinical relevance of exosomes in cancers. In particular, exosomes are valuable sources for biomarkers due to selective cargo loading and resemblance to their parental cells. In this review, we summarize the recent findings to utilize exosomes as cancer biomarkers for early detection, diagnosis and therapy selection.
Collapse
Affiliation(s)
- Young Hwa Soung
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| | - Shane Ford
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| | - Vincent Zhang
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| | - Jun Chung
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| |
Collapse
|
47
|
Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the Damaged Heart: Mesenchymal Stem Cells, Cell-Based Therapy, and Engineered Heart Tissue. Physiol Rev 2016; 96:1127-68. [PMID: 27335447 PMCID: PMC6345247 DOI: 10.1152/physrev.00019.2015] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are broadly distributed cells that retain postnatal capacity for self-renewal and multilineage differentiation. MSCs evade immune detection, secrete an array of anti-inflammatory and anti-fibrotic mediators, and very importantly activate resident precursors. These properties form the basis for the strategy of clinical application of cell-based therapeutics for inflammatory and fibrotic conditions. In cardiovascular medicine, administration of autologous or allogeneic MSCs in patients with ischemic and nonischemic cardiomyopathy holds significant promise. Numerous preclinical studies of ischemic and nonischemic cardiomyopathy employing MSC-based therapy have demonstrated that the properties of reducing fibrosis, stimulating angiogenesis, and cardiomyogenesis have led to improvements in the structure and function of remodeled ventricles. Further attempts have been made to augment MSCs' effects through genetic modification and cell preconditioning. Progression of MSC therapy to early clinical trials has supported their role in improving cardiac structure and function, functional capacity, and patient quality of life. Emerging data have supported larger clinical trials that have been either completed or are currently underway. Mechanistically, MSC therapy is thought to benefit the heart by stimulating innate anti-fibrotic and regenerative responses. The mechanisms of action involve paracrine signaling, cell-cell interactions, and fusion with resident cells. Trans-differentiation of MSCs to bona fide cardiomyocytes and coronary vessels is also thought to occur, although at a nonphysiological level. Recently, MSC-based tissue engineering for cardiovascular disease has been examined with quite encouraging results. This review discusses MSCs from their basic biological characteristics to their role as a promising therapeutic strategy for clinical cardiovascular disease.
Collapse
Affiliation(s)
- Samuel Golpanian
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Ariel Wolf
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Konstantinos E Hatzistergos
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
48
|
Abstract
Humans circulate quadrillions of exosomes at all times. Exosomes are a class of extracellular vesicles released by all cells, with a size range of 40-150 nm and a lipid bilayer membrane. Exosomes contain DNA, RNA, and proteins. Exosomes likely remove excess and/or unnecessary constituents from the cells, functioning like garbage bags, although their precise physiological role remains unknown. Additionally, exosomes may mediate specific cell-to-cell communication and activate signaling pathways in cells they fuse or interact with. Exosomes are detected in the tumor microenvironment, and emerging evidence suggests that they play a role in facilitating tumorigenesis by regulating angiogenesis, immunity, and metastasis. Circulating exosomes can be used as liquid biopsies and noninvasive biomarkers for early detection, diagnosis, and treatment of cancer patients.
Collapse
|
49
|
Abstract
Despite substantial clinical advances over the past 65 years, cardiovascular disease remains the leading cause of death in America. The past 15 years has witnessed major basic and translational interest in the use of stem and precursor cells as a therapeutic agent for chronically injured organs. Among the cell types under investigation, adult mesenchymal stem cells are widely studied, and in early stage, clinical studies show promise for repair and regeneration of cardiac tissues. The ability of mesenchymal stem cells to differentiate into mesoderm- and nonmesoderm-derived tissues, their immunomodulatory effects, their availability, and their key role in maintaining and replenishing endogenous stem cell niches have rendered them one of the most heavily investigated and clinically tested type of stem cell. Accumulating data from preclinical and early phase clinical trials document their safety when delivered as either autologous or allogeneic forms in a range of cardiovascular diseases, but also importantly define parameters of clinical efficacy that justify further investigation in larger clinical trials. Here, we review the biology of mesenchymal stem cells, their interaction with endogenous molecular and cellular pathways, and their modulation of immune responses. Additionally, we discuss factors that enhance their proliferative and regenerative ability and factors that may hinder their effectiveness in the clinical setting.
Collapse
Affiliation(s)
- Vasileios Karantalis
- From the University of Miami Miller School of Medicine, Interdisciplinary Stem Cell Institute, FL
| | - Joshua M Hare
- From the University of Miami Miller School of Medicine, Interdisciplinary Stem Cell Institute, FL.
| |
Collapse
|
50
|
Abstract
Cancer research has shifted in recent years from studying intracellular processes (identification of damaged genes and signaling pathways) to extracellular (hierarchy of tumor cells, cell transitions, clone competition) and tissue (interactions of a tumor with its environment) research. But then the next step seems to be logical: studying biochemistry of tumor-bearing organisms (namely, cancer-induced changes in cellular and tissue metabolism leading to the organism's death). These data can help to develop new methods of cancer treatment. This article discusses some of the challenges of contemporary oncology and possible ways to overcome them.
Collapse
Affiliation(s)
- A V Lichtenstein
- Blokhin Cancer Research Center, Institute of Carcinogenesis, Moscow, 115478, Russia.
| |
Collapse
|