1
|
Abedi A, Moosazadeh Moghaddam M, Kachuei R, Imani Fooladi AA. Exosomes as a Therapeutic Strategy in Cancer: Potential Roles as Drug Carriers and Immune Modulators. Biochim Biophys Acta Rev Cancer 2024; 1880:189238. [PMID: 39674417 DOI: 10.1016/j.bbcan.2024.189238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
Exosome-based cancer immunotherapy is advancing quickly on the concept of artificially activating the immune system to combat cancer. They can mechanistically change the tumor microenvironment, increase immune responses, and function as efficient drug delivery vehicles because of their inherent bioactivity, low toxicity, and immunogenicity. Accurate identification of the mechanisms of action of exosomes in tumor environments, along with optimization of their isolation, purification, and characterization methods, is necessary to increase clinical applications. Exosomes can be modified through cargo loading and surface modification to enhance their therapeutic applications, either before or after the donor cells' isolation. These engineered exosomes can directly target tumor cells at the tumor site or indirectly activate innate and adaptive immune responses in the tumor microenvironment. This approach is particularly effective when combined with traditional cancer immunotherapy techniques such as vaccines, immune checkpoints, and CAR-T cells. It can improve anti-tumor responses, induce long-term immunity, and address the limitations of traditional therapies, such as poor penetration in solid tumors and immunosuppressive environments. This review aims to provide a comprehensive and detailed overview of the direct role of engineered exosomes as drug delivery systems and their immunomodulatory effects on tumors as an indirect approach to fighting cancer. Additionally, it will discuss novel immunotherapy options.
Collapse
Affiliation(s)
- Azam Abedi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Bagheri R, Ghorbian M, Ghorbian S. Tumor circulating biomarkers in colorectal cancer. Cancer Treat Res Commun 2023; 38:100787. [PMID: 38194840 DOI: 10.1016/j.ctarc.2023.100787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024]
Abstract
CRC is a major global health concern and is responsible for a significant number of cancer-related deaths each year. The successful treatment of CRC becomes more difficult when it goes undetected until it has advanced to a later stage. Diagnostic biomarkers can play a critical role in the early detection of CRC, which leads to improved patient outcomes and increased survival rates. It is important to develop reliable biomarkers for the early detection of CRC to enable timely diagnosis and treatment. To date, CRC detection methods such as endoscopy, blood, and stool tests are imperfect and often only identify cases in the later stages of the disease. To overcome these limitations, researchers are turning to molecular biomarkers as a promising avenue for improving CRC detection. Diagnostic information can be provided more reliably through a noninvasive approach using biomarkers such as mRNA, circulating cell-free DNA, micro-RNA, long non-coding RNA, and proteins. These biomarkers can be found in blood, tissue, feces, and volatile organic compounds. The identification of molecular biomarkers with high sensitivity and specificity for early detection of CRC that are safe, cost-effective, and easily measurable remains a significant challenge for researchers. In this article, we will explore the latest advancements in blood-based diagnostic biomarkers for CRC and their potential impact on improving patient survival rates.
Collapse
Affiliation(s)
- Raana Bagheri
- Department of Molecular Genetics, Ahar Branch, Islamic Azad University, Ahar, Iran
| | - Mohsen Ghorbian
- Department of Computer Engineering, Qom Branch, Islamic Azad University, Qom, Iran
| | - Saeid Ghorbian
- Department of Molecular Genetics, Ahar Branch, Islamic Azad University, Ahar, Iran.
| |
Collapse
|
3
|
Zhong Q, Nie Q, Wu R, Huang Y. Exosomal miR-18a-5p promotes EMT and metastasis of NPC cells via targeting BTG3 and activating the Wnt/β-catenin signaling pathway. Cell Cycle 2023; 22:1544-1562. [PMID: 37287276 PMCID: PMC10361138 DOI: 10.1080/15384101.2023.2216508] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 02/28/2023] [Accepted: 04/26/2023] [Indexed: 06/09/2023] Open
Abstract
This study investigated the underlying mechanism of miR-18a-5p regulating the proliferation, invasion, and metastasis of nasopharyngeal carcinoma (NPC) cells in vitro and in vivo to indicate the pathogenesis of NPC. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was utilized to determine miR-18a-5p expression level in NPC tissues and cell lines. Besides, 2,5-diphenyl-2 H-tetrazolium bromide (MTT) and colony formation assays were employed to detect the effect of miR-18a-5p expression level on NPC cell proliferation. Wound healing and Transwell assays were utilized to detect the effect of miR-18a-5p on NPC cell invasion and migration. The expression levels of epithelial-mesenchymal transition (EMT)-related proteins (Vimentin, N-cadherin, and E-cadherin) were identified by Western blot assay. After collecting exosomes from CNE-2 cells, it was found that exosomal miR-18a-5p secreted from NPC cells promoted NPC cell proliferation, migration, invasion, and EMT, whereas inhibition of miR-18a-5p expression level led to the opposite results. The dual-luciferase reporter assay showed that BTG anti-proliferation factor 3 (BTG3) was the target gene of miR-18a-5p, and BTG3 could overturn the effect of miR-18a-5p on NPC cells. Xenograft mouse model of NPC nude mice showed that miR-18a-5p promoted NPC growth and metastasis in vivo. This study revealed that exosomal miR-18a-5p derived from NPC cells promoted angiogenesis via targeting BTG3 and activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Qiong Zhong
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| | - Qihong Nie
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| | - Renrui Wu
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| | - Yun Huang
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| |
Collapse
|
4
|
Antunes-Ferreira M, D'Ambrosi S, Arkani M, Post E, In 't Veld SGJG, Ramaker J, Zwaan K, Kucukguzel ED, Wedekind LE, Griffioen AW, Oude Egbrink M, Kuijpers MJE, van den Broek D, Noske DP, Hartemink KJ, Sabrkhany S, Bahce I, Sol N, Bogaard HJ, Koppers-Lalic D, Best MG, Wurdinger T. Tumor-educated platelet blood tests for Non-Small Cell Lung Cancer detection and management. Sci Rep 2023; 13:9359. [PMID: 37291189 PMCID: PMC10250384 DOI: 10.1038/s41598-023-35818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023] Open
Abstract
Liquid biopsy approaches offer a promising technology for early and minimally invasive cancer detection. Tumor-educated platelets (TEPs) have emerged as a promising liquid biopsy biosource for the detection of various cancer types. In this study, we processed and analyzed the TEPs collected from 466 Non-small Cell Lung Carcinoma (NSCLC) patients and 410 asymptomatic individuals (controls) using the previously established thromboSeq protocol. We developed a novel particle-swarm optimization machine learning algorithm which enabled the selection of an 881 RNA biomarker panel (AUC 0.88). Herein we propose and validate in an independent cohort of samples (n = 558) two approaches for blood samples testing: one with high sensitivity (95% NSCLC detected) and another with high specificity (94% controls detected). Our data explain how TEP-derived spliced RNAs may serve as a biomarker for minimally-invasive clinical blood tests, complement existing imaging tests, and assist the detection and management of lung cancer patients.
Collapse
Affiliation(s)
- Mafalda Antunes-Ferreira
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Silvia D'Ambrosi
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Mohammad Arkani
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Edward Post
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Sjors G J G In 't Veld
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Jip Ramaker
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Kenn Zwaan
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Ece Demirel Kucukguzel
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Laurine E Wedekind
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Mirjam Oude Egbrink
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - David P Noske
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Koen J Hartemink
- Department of Thoracic Surgery, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Siamack Sabrkhany
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Idris Bahce
- Department of Pulmonary Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Nik Sol
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
- Department of Neurology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Harm-Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | | | - Myron G Best
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Xiong J, Chi H, Yang G, Zhao S, Zhang J, Tran LJ, Xia Z, Yang F, Tian G. Revolutionizing anti-tumor therapy: unleashing the potential of B cell-derived exosomes. Front Immunol 2023; 14:1188760. [PMID: 37342327 PMCID: PMC10277631 DOI: 10.3389/fimmu.2023.1188760] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
B cells occupy a vital role in the functioning of the immune system, working in tandem with T cells to either suppress or promote tumor growth within the tumor microenvironment(TME). In addition to direct cell-to-cell communication, B cells and other cells release exosomes, small membrane vesicles ranging in size from 30-150 nm, that facilitate intercellular signaling. Exosome research is an important development in cancer research, as they have been shown to carry various molecules such as major histocompatibility complex(MHC) molecules and integrins, which regulate the TME. Given the close association between TME and cancer development, targeting substances within the TME has emerged as a promising strategy for cancer therapy. This review aims to present a comprehensive overview of the contributions made by B cells and exosomes to the tumor microenvironment (TME). Additionally, we delve into the potential role of B cell-derived exosomes in the progression of cancer.
Collapse
Affiliation(s)
- Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Songyun Zhao
- Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jing Zhang
- Division of Basic Biomedical Sciences, The University of South Dakota Sanford School of Medicine, Vermillion, SD, United States
| | - Lisa Jia Tran
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fang Yang
- Department of Ophthalmology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Ferlizza E, Romaniello D, Borrelli F, Pagano F, Girone C, Gelfo V, Kuhre RS, Morselli A, Mazzeschi M, Sgarzi M, Filippini DM, D'Uva G, Lauriola M. Extracellular Vesicles and Epidermal Growth Factor Receptor Activation: Interplay of Drivers in Cancer Progression. Cancers (Basel) 2023; 15:cancers15112970. [PMID: 37296932 DOI: 10.3390/cancers15112970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/12/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Extracellular vesicles (EVs) are of great interest to study the cellular mechanisms of cancer development and to diagnose and monitor cancer progression. EVs are a highly heterogeneous population of cell derived particles, which include microvesicles (MVs) and exosomes (EXOs). EVs deliver intercellular messages transferring proteins, lipids, nucleic acids, and metabolites with implications for tumour progression, invasiveness, and metastasis. Epidermal Growth Factor Receptor (EGFR) is a major driver of cancer. Tumour cells with activated EGFR could produce EVs disseminating EGFR itself or its ligands. This review provides an overview of EVs (mainly EXOs and MVs) and their cargo, with a subsequent focus on their production and effects related to EGFR activation. In particular, in vitro studies performed in EGFR-dependent solid tumours and/or cell cultures will be explored, thus shedding light on the interplay between EGFR and EVs production in promoting cancer progression, metastases, and resistance to therapies. Finally, an overview of liquid biopsy approaches involving EGFR and EVs in the blood/plasma of EGFR-dependent tumour patients will also be discussed to evaluate their possible application as candidate biomarkers.
Collapse
Affiliation(s)
- Enea Ferlizza
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Donatella Romaniello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Francesco Borrelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Federica Pagano
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Cinzia Girone
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Valerio Gelfo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Rikke Sofie Kuhre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Alessandra Morselli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Martina Mazzeschi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Michela Sgarzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Daria Maria Filippini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Gabriele D'Uva
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Mattia Lauriola
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
7
|
Fang H, Liu M, Jiang W. Nickel-Doped Microfluidic Chip for Rapid and Efficient Immunomagnetic Separation and Detection of Breast Cancer Cell-Derived Exosomes. Appl Biochem Biotechnol 2022; 195:3109-3121. [PMID: 36542270 DOI: 10.1007/s12010-022-04272-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Breast cancer cell-derived exosomes have high potential as biomarkers for continuous biopsies and longitudinal monitoring in breast cancer. However, it is extremely difficult to separate exosomes with high recovery and high purity from complex media, such as urine, plasma, saliva and cell culture supernatants. Here, we designed a flexible and simple microfluidic chip for exosome separation. The capture zone of the chip is a three-dimensional structure of interlaced cylinders doped with nickel powder. Exosomes were separated from cell culture supernatant by the immunomagnetic separation method in continuous flow mode and were detected by fluorescence imaging with high sensitivity. The chip achieved a high exosome recovery rate (> 74%) and purity (> 67%) at an injection rate of 3.6 mL/h. Thus, this chip was demonstrated to be a cutting-edge platform for the separation and detection of exosomes. It could also be applied to separate and detect other types of exosomes, microbubbles and cells.
Collapse
Affiliation(s)
- Huiying Fang
- Department of Breast Diseases, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, No. 181 Hanyu Rd, Shapingba District, 400030, Chongqing, China
| | - Mei Liu
- Department of Breast Diseases, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, No. 181 Hanyu Rd, Shapingba District, 400030, Chongqing, China
| | - Wei Jiang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, 400010, Chongqing, China.
| |
Collapse
|
8
|
Ginini L, Billan S, Fridman E, Gil Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022; 11:1375. [PMID: 35563681 PMCID: PMC9101098 DOI: 10.3390/cells11091375] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogamous lipid bilayer-enclosed membranous structures secreted by cells. They are comprised of apoptotic bodies, microvesicles, and exosomes, and carry a range of nucleic acids and proteins that are necessary for cell-to-cell communication via interaction on the cells surface. They initiate intracellular signaling pathways or the transference of cargo molecules, which elicit pleiotropic responses in recipient cells in physiological processes, as well as pathological processes, such as cancer. It is therefore important to understand the molecular means by which EVs are taken up into cells. Accordingly, this review summarizes the underlying mechanisms involved in EV targeting and uptake. The primary method of entry by EVs appears to be endocytosis, where clathrin-mediated, caveolae-dependent, macropinocytotic, phagocytotic, and lipid raft-mediated uptake have been variously described as being prevalent. EV uptake mechanisms may depend on proteins and lipids found on the surfaces of both vesicles and target cells. As EVs have been shown to contribute to cancer growth and progression, further exploration and targeting of the gateways utilized by EVs to internalize into tumor cells may assist in the prevention or deceleration of cancer pathogenesis.
Collapse
Affiliation(s)
- Lana Ginini
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Salem Billan
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
- Medical Oncology and Radiation Therapy Program, Oncology Section, Rambam Health Care Campus, HaAliya HaShniya Street 8, Haifa 3109601, Israel
| | - Eran Fridman
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Ziv Gil
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
| |
Collapse
|
9
|
Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem 2022; 232:114205. [DOI: 10.1016/j.ejmech.2022.114205] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
|
10
|
Filipović L, Kojadinović M, Popović M. Exosomes and exosome-mimetics as targeted drug carriers: Where we stand and what the future holds? J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Novel Diagnostic Biomarkers in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23020852. [PMID: 35055034 PMCID: PMC8776048 DOI: 10.3390/ijms23020852] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/27/2021] [Accepted: 01/03/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is still a leading cause of cancer death worldwide. Less than half of cases are diagnosed when the cancer is locally advanced. CRC is a heterogenous disease associated with a number of genetic or somatic mutations. Diagnostic markers are used for risk stratification and early detection, which might prolong overall survival. Nowadays, the widespread use of semi-invasive endoscopic methods and feacal blood tests characterised by suboptimal accuracy of diagnostic results has led to the detection of cases at later stages. New molecular noninvasive tests based on the detection of CRC alterations seem to be more sensitive and specific then the current methods. Therefore, research aiming at identifying molecular markers, such as DNA, RNA and proteins, would improve survival rates and contribute to the development of personalized medicine. The identification of “ideal” diagnostic biomarkers, having high sensitivity and specificity, being safe, cheap and easy to measure, remains a challenge. The purpose of this review is to discuss recent advances in novel diagnostic biomarkers for tumor tissue, blood and stool samples in CRC patients.
Collapse
|
12
|
Lu Y, Ye L, Jian X, Yang D, Zhang H, Tong Z, Wu Z, Shi N, Han Y, Mao H. Integrated microfluidic system for isolating exosome and analyzing protein marker PD-L1. Biosens Bioelectron 2021; 204:113879. [PMID: 35180692 DOI: 10.1016/j.bios.2021.113879] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023]
Abstract
Exosomes are lipid-bilayered nanovesicles secreted by cells to mediate intercellular communication. Various kinds of biomolecules involved in exosomes offer non-invasive approaches for detecting or monitoring disease and developing targeted therapeutics. Here, we present an integrated microfluidic exosome isolation and detection system (EXID system) to analyze the abundance of the exosomal PD-L1 protein marker, which is a transmembrane protein expressed by tumors to suppress immune activation of T cells. By incorporating exosome isolation and biomarker labelling and quantification within a single microfluidic chip, our system reduced the total analysis time below 2 h. Using the EXID system, 7 categories of cell lines including cancer cell lines and control samples were profiled, where significant differences in the fluorescence intensity were observed with the limit of detection (LOD) down to 10.76 per microliter. Such noticeable variations in PD-L1 abundance among cancer cell lines highlighted the need of personalized treatments. Furthermore, 16 clinical samples from 7 post-treated cancer patients, 3 prior-treatment patients and 6 healthy controls, are tested, among which differences in sensitivity toward immune response were subsistent. Because the abundance of PD-L1 reflects the sensibility for immune response, our results provide useful guides to design immunotherapy strategies for different types of tumors.
Collapse
Affiliation(s)
- Yunxing Lu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ling Ye
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoyu Jian
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Dawei Yang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hongwei Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhaoduo Tong
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhenhua Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Nan Shi
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
13
|
Serafini FL, Lanuti P, Delli Pizzi A, Procaccini L, Villani M, Taraschi AL, Pascucci L, Mincuzzi E, Izzi J, Chiacchiaretta P, Buca D, Catitti G, Bologna G, Simeone P, Pieragostino D, Caulo M. Diagnostic Impact of Radiological Findings and Extracellular Vesicles: Are We Close to Radiovesicolomics? BIOLOGY 2021; 10:biology10121265. [PMID: 34943180 PMCID: PMC8698452 DOI: 10.3390/biology10121265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022]
Abstract
Simple Summary Over the years, diagnostic tests such as in radiology and flow cytometry have become more and more powerful in the constant struggle against different pathologies, some of which are life-threatening. The possibility of using these “weapons” in a conjugated manner could result in higher healing and prevention rates, and a decrease in late diagnosis diseases. Different correlations among pathologies, extracellular vesicles (EVs), and radiological findings were recently demonstrated by many authors. Together with the increasing importance of “omics” sciences, and artificial intelligence in this new century, the perspective of a new research field called “radiovesicolomics” could be the missing link, enabling a different approach to disease diagnosis and treatment. Abstract Currently, several pathologies have corresponding and specific diagnostic and therapeutic branches of interest focused on early and correct detection, as well as the best therapeutic approach. Radiology never ceases to develop newer technologies in order to give patients a clear, safe, early, and precise diagnosis; furthermore, in the last few years diagnostic imaging panoramas have been extended to the field of artificial intelligence (AI) and machine learning. On the other hand, clinical and laboratory tests, like flow cytometry and the techniques found in the “omics” sciences, aim to detect microscopic elements, like extracellular vesicles, with the highest specificity and sensibility for disease detection. If these scientific branches started to cooperate, playing a conjugated role in pathology diagnosis, what could be the results? Our review seeks to give a quick overview of recent state of the art research which investigates correlations between extracellular vesicles and the known radiological features useful for diagnosis.
Collapse
Affiliation(s)
- Francesco Lorenzo Serafini
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Andrea Delli Pizzi
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
- Correspondence:
| | - Luca Procaccini
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Michela Villani
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Alessio Lino Taraschi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Luca Pascucci
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Erica Mincuzzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Jacopo Izzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Piero Chiacchiaretta
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Davide Buca
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Giulia Catitti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| |
Collapse
|
14
|
Wang Z, Wu Y, Zhao Z, Liu C, Zhang L. Study on Transorgan Regulation of Intervertebral Disc and Extra-Skeletal Organs Through Exosomes Derived From Bone Marrow Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:741183. [PMID: 34631718 PMCID: PMC8495158 DOI: 10.3389/fcell.2021.741183] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022] Open
Abstract
Exosomes are membranous lipid vesicles fused with intracellular multicellular bodies and then released into the extracellular environment. They contain various bioactive substances, including proteins, mRNA, miRNAs, lncRNAs, circRNAs, lipids, transcription factors, and cytokine receptors. Under certain conditions, bone marrow mesenchymal stem cells (BMSCs) can differentiate into osteoblasts, chondrocytes, adipocytes, and biological functions. This study provides a theoretical basis for the application of exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos) in osteology, exploring different sources of exosomes to improve bone microenvironment and resist bone metastasis. We also provided new ideas for the prevention and rehabilitation of human diseases by exosomes.
Collapse
Affiliation(s)
- Zhikun Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yangming Wu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Zhonghan Zhao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Chengyi Liu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Lingli Zhang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| |
Collapse
|
15
|
Zhang LY, Yang X, Wang SB, Chen H, Pan HY, Hu ZM. Membrane Derived Vesicles as Biomimetic Carriers for Targeted Drug Delivery System. Curr Top Med Chem 2021; 20:2472-2492. [PMID: 32962615 DOI: 10.2174/1568026620666200922113054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/25/2020] [Accepted: 04/25/2020] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are membrane vesicles (MVs) playing important roles in various cellular and molecular functions in cell-to-cell signaling and transmitting molecular signals to adjacent as well as distant cells. The preserved cell membrane characteristics in MVs derived from live cells, give them great potential in biological applications. EVs are nanoscale particulates secreted from living cells and play crucial roles in several important cellular functions both in physiological and pathological states. EVs are the main elements in intercellular communication in which they serve as carriers for various endogenous cargo molecules, such as RNAs, proteins, carbohydrates, and lipids. High tissue tropism capacity that can be conveniently mediated by surface molecules, such as integrins and glycans, is a unique feature of EVs that makes them interesting candidates for targeted drug delivery systems. The cell-derived giant MVs have been exploited as vehicles for delivery of various anticancer agents and imaging probes and for implementing combinational phototherapy for targeted cancer treatment. Giant MVs can efficiently encapsulate therapeutic drugs and deliver them to target cells through the membrane fusion process to synergize photodynamic/photothermal treatment under light exposure. EVs can load diagnostic or therapeutic agents using different encapsulation or conjugation methods. Moreover, to prolong the blood circulation and enhance the targeting of the loaded agents, a variety of modification strategies can be exploited. This paper reviews the EVs-based drug delivery strategies in cancer therapy. Biological, pharmacokinetics and physicochemical characteristics, isolation techniques, engineering, and drug loading strategies of EVs are discussed. The recent preclinical and clinical progresses in applications of EVs and oncolytic virus therapy based on EVs, the clinical challenges and perspectives are discussed.
Collapse
Affiliation(s)
- Le-Yi Zhang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People's Hospital Chun’an
Branch), Hangzhou 311700, China
| | - Xue Yang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Hong Chen
- Department of Stomatology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Hong-Ying Pan
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China,Department of Infectious Diseases, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Zhi-Ming Hu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China,Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
16
|
Huda MN, Nafiujjaman M, Deaguero IG, Okonkwo J, Hill ML, Kim T, Nurunnabi M. Potential Use of Exosomes as Diagnostic Biomarkers and in Targeted Drug Delivery: Progress in Clinical and Preclinical Applications. ACS Biomater Sci Eng 2021; 7:2106-2149. [PMID: 33988964 PMCID: PMC8147457 DOI: 10.1021/acsbiomaterials.1c00217] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Exosomes are cell-derived vesicles containing heterogeneous active biomolecules such as proteins, lipids, mRNAs, receptors, immune regulatory molecules, and nucleic acids. They typically range in size from 30 to 150 nm in diameter. An exosome's surfaces can be bioengineered with antibodies, fluorescent dye, peptides, and tailored for small molecule and large active biologics. Exosomes have enormous potential as a drug delivery vehicle due to enhanced biocompatibility, excellent payload capability, and reduced immunogenicity compared to alternative polymeric-based carriers. Because of active targeting and specificity, exosomes are capable of delivering their cargo to exosome-recipient cells. Additionally, exosomes can potentially act as early stage disease diagnostic tools as the exosome carries various protein biomarkers associated with a specific disease. In this review, we summarize recent progress on exosome composition, biological characterization, and isolation techniques. Finally, we outline the exosome's clinical applications and preclinical advancement to provide an outlook on the importance of exosomes for use in targeted drug delivery, biomarker study, and vaccine development.
Collapse
Affiliation(s)
- Md Nurul Huda
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - Md Nafiujjaman
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Isaac G Deaguero
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - Jude Okonkwo
- John A Paulson School of Engineering, Harvard University, Cambridge, MA 02138
| | - Meghan L. Hill
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Taeho Kim
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Md Nurunnabi
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX 79968
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968
| |
Collapse
|
17
|
Giacobino C, Canta M, Fornaguera C, Borrós S, Cauda V. Extracellular Vesicles and Their Current Role in Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13092280. [PMID: 34068657 PMCID: PMC8126043 DOI: 10.3390/cancers13092280] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In recent years, immunotherapy has shown great advancement, becoming a powerful tool to combat cancer. In this context, the use of biologically derived vesicles has also acquired importance for cancer immunotherapy. Extracellular vesicles are thus proposed to transport molecules able to trigger an immune response and thus fight cancer cells. As a particular immunotherapeutic approach, a new technique also consists in the exploitation of extracellular vesicles as new cancer vaccines. The present review provides basic notions on cancer immunotherapy and describes several clinical trials in which therapeutic anticancer vaccines are tested. In particular, the potential of extracellular vesicles-based therapeutic vaccines in the treatment of cancer patients is highlighted, even with advanced stage-cancer. A focus on the clinical studies, already completed or still in progress, is offered and a systematic collection and reorganization of the present literature on this topic is proposed to the reader. Abstract Extracellular vesicles (EVs) are natural particles formed by the lipid bilayer and released from almost all cell types to the extracellular environment both under physiological conditions and in presence of a disease. EVs are involved in many biological processes including intercellular communication, acting as natural carriers in the transfer of various biomolecules such as DNA, various RNA types, proteins and different phospholipids. Thanks to their transfer and targeting abilities, they can be employed in drug and gene delivery and have been proposed for the treatment of different diseases, including cancer. Recently, the use of EVs as biological carriers has also been extended to cancer immunotherapy. This new technique of cancer treatment involves the use of EVs to transport molecules capable of triggering an immune response to damage cancer cells. Several studies have analyzed the possibility of using EVs in new cancer vaccines, which represent a particular form of immunotherapy. In the literature there are only few publications that systematically group and collectively discuss these studies. Therefore, the purpose of this review is to illustrate and give a partial reorganization to what has been produced in the literature so far. We provide basic notions on cancer immunotherapy and describe some clinical trials in which therapeutic cancer vaccines are tested. We thus focus attention on the potential of EV-based therapeutic vaccines in the treatment of cancer patients, overviewing the clinically relevant trials, completed or still in progress, which open up new perspectives in the fight against cancer.
Collapse
Affiliation(s)
- Carla Giacobino
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (C.G.); (M.C.)
| | - Marta Canta
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (C.G.); (M.C.)
| | - Cristina Fornaguera
- Grup d’Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain; (C.F.); (S.B.)
| | - Salvador Borrós
- Grup d’Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain; (C.F.); (S.B.)
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (C.G.); (M.C.)
- Correspondence:
| |
Collapse
|
18
|
Obaya AA, Mohammed AA, Rashied H, Morsy AM, Osman G, Allam AS, Elsayed AM, Harb OA, Elsayed WSH. Evaluating the value of Amphiregulin, Phosphatase and Tensin Homologue (PTEN) and P21 Expression for Anti-EGFR Treatment in Metastatic Colorectal Carcinoma. Asian Pac J Cancer Prev 2021; 22:1025-1034. [PMID: 33906293 PMCID: PMC8325139 DOI: 10.31557/apjcp.2021.22.4.1025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Despite the significant progress in target therapy for the treatment of metastatic colorectal carcinoma (mCRC), the overall survival isn't satisfactory. METHODS We assessed the expression of Amphiregulin, PTEN, and P21 in sections from 23 paraffin blocks prepared from 23 patients with left-sided mCRC using immunohistochemistry (IHC). The relationship between their level of expressions, clinicopathological parameters, response to anti-EGFR, and prognosis were analyzed. RESULTS High Amphiregulin, PTEN and low P21 expression levels were associated with low tumor grade (p= 0.038 and 0.025 respectively), better response to anti-EGFR treatment (p <0.001), and favorable outcome {progression-free survival (PFS) and overall survival (OS)} (p <0.05). There was a direct relation between Amphiregulin and PTEN expressions (phi coefficient=+0.840), while there was an inverse relation between P21expression and both Amphiregulin (phi coefficient= -0.840) and PTEN expressions (phi coefficient = -1.000), which was statistically significant (P <0.001). CONCLUSION High Amphiregulin and PTEN expression levels and low P21 expression levels were associated with better response to anti-EGFR therapy and improved survival outcome. They might be considered predictive markers of response to anti-EGFR therapy in mCRC.<br /> <br />.
Collapse
Affiliation(s)
- Ahmed Ali Obaya
- Department of Clinical Oncology & Nuclear Medicine, Faculty of Medicine, Zagazig University, Egypt
| | - Amrallah A Mohammed
- Department of Medical Oncology, Faculty of Medicine, Zagazig University, Egypt
| | - Hanaa Rashied
- Department of Clinical Oncology & Nuclear Medicine, Elmabara Hospital of Zagazig, Health Insurance Organization, Zagazig, Egypt
| | - Adel Mahmoud Morsy
- Department of General Surgery, Faculty of Medicine, Zagazig University, Egypt
| | - Gamal Osman
- Department of General Surgery, Faculty of Medicine, Zagazig University, Egypt
| | - Ahmed S Allam
- Department of Internal Medicine, Faculty of Medicine, Zagazig University, Egypt
| | - Ahmed M Elsayed
- Department of Tropical Medicine, Faculty of Medicine, Zagazig University, Egypt
| | - Ola A Harb
- Department of Pathology, Faculty of Medicine, Zagazig University, Egypt
| | - Walid S H Elsayed
- Department of Pathology, Faculty of Medicine, Zagazig University, Egypt
| |
Collapse
|
19
|
Subia B, Dahiya UR, Mishra S, Ayache J, Casquillas GV, Caballero D, Reis RL, Kundu SC. Breast tumor-on-chip models: From disease modeling to personalized drug screening. J Control Release 2021; 331:103-120. [PMID: 33417986 PMCID: PMC8172385 DOI: 10.1016/j.jconrel.2020.12.057] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023]
Abstract
Breast cancer is one of the leading causes of mortality worldwide being the most common cancer among women. Despite the significant progress obtained during the past years in the understanding of breast cancer pathophysiology, women continue to die from it. Novel tools and technologies are needed to develop better diagnostic and therapeutic approaches, and to better understand the molecular and cellular players involved in the progression of this disease. Typical methods employed by the pharmaceutical industry and laboratories to investigate breast cancer etiology and evaluate the efficiency of new therapeutic compounds are still based on traditional tissue culture flasks and animal models, which have certain limitations. Recently, tumor-on-chip technology emerged as a new generation of in vitro disease model to investigate the physiopathology of tumors and predict the efficiency of drugs in a native-like microenvironment. These microfluidic systems reproduce the functional units and composition of human organs and tissues, and importantly, the rheological properties of the native scenario, enabling precise control over fluid flow or local gradients. Herein, we review the most recent works related to breast tumor-on-chip for disease modeling and drug screening applications. Finally, we critically discuss the future applications of this emerging technology in breast cancer therapeutics and drug development.
Collapse
Affiliation(s)
- Bano Subia
- Elvesys Microfluidics Innovation Centre, Paris 75011, France..
| | | | - Sarita Mishra
- CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India..
| | - Jessica Ayache
- Elvesys Microfluidics Innovation Centre, Paris 75011, France..
| | | | - David Caballero
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| | - Subhas C Kundu
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| |
Collapse
|
20
|
Ferlizza E, Solmi R, Sgarzi M, Ricciardiello L, Lauriola M. The Roadmap of Colorectal Cancer Screening. Cancers (Basel) 2021; 13:1101. [PMID: 33806465 PMCID: PMC7961708 DOI: 10.3390/cancers13051101] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common form of cancer in terms of incidence and the second in terms of mortality worldwide. CRC develops over several years, thus highlighting the importance of early diagnosis. National screening programs based on fecal occult blood tests and subsequent colonoscopy have reduced the incidence and mortality, however improvements are needed since the participation rate remains low and the tests present a high number of false positive results. This review provides an overview of the CRC screening globally and the state of the art in approaches aimed at improving accuracy and participation in CRC screening, also considering the need for gender and age differentiation. New fecal tests and biomarkers such as DNA methylation, mutation or integrity, proteins and microRNAs are explored, including recent investigations into fecal microbiota. Liquid biopsy approaches, involving novel biomarkers and panels, such as circulating mRNA, micro- and long-non-coding RNA, DNA, proteins and extracellular vesicles are discussed. The approaches reported are based on quantitative PCR methods that could be easily applied to routine screening, or arrays and sequencing assays that should be better exploited to describe and identify candidate biomarkers in blood samples.
Collapse
Affiliation(s)
- Enea Ferlizza
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| | - Rossella Solmi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| | - Michela Sgarzi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| | - Luigi Ricciardiello
- Gastroenterology Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| |
Collapse
|
21
|
Wang S, Dong Y, Gong A, Kong H, Gao J, Hao X, Liu Y, Wang Z, Fan Y, Liu C, Xu W. Exosomal circRNAs as novel cancer biomarkers: Challenges and opportunities. Int J Biol Sci 2021; 17:562-573. [PMID: 33613113 PMCID: PMC7893596 DOI: 10.7150/ijbs.48782] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/19/2020] [Indexed: 12/14/2022] Open
Abstract
Identifying high specificity and sensitivity biomarkers has always been the focus of research in the field of non-invasive cancer diagnosis. Exosomes are extracellular vesicles with a lipid bilayer membrane that can be released by all types of cells, which contain a variety of proteins, lipids, and a variety of non-coding RNAs. Increasing research has shown that the lipid bilayer can effectively protect the nucleic acid in exosomes. In cancers, tumor cell-derived exosomal circRNAs can act on target cells or organs through the transport of exosomes, and then participate in the regulation of tumor development and metastasis. Since exosomes exist in various body fluids and circRNAs in exosomes exhibit high stability, exosomal circRNAs have the potential as biomarkers for early and minimally invasive cancer diagnosis and prognosis judgment. In this review, we summarized circRNAs and their biological roles in cancers, with the emerging value biomarkers in cancer diagnosis, disease judgment, and prognosis observation. In addition, we briefly compared the advantages of exosomal circRNAs as biomarkers and the current obstacles in the exosome isolation technology, shed light to the future development of this technology.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Yanhan Dong
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Anjing Gong
- Department of Neurosurgery, The affiliated hospital of Qingdao University, Qingdao 266003, China
| | - Huimin Kong
- School Hospital, Shandong University of Science and Technology, Qingdao 266003, China
| | - Jinning Gao
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Xiaodan Hao
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Yongmei Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Zibo Wang
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Yuqiao Fan
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Chengyu Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Wenhua Xu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| |
Collapse
|
22
|
Antunes‐Ferreira M, Koppers‐Lalic D, Würdinger T. Circulating platelets as liquid biopsy sources for cancer detection. Mol Oncol 2020; 15:1727-1743. [PMID: 33219615 PMCID: PMC8169446 DOI: 10.1002/1878-0261.12859] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/08/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Nucleic acids and proteins are shed into the bloodstream by tumor cells and can be exploited as biomarkers for the detection of cancer. In addition, cancer detection biomarkers can also be nontumor‐derived, having their origin in other organs and cell types. Hence, liquid biopsies provide a source of direct tumor cell‐derived biomolecules and indirect nontumor‐derived surrogate markers that circulate in body fluids or are taken up by circulating peripheral blood cells. The capacity of platelets to take up proteins and nucleic acids and alter their megakaryocyte‐derived transcripts and proteins in response to external signals makes them one of the richest liquid biopsy biosources. Platelets are the second most abundant cell type in peripheral blood and are routinely isolated through well‐established and fast methods in clinical diagnostics but their value as a source of cancer biomarkers is relatively recent. Platelets do not have a nucleus but have a functional spliceosome and protein translation machinery, to process RNA transcripts. Platelets emerge as important repositories of potential cancer biomarkers, including several types of RNAs (mRNA, miRNA, circRNA, lncRNA, and mitochondrial RNA) and proteins, and several preclinical studies have highlighted their potential as a liquid biopsy source for detecting various types and stages of cancer. Here, we address the usability of platelets as a liquid biopsy for the detection of cancer. We describe several studies that support the use of platelet biomarkers in cancer diagnostics and discuss what is still lacking for their implementation into the clinic.
Collapse
Affiliation(s)
- Mafalda Antunes‐Ferreira
- Department of NeurosurgeryCancer Center AmsterdamAmsterdam University Medical CentersVU University Medical CenterAmsterdamThe Netherlands
| | - Danijela Koppers‐Lalic
- Department of NeurosurgeryCancer Center AmsterdamAmsterdam University Medical CentersVU University Medical CenterAmsterdamThe Netherlands
| | - Thomas Würdinger
- Department of NeurosurgeryCancer Center AmsterdamAmsterdam University Medical CentersVU University Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
23
|
Mehryab F, Rabbani S, Shahhosseini S, Shekari F, Fatahi Y, Baharvand H, Haeri A. Exosomes as a next-generation drug delivery system: An update on drug loading approaches, characterization, and clinical application challenges. Acta Biomater 2020; 113:42-62. [PMID: 32622055 DOI: 10.1016/j.actbio.2020.06.036] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are small nanoparticles secreted by almost all cells and have a well-known role in intercellular communication. They are found in different body fluids and can also be isolated from cell culture media. They contain a natural cargo including various protein and nucleic acid molecules originated from their donor cells. In recent years, exosomes have emerged as a desired drug delivery system. They are believed to provide a targeted delivery of drug molecules, supplemented with their natural function. Furthermore, they have a membranous structure similar to liposomes, and that motivated researchers to apply their previous experience of drug loading into liposomes for exosomes. Herein, we discuss applied methods for the encapsulation of different drugs into exosomes, parameters affecting the incorporation of drug molecules into exosomes, characterization techniques, recent achievements, commercialization challenges and the potential future developments of exosomal drugs. Overall, while the application of exosomes as a drug delivery system is still in its infancy, they are considered to be a new class of natural nanocarriers with great potential for clinical application. Understanding of their key formulation parameters, pharmaceutical properties, in vivo behavior and applicable scale-up production will pave their way to the market. STATEMENT OF SIGNIFICANCE: Details of loading methods, characterization and biopharmaceutical properties of drug-incorporated exosomes are presented. Most parameters affecting encapsulation of drugs into exosomes are mentioned to serve as a guide for future studies in this field. Moreover, challenges on the way of exosomes to the market and clinic are described.
Collapse
|
24
|
Cheng Y, Qu X, Dong Z, Zeng Q, Ma X, Jia Y, Li R, Jiang X, Williams C, Wang T, Xia W. Comparison of serum exosome isolation methods on co-precipitated free microRNAs. PeerJ 2020; 8:e9434. [PMID: 32923177 PMCID: PMC7457927 DOI: 10.7717/peerj.9434] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/06/2020] [Indexed: 12/15/2022] Open
Abstract
Background Exosomes are nano-sized extracellular vesicles containing different biomolecules such as proteins and microRNAs (miRNAs) that mediate intercellular communication. Recently, numerous studies have reported the important functions of exosomal miRNAs in disease development and the potential clinical application as diagnostic biomarkers. Up to now, the most commonly used methods to extract exosomes are ultracentrifugation (UC) and precipitation-based commercial kit (e.g., ExoQuick). Generally, both UC and ExoQuick method could co-isolate contaminating proteins along with exosomes, with the UC method yielding even purer exosomes than ExoQuick. However, the comparison of these two methods on co-precipitated free miRNAs is still unknown. Methods In this study, we isolated exosomes from the human serum with exogenously added cel-miR-39 by UC and ExoQuick and compared the proportion of cel-miR-39 co-precipitated with exosomes extracted by these two methods. Results Using exogenous cel-miR-39 as free miRNAs in serum, we concluded that ExoQuick co-isolates a small proportion of free miRNAs while UC hardly precipitates any free miRNAs. We also found that incubation at 37 °C for 1 h could decrease the proportion of free miRNAs, and exosomal miRNAs like miR-126 and miR-152 also decreased when RNase A was used. In conclusion, our findings provide essential information about the details of serum exosome isolation methods for further research on exosomal miRNAs.
Collapse
Affiliation(s)
- Yirui Cheng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangyun Qu
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Zhaonan Dong
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Qingyu Zeng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xueqing Ma
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Yunli Jia
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Ruochen Li
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Xiaoxu Jiang
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Cecilia Williams
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Tao Wang
- Jiangsu Cancer Molecular Diagnostics Engineering Research Center, Suzhou MicroDiag Biomedicine Co., Ltd, Suzhou, China
| | - Weiliang Xia
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Wang W, Zhu N, Yan T, Shi YN, Chen J, Zhang CJ, Xie XJ, Liao DF, Qin L. The crosstalk: exosomes and lipid metabolism. Cell Commun Signal 2020; 18:119. [PMID: 32746850 PMCID: PMC7398059 DOI: 10.1186/s12964-020-00581-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/13/2020] [Indexed: 02/08/2023] Open
Abstract
Exosomes have been considered as novel and potent vehicles of intercellular communication, instead of "cell dust". Exosomes are consistent with anucleate cells, and organelles with lipid bilayer consisting of the proteins and abundant lipid, enhancing their "rigidity" and "flexibility". Neighboring cells or distant cells are capable of exchanging genetic or metabolic information via exosomes binding to recipient cell and releasing bioactive molecules, such as lipids, proteins, and nucleic acids. Of note, exosomes exert the remarkable effects on lipid metabolism, including the synthesis, transportation and degradation of the lipid. The disorder of lipid metabolism mediated by exosomes leads to the occurrence and progression of diseases, such as atherosclerosis, cancer, non-alcoholic fatty liver disease (NAFLD), obesity and Alzheimer's diseases and so on. More importantly, lipid metabolism can also affect the production and secretion of exosomes, as well as interactions with the recipient cells. Therefore, exosomes may be applied as effective targets for diagnosis and treatment of diseases. Video abstract.
Collapse
Affiliation(s)
- Wei Wang
- School of Pharmacy, Hanpu Science and Education District, Hunan University of Chinese Medicine, 300 Xueshi Road, Changsha, 410208, Hunan, China.,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Neng Zhu
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tao Yan
- School of Pharmacy, Hanpu Science and Education District, Hunan University of Chinese Medicine, 300 Xueshi Road, Changsha, 410208, Hunan, China.,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ya-Ning Shi
- School of Pharmacy, Hanpu Science and Education District, Hunan University of Chinese Medicine, 300 Xueshi Road, Changsha, 410208, Hunan, China.,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Chen
- Department of Neurosurgery in Changsha, 921 hospital, joint service support force of People's Liberation Army, Changsha, China
| | - Chan-Juan Zhang
- School of Pharmacy, Hanpu Science and Education District, Hunan University of Chinese Medicine, 300 Xueshi Road, Changsha, 410208, Hunan, China.,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xue-Jiao Xie
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Duan-Fang Liao
- School of Pharmacy, Hanpu Science and Education District, Hunan University of Chinese Medicine, 300 Xueshi Road, Changsha, 410208, Hunan, China. .,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| | - Li Qin
- School of Pharmacy, Hanpu Science and Education District, Hunan University of Chinese Medicine, 300 Xueshi Road, Changsha, 410208, Hunan, China. .,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
26
|
Eslami-S Z, Cortés-Hernández LE, Cayrefourcq L, Alix-Panabières C. The Different Facets of Liquid Biopsy: A Kaleidoscopic View. Cold Spring Harb Perspect Med 2020; 10:a037333. [PMID: 31548226 PMCID: PMC7263091 DOI: 10.1101/cshperspect.a037333] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The current limitations of cancer diagnosis and molecular profiling based on invasive tissue biopsies or clinical imaging have led to the development of the liquid biopsy field. Liquid biopsy includes the isolation of circulating tumor cells (CTCs), circulating free or tumor DNA (cfDNA or ctDNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) from body fluid samples and their molecular characterization to identify biomarkers for early cancer diagnosis, prognosis, therapeutic prediction, and follow-up. These innovative biosources show similar features as the primary tumor from where they originated or interacted. This review describes the different technologies and methods used for processing these biosources as well as their main clinical applications with their advantages and limitations.
Collapse
Affiliation(s)
- Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, 34093 Montpellier, France
| | - Luis Enrique Cortés-Hernández
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, 34093 Montpellier, France
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, 34093 Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, 34093 Montpellier, France
| |
Collapse
|
27
|
Liu X, Chu KM. Exosomal miRNAs as circulating biomarkers for prediction of development of haematogenous metastasis after surgery for stage II/III gastric cancer. J Cell Mol Med 2020; 24:6220-6232. [PMID: 32383554 PMCID: PMC7294143 DOI: 10.1111/jcmm.15253] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 12/24/2022] Open
Abstract
Exosomes secreted by living cancer cells can regulate metastasis. Exosomal miRNAs can reflect pathological conditions of the original cancer cells. Therefore, we aim to identify exosomal miRNAs as circulating biomarkers for haematogenous metastasis of gastric cancer. Pre‐treatment serum samples of eighty‐nine patients with stage II/III gastric cancer were collected. Thirty‐four of them developed haematogenous metastasis after surgery and the other fifty‐five did not. Extraction of exosomes was validated by western blot, transmission electron microscopy and nanoparticle tracking analysis. MiRNA qPCR array was performed in three matched pairs of samples. Internal control was selected from PCR array and validated in the remaining samples. Expressions of exosomal miRNAs were evaluated in the remaining samples by RT‐qPCR, as well as in gastric cancer tissue samples and cell culture medium. Expression levels of exosomal miRNAs were analysed with clinical characteristics. The results indicated thirteen up‐regulated and six down‐regulated miRNAs were found after normalization. MiR‐379‐5p and miR‐410‐3p were significantly up‐regulated in metastatic patients (P < .01). Higher expression of exosomal miR‐379‐5p or miR‐410‐3p showed shorter progression‐free survival of the patients (P < .05). It was also found that miR‐379‐5p and miR‐410‐3p were down‐regulated in gastric cancer tissue samples, while they were significantly up‐regulated in gastric cancer cell culture medium compared with cancer cells. In conclusion, exosomal miRNAs are promising circulating biomarkers for prediction of development of haematogenous metastasis after surgery for stage II/III gastric cancer.
Collapse
Affiliation(s)
- Xin Liu
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kent-Man Chu
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong
| |
Collapse
|
28
|
Cheng Y, Zeng Q, Han Q, Xia W. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell 2020; 10:295-299. [PMID: 29616487 PMCID: PMC6418301 DOI: 10.1007/s13238-018-0529-4] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Yirui Cheng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qingyu Zeng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qing Han
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Weiliang Xia
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
29
|
Aghebati-Maleki A, Nami S, Baghbanzadeh A, Karzar BH, Noorolyai S, Fotouhi A, Aghebati-Maleki L. Implications of exosomes as diagnostic and therapeutic strategies in cancer. J Cell Physiol 2019; 234:21694-21706. [PMID: 31161617 DOI: 10.1002/jcp.28875] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/28/2019] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Abstract
Exosomes offer a new perspective on the biology of cancer with both diagnostic and therapeutic concepts. Due to the cell-to-cell association, exosomes are involved in the progression, metastasis, and therapeutic efficacy of the tumor. They can be isolated from blood and other body fluids to determine the disease progression in the body, including cancer growth. In addition to being reservoirs of biochemical markers of cancer, exomes can be designed to restore tumor immunity. Tumor exosomes interact with different cells in the tumor microenvironment to confer beneficial modulations, responsible for stromal activity, angiogenesis, increased vascular permeability, and immune evasion. Exosomes also contribute to the metastasis with the aim of epithelial transmission to the mesenchyme and the formation of premetastatic niches. Moreover, exosomes protect cells against the cytotoxic effects of chemotherapeutic drugs and prevent the transmission of chemotherapy resistance to adjacent cells. Therefore, exosomes are essential for many fatal cancer agents, and understanding their origins and role in cancer is important. In this article, we attempted to clarify the potential of exosomes for the application in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Ali Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Nami
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bita H Karzar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Fotouhi
- Department of Orthopedic Surgery, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Modulating Pro-inflammatory Cytokines, Tissue Damage Magnitude, and Motor Deficit in Spinal Cord Injury with Subventricular Zone-Derived Extracellular Vesicles. J Mol Neurosci 2019; 70:458-466. [DOI: 10.1007/s12031-019-01437-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
|
31
|
Rahmati S, Shojaei F, Shojaeian A, Rezakhani L, Dehkordi MB. An overview of current knowledge in biological functions and potential theragnostic applications of exosomes. Chem Phys Lipids 2019; 226:104836. [PMID: 31678051 DOI: 10.1016/j.chemphyslip.2019.104836] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/19/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022]
Abstract
Exosomes are cup-shaped structures, made of two lipid layers. Their size is in the range of 30-150 nm. Exosomes are excreted to the extracellular space and function in local and systemic cellular communication. Based on their primary origins, they can contain substantial amounts of RNA, protein, and miRNA; the horizontal transfer of these contents significantly determines the exosome's biological effects. The endosomal origins of exosomes can be deduced based on their surface protein markers. The use of exosomes as a diagnostic biomarker and therapeutic tool, has numerous advantages because they do not pose risks such as aneuploidy and transplant rejection. This - overview highlights the recent findings in exosome development and current knowledge in exosome-based therapies.
Collapse
Affiliation(s)
- Shima Rahmati
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Fereshteh Shojaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Shojaeian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Leila Rezakhani
- Department of Tissue Reengineering, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehdi Banitalebi Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
32
|
Yu Y, Abudula M, Li C, Chen Z, Zhang Y, Chen Y. Icotinib-resistant HCC827 cells produce exosomes with mRNA MET oncogenes and mediate the migration and invasion of NSCLC. Respir Res 2019; 20:217. [PMID: 31606039 PMCID: PMC6790059 DOI: 10.1186/s12931-019-1202-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/27/2019] [Indexed: 12/26/2022] Open
Abstract
Background Icotinib has been widely used in patients with non-small cell lung cancer (NSCLC), and have significantly enhanced the overall survival rate of NSCLC patients. However, acquired drug resistance limits its clinical efficacy. Tumor cell-derived exosomes have been reported to participate in various biological processes, including tumor invasion, metastasis and drug resistance. Materials and methods In the present study, drug resistance was measured by MTT assay. Exosomes were extracted from the cell supernatant using ultracentrifugation and identified by exosomal marker. HCC827 cells were treated with exosomes derived from icotinib-resistant (IR) HCC827 to observe the invasion and migration of parent cells. The expression of exo-mRNA was analyzed by reverse transcription-quantitative polymerase chain reaction (RT-PCR). In addition, 10 exo-mRNAs detecting from the plasma and bronchoalveolar lavage fluid (BALF) of NSCLC patients with icotinib treatment were used to establish a new drug resistant-warning formula. Results The oncogene MET into exosomes was identified from icotinib-resistant lung cancer cells, and this was also presented in exosomes in NSCLC patients diagnosed with cancer metastasis after icotinib treatment. The knockdown of MET in exosomes significantly decreased the ability of invasion and migration in HCC827 cells. Conclusion It was suggested that MET might be specifically package and transferred by exosomes to modify the invasion and migration ability of the surrounding icotinib-sensitive cells.
Collapse
Affiliation(s)
- Yiming Yu
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | | | | | - Zhongbo Chen
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Yun Zhang
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Yichen Chen
- Ningbo Institution of Medical Science, Ningbo, China.
| |
Collapse
|
33
|
Galamb O, Barták BK, Kalmár A, Nagy ZB, Szigeti KA, Tulassay Z, Igaz P, Molnár B. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol 2019; 25:5026-5048. [PMID: 31558855 PMCID: PMC6747286 DOI: 10.3748/wjg.v25.i34.5026] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/26/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are members of the non-protein coding RNA family longer than 200 nucleotides. They participate in the regulation of gene and protein expression influencing apoptosis, cell proliferation and immune responses, thereby playing a critical role in the development and progression of various cancers, including colorectal cancer (CRC). As CRC is one of the most frequently diagnosed malignancies worldwide with high mortality, its screening and early detection are crucial, so the identification of disease-specific biomarkers is necessary. LncRNAs are promising candidates as they are involved in carcinogenesis, and certain lncRNAs (e.g., CCAT1, CRNDE, CRCAL1-4) show altered expression in adenomas, making them potential early diagnostic markers. In addition to being useful as tissue-specific markers, analysis of circulating lncRNAs (e.g., CCAT1, CCAT2, BLACAT1, CRNDE, NEAT1, UCA1) in peripheral blood offers the possibility to establish minimally invasive, liquid biopsy-based diagnostic tests. This review article aims to describe the origin, structure, and functions of lncRNAs and to discuss their contribution to CRC development. Moreover, our purpose is to summarise lncRNAs showing altered expression levels during tumor formation in both colon tissue and plasma/serum samples and to demonstrate their clinical implications as diagnostic or prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Orsolya Galamb
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| | - Barbara K Barták
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Alexandra Kalmár
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| | - Zsófia B Nagy
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Krisztina A Szigeti
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| | - Peter Igaz
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Béla Molnár
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| |
Collapse
|
34
|
Abstract
Exosomes are bilayer vesicles with particle sizes between 50 and 150 nm. Owing to their bilayer membrane structure, cell-to-cell communication, and good absorbability, exosomes are increasingly used as carriers for drug delivery through phospholipid membrane structures to lesion sites with enhanced targeting. Exosome sources and drug-loading methods are important factors affecting their use as drug carriers. There are various ways to pack species in exosomes, and researchers are constantly seeking new and improved approaches. In both in vivo and in vitro evaluations, exosomal vectors have achieved good antitumor efficacies. Despite the importance of exosomes as drug delivery systems with accurate targeting ability and biocompatibility, improvements are needed to facilitate their widespread clinical use. This review focuses on the preparation of exosomes as carriers and their utilization in antitumor research.
Collapse
Affiliation(s)
- Yanyan Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kuan Zhou
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
35
|
Jin Y, Long D, Li J, Yu R, Song Y, Fang J, Yang X, Zhou S, Huang S, Zhao Z. Extracellular vesicles in bone and tooth: A state-of-art paradigm in skeletal regeneration. J Cell Physiol 2019; 234:14838-14851. [PMID: 30847902 DOI: 10.1002/jcp.28303] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 02/05/2023]
Abstract
Bone and tooth, fundamental parts of the craniofacial skeleton, are anatomically and developmentally interconnected structures. Notably, pathological processes in these tissues underwent together and progressed in multilevels. Extracellular vesicles (EVs) are cell-released small organelles and transfer proteins and genetic information into cells and tissues. Although EVs have been identified in bone and tooth, particularly EVs have been identified in the bone formation and resorption, the concrete roles of EVs in bone and tooth development and diseases remain elusive. As such, we review the recent progress of EVs in bone and tooth to highlight the novel findings of EVs in cellular communication, tissue homeostasis, and interventions. This will enhance our comprehension on the skeletal biology and shed new light on the modulation of skeletal disorders and the potential of genetic treatment.
Collapse
Affiliation(s)
- Ying Jin
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Juan Li
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Ruichao Yu
- Department of Pulmonary, Brigham and Women's Hospital, Harvard Medical School, Massachusetts
| | - Yueming Song
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Fang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xi Yang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shu Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, P.R. China
| | - Shishu Huang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
36
|
Chen W, Li H, Su W, Qin J. Microfluidic device for on-chip isolation and detection of circulating exosomes in blood of breast cancer patients. BIOMICROFLUIDICS 2019; 13:054113. [PMID: 31893011 PMCID: PMC6932858 DOI: 10.1063/1.5110973] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/08/2019] [Indexed: 05/04/2023]
Abstract
Tumor-derived circulating exosomes have been recognized as a promising biomarker source for cancer diagnosis via a less invasive procedure. The integration of isolation and detection of exosomes in routine clinical settings is still challenging. In this study, we developed a new microfluidic device for immunomagnetic separation and detection of blood exosomes in situ. The microfluidic device may empower the integration of target exosome analysis via high surface to volume ratios of immunomagnetic beads and highly precise fluid control with the aid of microvalves. The obtained microfluidic device was capable of on-chip isolation and detection of circulating exosomes within 1.5 h. The captured exosomes could be directly visualized with an inverted fluorescence microscope in situ by tetramethylbenzidine-based colorimetric sensing. It was revealed that a statistically significant increase (p < 0.01) in EpCAM-positive exosomes was captured for cancer patients (n = 10) on the device when compared to healthy individuals (n = 10). The device also demonstrated high predicting accuracy for tumor exosomal markers with a sensitivity of 90% and a specificity of >95% using receiver operating characteristic curves. The microfluidic device might provide a new platform to assist cancer diagnosis and molecular classification in an automated and simple fashion.
Collapse
Affiliation(s)
| | - Hongjing Li
- First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Wentao Su
- Authors to whom correspondence should be addressed: and
| | - Jianhua Qin
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
37
|
Deng Y, Chen D, Gao F, Lv H, Zhang G, Sun X, Liu L, Mo D, Ma N, Song L, Huo X, Yan T, Zhang J, Miao Z. Exosomes derived from microRNA-138-5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2. J Biol Eng 2019; 13:71. [PMID: 31485266 PMCID: PMC6714399 DOI: 10.1186/s13036-019-0193-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 06/30/2019] [Indexed: 12/16/2022] Open
Abstract
Background MicroRNAs (miRNAs) are implicated in the progression of ischemic stroke (IS) and bone marrow-derived mesenchymal stem cells (BMSCs)-derived exosomes play a role in IS therapy. Herein we hypothesized that the BMSCs-derived exosomes containing overexpressed miR-138-5p could protect the astrocytes following IS involved with lipocalin 2 (LCN2). Methods The differentially expressed gene related to IS was initially identified by bioinformatics analysis. miR-138-5p was predicted to regulate LCN2. The expression of miR-138-5p and LCN2 was altered in the oxygen-glucose deprivation (OGD)-induced astrocytes. Furthermore, the cell behaviors and inflammatory responses were evaluated both in astrocytes alone and astrocytes co-cultured with exosomes derived from BMSCs overexpressing miR-138-5p to explore the involvement of miR-138-5p and LCN2 in IS. Besides, middle cerebral artery occlusion (MCAO) mouse model was established to explore the effect of BMSCs-derived exosomal miR-138-5p in IS in vivo. Results LCN2 was highly expressed in IS. Besides, LCN2 was a target gene of miR-138-5p. BMSCs-derived exosomes could be endocytosed by astrocytes via co-culture. Overexpression of miR-138-5p promoted the proliferation and inhibited apoptosis of astrocytes injured by OGD, accompanied by the reduced expression of inflammatory factors, which was achieved by down-regulating LCN2. More importantly, BMSCs delivered miR-138-5p to the astrocytes via exosomes and BMSCs-derived exosomal miR-138-5p alleviated neuron injury in IS mice. Conclusion BMSCs-derived exosomal miR-138-5p reduces neurological impairment by promoting proliferation and inhibiting inflammatory responses of astrocytes following IS by targeting LCN2, which may provide a novel target for IS treatment.
Collapse
Affiliation(s)
- Yiming Deng
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Duanduan Chen
- 4School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Feng Gao
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Hong Lv
- 5Departments of Clinical Laboratory, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050 People's Republic of China
| | - Guojun Zhang
- 5Departments of Clinical Laboratory, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050 People's Republic of China
| | - Xuan Sun
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Lian Liu
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Dapeng Mo
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Ning Ma
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Ligang Song
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Xiaochuan Huo
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Tianyi Yan
- 4School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Jingbo Zhang
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Zhongrong Miao
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| |
Collapse
|
38
|
Li FXZ, Liu JJ, Xu F, Lin X, Zhong JY, Wu F, Yuan LQ. Role of tumor-derived exosomes in bone metastasis. Oncol Lett 2019; 18:3935-3945. [PMID: 31579412 PMCID: PMC6757296 DOI: 10.3892/ol.2019.10776] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/05/2019] [Indexed: 12/13/2022] Open
Abstract
Tight coupling between bone resorption and formation is essential for bone remodeling. Disruption of this equilibrium can lead to skeletal disorders. Osseous metastatic disease is a severe consequence of tumor cell dissemination from numerous primary cancer sites, including the prostate, lungs and breasts. Metastatic disease is one of the most common causes of mortality in patients with cancer. Rapid advances in the therapeutic options for bone disease, including the use of bisphosphonates, have achieved effective clinical effects. However, the overall survival time of patients with bone metastatic has not significantly improved. Exosomes, which originate from tumor tissue and preferentially the bone, provide a reasonable way to understand the mechanism of neoplastic bone metastasis. Recently, several studies have indicated that tumor-derived exosomes are involved in cancer progression. However, the potential role that exosomes serve in the pathological communication between tumor and bone cells within the skeletal microenvironment remains an emerging field. The present review reports some recent findings on the detrimental roles of exosomes in bone metastasis. In addition, since exosomes are involved in metabolic organ cross-talk, this review highlights the involvement of cancer-derived exosomes in the regulation of skeletal metastatic diseases. Lastly, the potential promising clinical applications and emerging therapeutic opportunities targeting exosomes are discussed as novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Fu-Xing-Zi Li
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jun-Jie Liu
- Department of Pathology, The Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Feng Xu
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiao Lin
- Department of Geriatrics, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jia-Yu Zhong
- Department of Geriatrics, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Feng Wu
- Department of Pathology, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Ling-Qing Yuan
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
39
|
Guo XY, Xiao F, Li J, Zhou YN, Zhang WJ, Sun B, Wang G. Exosomes and pancreatic diseases: status, challenges, and hopes. Int J Biol Sci 2019; 15:1846-1860. [PMID: 31523187 PMCID: PMC6743302 DOI: 10.7150/ijbs.35823] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic disease, including pathologies such as acute pancreatitis (AP), chronic pancreatitis (CP), and pancreatic cancer (PC), is a complicated and dangerous clinical condition involving the disruption of exocrine or endocrine function. PC has one of the highest mortality rates among cancers due to insufficient diagnosis in early stages. Furthermore, efficient treatment options for the disease etiologies of AP and CP are lacking. Thus, the identification of new therapeutic targets and reliable biomarkers is required. As essential couriers in intercellular communication, exosomes have recently been confirmed to play an important role in pancreatic disease, but the specific underlying mechanisms are unknown. Herein, we summarize the current knowledge of exosomes in pancreatic disease with respect to diagnosis, molecular mechanisms, and treatment, proposing new ideas for the study of pancreatic disease.
Collapse
Affiliation(s)
- Xiao-Yu Guo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fan Xiao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jie Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yi-Nan Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wang-Jun Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
40
|
Bai Y, Lu Y, Wang K, Cheng Z, Qu Y, Qiu S, Zhou L, Wu Z, Liu H, Zhao J, Mao H. Rapid Isolation and Multiplexed Detection of Exosome Tumor Markers Via Queued Beads Combined with Quantum Dots in a Microarray. NANO-MICRO LETTERS 2019; 11:59. [PMID: 34137993 PMCID: PMC7770845 DOI: 10.1007/s40820-019-0285-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 06/10/2019] [Indexed: 05/04/2023]
Abstract
Tumor-derived exosomes are actively involved in cancer progression and metastasis and have emerged as a promising marker for cancer diagnosis in liquid biopsy. Because of their nanoscale size, complex biogenesis, and methodological limitations related to exosome isolation and detection, advancements in their analysis remain slow. Microfluidic technology offers a better analytic approach compared with conventional methods. Here, we developed a bead-based microarray for exosome isolation and multiplexed tumor marker detection. Using this method, exosomes are isolated by binding to antibodies on the bead surface, and tumor markers on the exosomes are detected through quantum dot (QD) probes. The beads are then uniformly trapped and queued among micropillars in the chip. This design benefits fluorescence observation by dispersing the signals into every single bead, thereby avoiding optical interference and enabling more accurate test results. We analyzed exosomes in the cell culture supernatant of lung cancer and endothelial cell lines, and different lung cancer markers labeled with three QD probes were used to conduct multiplexed detection of exosome surface protein markers. Lung cancer-derived samples showed much higher (~ sixfold-tenfold) fluorescence intensity than endothelial cell samples, and different types of lung cancer samples showed distinctive marker expression levels. Additionally, using the chip to detect clinical plasma samples from cancer patients showed good diagnostic power and revealed a well consistency with conventional tests for serological markers. These results provide insight into a promising method for exosome tumor marker detection and early-stage cancer diagnosis.
Collapse
Affiliation(s)
- Yanan Bai
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yunxing Lu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Kun Wang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Zule Cheng
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Youlan Qu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
- School of Stomatology, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Shihui Qiu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Lin Zhou
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Zhenhua Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Huiying Liu
- School of Stomatology, Dalian Medical University, Dalian, 116044, People's Republic of China.
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
41
|
Zeng Q, Hong S, Wang X, Cheng Y, Sun J, Xia W. Regulation of exosomes secretion by low-intensity pulsed ultrasound in lung cancer cells. Exp Cell Res 2019; 383:111448. [PMID: 31152706 DOI: 10.1016/j.yexcr.2019.05.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 01/06/2023]
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a noninvasive therapeutic method which gradually being used in clinic including cancers. Exosomes mediate intercellular communication functions in disease development and the potential clinical applications in diagnosis and therapy. However, few studies have discussed the relationship between LIPUS and exosomes. Herein, we show that low intensity (0.6-2.1 W/cm2 or 0.6-3.4 W/cm2) LIPUS promoted exosomes secretion whereas higher intensity (3.4-5.0 W/cm2 or 5.0 W/cm2) LIPUS inhibited exosomes secretion, and this phenomenon is associated with autophagy. Pretreatment with 3-MA or down-regulation of LC3 potentiated low intensity LIPUS's promotion of exosomes secretion and conferred resistance to higher intensity LIPUS's effects on exosomes secretion. Furthermore, pretreatment with PP242 attenuated LIPUS-influenced exosomes secretion while expression of constitutively active Akt (Ad-myr-Akt) elevated LIPUS-influenced exosomes secretion, implying mTOR-dependent mechanism involved. The findings indicate that LIPUS influences exosomes secretion by targeting mTOR-mediated LC3 signaling in SPC-A1 and SPC-A1-BM cells. Our data provided initial evidence to connect LIPUS and secretion of exosomes, and highlight that LIPUS may be exploited in exosome-related diseases.
Collapse
Affiliation(s)
- Qingyu Zeng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, Shanghai, 200030, China
| | - Shibin Hong
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, Shanghai, 200030, China
| | - Xue Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, West Huaihai Road 241, Shanghai, 20030, China
| | - Yirui Cheng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, Shanghai, 200030, China
| | - Junfeng Sun
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, Shanghai, 200030, China.
| | - Weiliang Xia
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, Shanghai, 200030, China.
| |
Collapse
|
42
|
Ren W, Zhang X, Li W, Feng Q, Feng H, Tong Y, Rong H, Wang W, Zhang D, Zhang Z, Tu S, Zhu X, Zhang Q. Exosomal miRNA-107 induces myeloid-derived suppressor cell expansion in gastric cancer. Cancer Manag Res 2019; 11:4023-4040. [PMID: 31190980 PMCID: PMC6511657 DOI: 10.2147/cmar.s198886] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 02/20/2019] [Indexed: 12/23/2022] Open
Abstract
Background: Myeloid-derived suppressor cells (MDSCs) promote immunosuppression in the tumor microenvironment, support tumor growth and survival, and may contribute to immunotherapy resistance. Recent studies showed that tumor-derived exosomes (TDEs) can induce MDSCs accumulation and expansion, the mechanisms of which are largely unknown. Methods: The morphologies and sizes of the exosomes was observed by using a JEM-1400 transmission electron microscope. MicroRNA(miR)-107 and ARG1, DICER1, PTEN, PI3K, AKT, mTOR, and NF-kB mRNAs were quantified by quantitative reverse tanscription PCR. Dual-Luciferase Reports Assay were used to examine the expression of genes which was targeted by miR-107. The expression of proteins were analyzed by using western blot. Results: MiR-107 was not only overexpressed in gastric cancer cells but also enriched in their secreted TDEs. Also, these miR-107 enriched TDEs could be taken up by HLA-DR-CD33+MDSCs, where miR-107 was able to target and suppress expression of DICER1 and PTEN genes. Dampened DICER1 expression supported expansion of MDSCs , while decreased PTEN led to activation of the PI3K pathway, resulting in increased ARG1 expression. Furthemore, gastric cancer-derived miR-107 TDEs, when dosed intravenously into mice, were also capable of inducing expansion of CD11b+Gr1+/high MDSCs in mouse peripheral blood and altering expression of DICER1, PTEN, ARG1, and NOS2 in the MDSCs. Conclusions: Our findings demonstrate for the first time that gastric cancer-secreted exosomes are able to deliver miR-107 to the host MDSCs where they induce their expansion and activition by targeting DICER1 and PTEN genes, thereby may provide novel cancer therapeutics target for gastric cancer.
Collapse
Affiliation(s)
- WeiHong Ren
- Department of Histology and Embryology, College of Basic Medicine, Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China.,Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - XuRan Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - WenBo Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - Qian Feng
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - HuiJie Feng
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - Yan Tong
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - Hao Rong
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - Wei Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - Dai Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - ZhenQiang Zhang
- Immunology Laboratory of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan Province, People's Republic of China
| | - ShiChun Tu
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - XiaoYan Zhu
- Department of Histology and Embryology, College of Basic Medicine, Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - QinXian Zhang
- Department of Histology and Embryology, College of Basic Medicine, Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| |
Collapse
|
43
|
Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two-edged sword in cancer therapy. Int J Nanomedicine 2019; 14:2847-2859. [PMID: 31114198 PMCID: PMC6488158 DOI: 10.2147/ijn.s200036] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells present in various adult tissues. Several studies suggest that MSCs secrete exosomes that perform as mediators in the tumor niche and play several roles in tumorigenesis, angiogenesis, and metastasis. In contrast, there are other studies supporting the tumor-suppressing effects of MSC-derived exosomes. Therefore, the exact association of MSC exosomes and tumor cells remains open to debate. This review aimed to demonstrate the present knowledge of MSC-derived exosomes in cancer research and to illustrate current approaches to make use of modified exosomes as a platform in therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Faezeh Vakhshiteh
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran,
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran,
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran,
| | - Seyed Nasser Ostad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Analysis of the thrombotic and fibrinolytic activities of tumor cell-derived extracellular vesicles. Blood Adv 2019; 2:1054-1065. [PMID: 29752284 DOI: 10.1182/bloodadvances.2017015479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/04/2018] [Indexed: 02/06/2023] Open
Abstract
Exosomes and microvesicles (MVs) are small extracellular vesicles secreted by tumor cells and are suggested to contribute to the thrombotic events that commonly occur in patients with advanced malignancies. Paradoxically, these vesicles have been reported to also possess fibrinolytic activity. To determine whether thrombotic or fibrinolytic activity is a predominant characteristic of these extracellular vesicles, we prepared exosomes and MVs from 2 breast cancer cell lines (MDA-MB-231 and MCF7), a lung cancer cell line (A549), and a leukemia cell line (NB4) and assayed their thrombotic and fibrinolytic activities. We observed that thrombotic activity was a common feature of MVs but not exosomes. Exosomes and/or MVs from several cell lines, with the exception of the A549 cell line, displayed fibrinolytic activity toward a pure fibrin clot, but only exosomes from MDA-MB-231 cells could degrade a fibrin clot formed in plasma. Increasing the malignant potential of MCF7 cells decreased the thrombotic activity of their MVs but did not alter their fibrinolytic activity. Decreasing the malignant potential of NB4 cells did not alter the thrombotic or fibrinolytic activity of their MVs or exosomes. Finally, the incubation of MDA-MB-231 cell-derived exosomes with A549 cells increased plasmin generation by these cells. Our data indicate that MVs generally have thrombotic activity, whereas thrombotic activity is not commonly observed for exosomes. Furthermore, exosomes and MVs generally do not display fibrinolytic activity under physiological conditions.
Collapse
|
45
|
Mousavi S, Moallem R, Hassanian SM, Sadeghzade M, Mardani R, Ferns GA, Khazaei M, Avan A. Tumor-derived exosomes: Potential biomarkers and therapeutic target in the treatment of colorectal cancer. J Cell Physiol 2019; 234:12422-12432. [PMID: 30637729 DOI: 10.1002/jcp.28080] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/07/2018] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is the third most common cause of cancer-related death in men and women in many countries. Early detection of CRC helps to prevent the advanced stages of the disease, and may thereby improve the survival of these patients. A noninvasive test with high specificity and sensitivity is required for this. Exosomes are lipid bilayer membrane nanovesicles that are released into most body fluids and especially in the microenvironment of cancer. They carry various proteins, lipids, and nucleic materials such as DNA, RNA, messenger RNA (mRNA), and microRNA (miRNA), and may also alter the function of target cells. In this review, we aimed to describe the biogenesis, composition, function, and the role of tumor-derived exosomes in cancer progression. Moreover, their applications in tumor diagnosis and treatment are described, with a particular focus on CRC.
Collapse
Affiliation(s)
- Sousan Mousavi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roya Moallem
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Sadeghzade
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Mardani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, Sussex, UK
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology and School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
46
|
Differential Exosomic Proteomic Patterns and Their Influence in Resveratrol Sensitivities of Glioblastoma Cells. Int J Mol Sci 2019; 20:ijms20010191. [PMID: 30621053 PMCID: PMC6337327 DOI: 10.3390/ijms20010191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/27/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the commonest primary brain malignancy with extremely poor prognosis. Resveratrol posseses anti-cancer effects, while GBM cells respond differently to it due to certain unknown reason(s). Because the tumor-derived exosomes are supposed to influence chemosensitivity, the exosomic proteins released from resveratrol-sensitive U251 and resveratrol-resistant glioblastoma LN428 cells are profiled before (N/Exo) and after drug treatment (Res/Exo) by label-free liquid chromatography-mass spectrometry (LC-MS). The therapeutic implications of the proteomic findings are estimated by gene ontology enrichment analysis (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG)-based bioinformatic analyses and further elucidated by exosome co-incubating. The results reveal that U251/N/Exo but not U251/Res/Exo enhances resveratrol sensitivity of resveratrol-resistant LN428 cells. The resveratrol sensitive properties of U251 cells are not altered by either LN428/N/Exo or LN428/Res/Exo. U251/N/Exo contains higher levels of chromatin silencing and epidermis development proteins, while U251/Res/Exo has more oxygen transport and G protein-coupled receptor. Both of LN428/N/Exo and LN428/Res/Exo are rich in the proteins related with nucleosome assembly, microtubule-based process and chromatin silencing. In conclusion, U251/N/Exo sensitizes LN428 cells to resveratrol via delivering drug sensitizing signals, suggesting the presence of additional factor(s) that may determine the resveratrol sensitivities of glioblastoma cells.
Collapse
|
47
|
Das CK, Jena BC, Banerjee I, Das S, Parekh A, Bhutia SK, Mandal M. Exosome as a Novel Shuttle for Delivery of Therapeutics across Biological Barriers. Mol Pharm 2018; 16:24-40. [PMID: 30513203 DOI: 10.1021/acs.molpharmaceut.8b00901] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The effective delivery of target-specific siRNA to the brain by exploiting the exosomes derived from dendritic cells renders the paradigm shift for the prospective use of nanosized exosomes as a delivery system. Although the in vivo targeting strategies by other nanovesicles like liposomes exist, still this novel exosome-based delivery approach holds an inclusive dominance of in vivo security and reduced immunogenicity. Achieving promising exosome-based delivery strategies warrants more desirable exploration of their biology. Over the years, the invention of novel production, characterization, targeting strategies, and cargo loading techniques of exosome improved its ability to reach clinics. Essentially, exosome-based delivery of therapeutics assures to conquer the major hurdles, like delivery of cargos across impermeable biological barriers, like the blood-brain barrier, biocompatibility, increased solubility, metabolic stability, improved circulation time, target specific delivery, and pharmacokinetics, and thereby enhanced the efficacy of loaded therapeutic agents. In this article, we cover the current status of exosome as a delivery vehicle for therapeutics and the challenges that need to be overcome, and we also discuss future perspectives of this exciting field of research to transform it from bench to clinical reality.
Collapse
Affiliation(s)
- Chandan Kanta Das
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Bikash Chandra Jena
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Indranil Banerjee
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Subhayan Das
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Aditya Parekh
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| | - Sujit Kumar Bhutia
- Department of Life Science , National Institute of Technology , Rourkela , India
| | - Mahitosh Mandal
- School of Medical Science and Technology , Indian Institute of Technology Kharagpur , Kharagpur , West Bengal 721302 , India
| |
Collapse
|
48
|
Chen Z, Cheng SB, Cao P, Qiu QF, Chen Y, Xie M, Xu Y, Huang WH. Detection of exosomes by ZnO nanowires coated three-dimensional scaffold chip device. Biosens Bioelectron 2018; 122:211-216. [DOI: 10.1016/j.bios.2018.09.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/09/2018] [Indexed: 12/20/2022]
|
49
|
The Expanding Role of Vesicles Containing Aquaporins. Cells 2018; 7:cells7100179. [PMID: 30360436 PMCID: PMC6210599 DOI: 10.3390/cells7100179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 12/11/2022] Open
Abstract
In animals and plants, membrane vesicles containing proteins have been defined as key for biological systems involving different processes such as trafficking or intercellular communication. Docking and fusion of vesicles to the plasma membrane occur in living cells in response to different stimuli, such as environmental changes or hormones, and therefore play an important role in cell homeostasis as vehicles for certain proteins or other substances. Because aquaporins enhance the water permeability of membranes, their role as proteins immersed in vesicles formed of natural membranes is a recent topic of study. They regulate numerous physiological processes and could hence serve new biotechnological purposes. Thus, in this review, we have explored the physiological implications of the trafficking of aquaporins, the mechanisms that control their transit, and the proteins that coregulate the migration. In addition, the importance of exosomes containing aquaporins in the cell-to-cell communication processes in animals and plants have been analyzed, together with their potential uses in biomedicine or biotechnology. The properties of aquaporins make them suitable for use as biomarkers of different aquaporin-related diseases when they are included in exosomes. Finally, the fact that these proteins could be immersed in biomimetic membranes opens future perspectives for new biotechnological applications.
Collapse
|
50
|
Sun Z, Shi K, Yang S, Liu J, Zhou Q, Wang G, Song J, Li Z, Zhang Z, Yuan W. Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer 2018; 17:147. [PMID: 30309355 PMCID: PMC6182840 DOI: 10.1186/s12943-018-0897-7] [Citation(s) in RCA: 539] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023] Open
Abstract
Exosomes, extracellular vesicles with diameters ranging from 30 to 150 nm, are widely present in various body fluids. Recently, microRNAs (miRNAs) have been identified in exosomes, the biogenesis, release, and uptake of which may involve the endosomal sorting complex required for transport (ESCRT complex) and relevant proteins. After release, exosomes are taken up by neighboring or distant cells, and the miRNAs contained within modulate such processes as interfering with tumor immunity and the microenvironment, possibly facilitating tumor growth, invasion, metastasis, angiogenesis and drug resistance. Therefore, exosomal miRNAs have a significant function in regulating cancer progression. Here, we briefly review recent findings regarding tumor-derived exosomes, including RNA sorting and delivering mechanism. We then describe the intercommunication occurring between different cells via exosomal miRNAs in tumor microenvironmnt, with impacts on tumor proliferation, vascularization, metastasis and other biological characteristics. Finally, we highlight the potential role of these molecules as biomarkers in cancer diagnosis and prognosis and tumor resistance to therapeutics.
Collapse
Affiliation(s)
- Zhenqiang Sun
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Ke Shi
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shuaixi Yang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinbo Liu
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Quanbo Zhou
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Guixian Wang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Junmin Song
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhen Li
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhiyong Zhang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Weitang Yuan
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|