1
|
Davidson CL, Kshirsagar P, Vengoji R, Shonka N. Exosomes in Glioma: Diagnostic and Therapeutic Potentials. ADVANCES IN ONCOLOGY 2025; 5:151-162. [PMID: 40443744 PMCID: PMC12119113 DOI: 10.1016/j.yao.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Affiliation(s)
- Caroline L. Davidson
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE 68198-8437, USA
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198-6840, USA
| |
Collapse
|
2
|
Fukutomi K, Fujimoto E, Shimokawatoko M, Takano E, Sunayama H, Takeuchi T, Tawa K. Single-Extracellular-Vesicle Detection with a Plasmonic Chip and Enhanced Fluorescence Microscopy. ACS OMEGA 2024; 9:44396-44406. [PMID: 39524643 PMCID: PMC11541535 DOI: 10.1021/acsomega.4c05678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/14/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Endocytosis-derived extracellular vesicles (EVs), which can be as small as 100 nm, are useful for disease prediction. However, very small EVs are below the optical diffraction limit and are difficult to visualize with conventional fluorescence microscopy. In this study, single EVs captured on a plasmonic chip, where fluorescently labeled antibodies were bound over the EV surface, were detected as bright spots using plasmon-field enhanced fluorescence without any pretreatment of isolating labeled EVs, followed by analyzing the full width at half-maximum and the fluorescence peak value for each enhanced fluorescence bright spot. Bright spots smaller than the threshold determined by the observation of the fluorescent nanospheres were attributed to single EVs. The number of single EVs was quantitatively evaluated against the concentration of EV solution injected in the 1.4 pM-95 fM range. Furthermore, single EVs were detected by labeling two different membrane proteins. A molecularly imprinted polymer was applied to a capture interface on a plasmonic chip, and it is found that nonspecific adsorption of aggregates was suppressed. To accurately distinguish single EVs from aggregates of labeled antibodies, the fluorescence microscopy with transmitted light was superior to the epifluorescence method. Finally, single EVs were successfully detected with multiple targets at multiple wavelengths by using different fluorescently labeled antibodies.
Collapse
Affiliation(s)
- Kazuma Fukutomi
- Graduate
School of Science and Technology, Kwansei
Gakuin University, 1
GakuenUegahara, Sanda, Hyogo 669-1330, Japan
| | - Eri Fujimoto
- Graduate
School of Science and Technology, Kwansei
Gakuin University, 1
GakuenUegahara, Sanda, Hyogo 669-1330, Japan
| | - Masaya Shimokawatoko
- Graduate
School of Science and Technology, Kwansei
Gakuin University, 1
GakuenUegahara, Sanda, Hyogo 669-1330, Japan
| | - Eri Takano
- Graduate
School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Hirobumi Sunayama
- Graduate
School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Toshifumi Takeuchi
- Innovation
Commercialization Division, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Keiko Tawa
- Graduate
School of Science and Technology, Kwansei
Gakuin University, 1
GakuenUegahara, Sanda, Hyogo 669-1330, Japan
| |
Collapse
|
3
|
Jouybari MT, Mojtahedi F, Babaahmadi M, Faeed M, Eslaminejad MB, Taghiyar L. Advancements in extracellular vesicle targeted therapies for rheumatoid arthritis: insights into cellular origins, current perspectives, and emerging challenges. Stem Cell Res Ther 2024; 15:276. [PMID: 39227964 PMCID: PMC11373471 DOI: 10.1186/s13287-024-03887-x] [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: 04/09/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024] Open
Abstract
Rheumatoid arthritis (RA) remains a challenging chronic autoimmune disorder characterized by persistent joint inflammation and damage. While modern regenerative strategies, encompassing cell/stem cell-based therapies, gene therapy, and tissue engineering, have advanced tissue repair efforts, a definitive cure for RA remains elusive. Consequently, there is growing interest in developing targeted therapies that directly address the underlying mechanisms driving RA pathogenesis, such as extracellular vesicles (EVs). These small membrane-bound particles can modulate immune responses within the inflammatory microenvironment of damaged cartilage. To launch the clinical potential of EVs, they can be isolated from various cell types through several techniques. EVs can carry various bioactive molecules and anti-inflammatory or pro-regenerative drugs, deliver them directly to the affected joints, and affect the behavior of injured cells, making them a compelling choice for targeted therapy and drug delivery in RA patients. However, there are still several challenges and limitations associated with EV-based therapy, including the absence of standardized protocols for EV isolation, characterization, and delivery. This review provides a comprehensive overview of the cellular sources of EVs in RA and delves into their therapeutic potential and the hurdles they must overcome.
Collapse
Affiliation(s)
- Maryam Talebi Jouybari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem Square, Banihashem St., Resalat Highway, PO Box: 16635-148, Tehran, 1665659911, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Fatemeh Mojtahedi
- Department of Immunology, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Mahnaz Babaahmadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem Square, Banihashem St., Resalat Highway, PO Box: 16635-148, Tehran, 1665659911, Iran
| | - Maryam Faeed
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mohammadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem Square, Banihashem St., Resalat Highway, PO Box: 16635-148, Tehran, 1665659911, Iran.
| | - Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem Square, Banihashem St., Resalat Highway, PO Box: 16635-148, Tehran, 1665659911, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
4
|
Jacobson R, Ha S, Tani S, Ghosh S, Jarajapu YPR, Brand RE, Kim J, Choi Y. Differential extracellular vesicle concentration and their biomarker expression of integrin α v/β 5, EpCAM, and glypican-1 in pancreatic cancer models. Sci Rep 2024; 14:14273. [PMID: 38902362 PMCID: PMC11189911 DOI: 10.1038/s41598-024-65209-8] [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: 10/25/2023] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
Tumor-derived extracellular vesicles (EVs) show great potential as biomarkers for several diseases, including pancreatic cancer, due to their roles in cancer development and progression. However, the challenge of utilizing EVs as biomarkers lies in their inherent heterogeneity in terms of size and concentration, making accurate quantification difficult, which is highly dependent on the isolation and quantification methods used. In our study, we compared three EV isolation techniques and two EV quantification methods. We observed variations in EV concentration, with approximately 1.5-fold differences depending on the quantification method used. Interestingly, all EV isolation techniques consistently yielded similar EV quantities, overall size distribution, and modal sizes. In contrast, we found a notable increase in total EV amounts in samples from pancreatic cancer cell lines, mouse models, and patient plasma, compared to non-cancerous conditions. Moreover, individual tumor-derived EVs exhibited at least a 3-fold increase in several EV biomarkers. Our data, obtained from EVs isolated using various techniques and quantified through different methods, as well as originating from various pancreatic cancer models, suggests that EV profiling holds promise for the identification of unique and cancer-specific biomarkers in pancreatic cancer.
Collapse
Affiliation(s)
- Reed Jacobson
- Departments of Physics, North Dakota State University, Fargo, ND, 58108, USA
- Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Sangdeuk Ha
- Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Sakurako Tani
- Departments of Physics, North Dakota State University, Fargo, ND, 58108, USA
| | - Shrinwanti Ghosh
- Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA
| | - Yagna P R Jarajapu
- Pharmaceutical Sciences, North Dakota State University, Fargo, ND, 58108, USA
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, ND, 58108, USA
| | - Randall E Brand
- Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15232, USA
| | - Jiha Kim
- Biological Sciences, North Dakota State University, Fargo, ND, 58108, USA.
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, ND, 58108, USA.
| | - Yongki Choi
- Departments of Physics, North Dakota State University, Fargo, ND, 58108, USA.
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, ND, 58108, USA.
| |
Collapse
|
5
|
Omrani M, Beyrampour-Basmenj H, Jahanban-Esfahlan R, Talebi M, Raeisi M, Serej ZA, Akbar-Gharalari N, Khodakarimi S, Wu J, Ebrahimi-Kalan A. Global trend in exosome isolation and application: an update concept in management of diseases. Mol Cell Biochem 2024; 479:679-691. [PMID: 37166542 PMCID: PMC10173230 DOI: 10.1007/s11010-023-04756-6] [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: 03/17/2023] [Accepted: 04/28/2023] [Indexed: 05/12/2023]
Abstract
Extracellular vesicles (EVs) secreted by various cells offer great potential for use in the diagnosis and treatment of disease. EVs are heterogeneous membranous vesicles. Exosomes are a subtype of EVs, 40-150 nm spherical vesicles with a lipid layer derived from endosomes. Exosomes, which are involved in signal transduction and maintain homeostasis, are released from almost all cells, tissues, and body fluids. Although several methods exist to isolate and characterize EVs and exosomes, each technique has significant drawbacks and limitations that prevent progress in the field. New approaches in the biology of EVs show great potential for isolating and characterizing EVs, which will help us better understand their biological function. The strengths and limitations of conventional strategies and novel methods (microfluidic) for EV isolation are outlined in this review. We also present various exosome isolation techniques and kits that are commercially available and assess the global market demand for exosome assays.
Collapse
Affiliation(s)
- Mohammadhassan Omrani
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Beyrampour-Basmenj
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Talebi
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mortaza Raeisi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Aliyari Serej
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naeimeh Akbar-Gharalari
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Khodakarimi
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jiaqian Wu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, 77030, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Ishimoto T, Yamashita R, Matsumoto R, Matsumoto S, Matsuo Y, Nakao S, Masuo Y, Suzuki M, Kato Y. TrkB phosphorylation in serum extracellular vesicles correlates with cognitive function enhanced by ergothioneine in humans. NPJ Sci Food 2024; 8:11. [PMID: 38321007 PMCID: PMC10847428 DOI: 10.1038/s41538-024-00250-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024] Open
Abstract
Oral administration of the food-derived antioxidant amino acid ergothioneine (ERGO) results in its efficient distribution in the brain and enhances cognitive function. However, effect of ERGO deficiency on cognitive impairment and the underlying mechanisms remain unknown. We revealed that cognitive function and hippocampal neurogenesis were lower in mice fed an ERGO-free diet than in those fed the control diet. Furthermore, ERGO supplementation to achieve the control diet ERGO levels reversed these effects and restored ERGO concentrations in the plasma and hippocampus. The ERGO-induced recovery of cognitive function and hippocampal neurogenesis was blocked by inhibiting the neurotrophic factor receptor tropomyosin receptor kinase B (TrkB), with a concomitant reduction in hippocampal phosphorylated TrkB, suggesting the involvement of TrkB in these events in mice. Phosphorylated TrkB was also detected in extracellular vesicles (EVs) derived from serum of volunteers who had been orally administered placebo or ERGO-containing tablets. Importantly, the ratio of serum EV-derived phosphorylated TrkB was significantly higher in the ERGO-treated group than in the placebo-treated group and was positively correlated with both serum ERGO concentrations and several cognitive domain scores from Cognitrax. Altogether, TrkB phosphorylation is involved in ERGO-induced cognitive enhancement in mice, and TrkB phosphorylation levels in serum EVs may quantitatively represent ERGO-induced cognitive enhancement in humans.
Collapse
Affiliation(s)
- Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Reiya Yamashita
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Ruri Matsumoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Satoshi Matsumoto
- L·S Corporation Co. Ltd., 3-10-1 Ningyocho-Nihonbashi, Chuo-ku, Tokyo, 103-0013, Japan
| | - Yusuke Matsuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Shunsuke Nakao
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Makoto Suzuki
- L·S Corporation Co. Ltd., 3-10-1 Ningyocho-Nihonbashi, Chuo-ku, Tokyo, 103-0013, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
7
|
Du Y, Zhang Y, Luo W, Gan F, Yang M, Gong P, Yao Y. The influence of radiation-induced bystander effect in osteoblasts mediated by plasma-derived extracellular vesicles (EVs). Biochem Biophys Res Commun 2024; 695:149425. [PMID: 38211533 DOI: 10.1016/j.bbrc.2023.149425] [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: 09/22/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVES Head and neck tumor patients may develop post-radiotherapy diseases after radiotherapy treatment. And radiotherapy can elicit radiation-induced bystander effect, wherein extracellular vesicles (EVs) play a crucial role. For normal parts of the body that have not been directly irradiated, the effect of EVs on them needs to be further explored. This study aims to investigate the functions of plasma-derived EVs in regulating normal osteoblasts during radiation-induced bystander effects. METHODS AND MATERIALS Rat plasma-derived EVs were isolated and identified firstly, followed by an evaluation of their intracellular biological effects on normal osteoblasts in vitro. Transcriptome sequencing analysis and confirmations were performed to identify potential mechanisms. RESULTS Irradiated plasma-derived EVs were found to enhance osteoblast proliferation, migration, and cell cycle progression, concurrently suppressing the expression of osteogenesis-related genes and proteins. Furthermore, these EVs attenuated the expression of osteogenesis and oxidative stress resistance related genes, while upregulating the PI3K-AKT pathway and intracellular reactive oxygen species in osteoblasts. CONCLUSIONS Irradiated plasma-derived EVs could alter the biological effects in osteoblasts, which is closely associated with the levels of GPX1 and the PI3K-AKT signaling pathway. This suggests that plasma-derived EVs serve as a crucial factor contributing to radiation-induced bystander effect in osteoblasts.
Collapse
Affiliation(s)
- Yu Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, China.
| | - Yixin Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, China.
| | - Wenqiong Luo
- Department of Stomatology, The First People's Hospital of Liangshan Yi Autonomous Prefecture, Sichuan province, China.
| | - Feihong Gan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, China.
| | - Mao Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, China.
| | - Ping Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, China.
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
8
|
Yi F, Xiao H, Song M, Huang L, Huang Q, Deng J, Yang H, Zheng L, Wang H, Gu W. BMSC-derived exosomal miR-148b-3p attenuates OGD/R-induced HMC3 cell activation by targeting DLL4 and Notch1. Neurosci Res 2024; 199:36-47. [PMID: 37741572 DOI: 10.1016/j.neures.2023.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023]
Abstract
Bone mesenchymal stem cell (BMSC)-derived exosome (BMSC-Exo) could be a treatment method for ischemic injury. In ischemic cerebrovascular disease (IC), microglia is pivotal in neuronal damage and remodeling. This study explores the mechanisms of BMSC-Exo miR-148b-3p in regulating oxygen-glucose deprivation/reoxygenation (OGD/R)-induced human microglial clone 3 (HMC3) cell activation. Transmission electron microscopy (TEM) and qNano were used to assess BMSC-Exo features. The functions of BMSC-Exo miR-148 b-3p in OGD/R-induced HMC3 cell activation were explored via MTT assay, flow cytometry, scratch, transwell, and enzyme-linked immunosorbent assay (ELISA) assays. A dual-luciferase reporter assay was performed to determine the relationship between miR-148b-3p and Delta-like ligand 4(DDL4) or neurogenic locus notch homolog protein 1 (Notch1). OGD/R decreased miR-148b-3p expression in HMC3 cells. After BMSC-Exo treatment, miR-148b-3p expression was upregulated, cell viability and migration were inhibited, cell cycles remained in the G0/G1 phase, and proinflammatory cytokines were decreased in OGD/R-induced HMC3 cells. More importantly, BMSC-Exo miR-148b-3p could further strengthen BMSC-Exo effects. DDL4 and Notch1 are direct targets of miR-148b-3p, respectively. Moreover, the knockdown of DLL4 or Notch1 could inhibit OGD/R-induced HMC3 cell activation. BMSC-Exo miR-148b-3p inhibited OGD/R-induced HMC3 cell activation via inhibiting DLL4 and Notch1 expression, which provided a new strategy for treating cerebral ischemia.
Collapse
Affiliation(s)
- Fang Yi
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Hui Xiao
- Department of Neurology, Changsha Central Hospital, Changsha 410004, Hunan, PR China
| | - Mingyu Song
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan 410008, PR China
| | - Lei Huang
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410000, Hunan, PR China
| | - Qianyi Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jun Deng
- Department of Neurology, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha 410000, Hunan, PR China
| | - Han Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Lan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Hong Wang
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan 410008, PR China.
| |
Collapse
|
9
|
Yu F, Chen Y, Yi W, Guan M, Lin N, Zhuo Y, Lin J, Lai F. Lung-specific exosomes for doxorubicin delivery in lung adenocarcinoma therapy. Biotechnol J 2024; 19:e2300296. [PMID: 38403456 DOI: 10.1002/biot.202300296] [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: 06/16/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 02/27/2024]
Abstract
Doxorubicin (DOX) could be utilized to treat lung adenocarcinoma (LUAD), while dose-limiting cardiotoxicity limits its clinical utilization. MDA-MB-231 cell-derived exosomes show lung-specific organotropism features. In this study, we aimed to explore the potential of MDA-MB-231 cell-derived exosomes in DOX specific delivery to the lung. MDA-MB-231 cell-derived exosomes were coincubated with to construct for the doxorubicin delivery system (D-EXO). Exosomes labeled with fluorescein isothiocyanate were incubated with A549 cells or 293T cells, and the engulf and the mean intensity of the fluorescence were detected with immunofluorescence and flow cytometry assay. Cell viability was detected with cell counting kit-8 (CCK-8), and cell migration was determined by scratch test. The protein expression was detected by Western blot assay. A549 cell line-derived xenograft mouse model was constructed to examine the treatment effect of D-EXO. MDA-MB-231 cell-derived exosomes could be specially taken up by A549 cells with diminished cell viability but not engulfed by 293T cells. D-EXO inhibited A549 cell migration, and upregulated the protein expression of caspase 3 and cleaved caspase 3 expression, while did not show any inhibition on 293T cells. In vivo orthotopic xenotransplantation model indicated that D-EXO inhibited tumor growth characterized by diminished tumor weight and improved survival rate. No significant change in body weight was observed after the D-EXO treatment. In conclusion, D-EXO proposed in this study could be utilized to treat LUAD with lung-specific delivery effects to improve the survival rate.
Collapse
Affiliation(s)
- Fengqiang Yu
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yanxun Chen
- Department of Thoracic Surgery, Quangang District Hospital, Quanzhou, China
| | - Weiqiang Yi
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Maohao Guan
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Nanlong Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yi Zhuo
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianbo Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fancai Lai
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
10
|
Singh S, Dansby C, Agarwal D, Bhat PD, Dubey PK, Krishnamurthy P. Exosomes: Methods for Isolation and Characterization in Biological Samples. Methods Mol Biol 2024; 2835:181-213. [PMID: 39105917 DOI: 10.1007/978-1-0716-3995-5_17] [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] [Indexed: 08/07/2024]
Abstract
Exosomes are small lipid bilayer-encapsulated nanosized extracellular vesicles of endosomal origin. Exosomes are secreted by almost all cell types and are a crucial player in intercellular communication. Exosomes transmit cellular information from donor to recipient cells in the form of proteins, lipids, and nucleic acids and influence several physiological and pathological responses. Due to their capacity to carry a variety of cellular cargo, low immunogenicity and cytotoxicity, biocompatibility, and ability to cross the blood-brain barrier, these nanosized vesicles are considered excellent diagnostic tools and drug-delivery vehicles. Despite their tremendous potential, the progress in therapeutic applications of exosomes is hindered by inadequate isolation techniques, poor characterization, and scarcity of specific biomarkers. The current research in the field is focused on overcoming these limitations. In this chapter, we have reviewed conventional exosome isolation and characterization methods and recent advancements, their advantages and limitations, persistent challenges in exosome research, and future directions.
Collapse
Affiliation(s)
- Sarojini Singh
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cassidy Dansby
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Divyanshi Agarwal
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Purnima Devaki Bhat
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Praveen Kumar Dubey
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
Amina SJ, Azam T, Dagher F, Guo B. A review on the use of extracellular vesicles for the delivery of drugs and biological therapeutics. Expert Opin Drug Deliv 2024; 21:45-70. [PMID: 38226932 DOI: 10.1080/17425247.2024.2305115] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
INTRODUCTION Exosomes, a type of extracellular vesicles, are effective tools for delivering small-molecule drugs and biological therapeutics into cells and tissues. Surface modifications with targeting ligands ensure precise delivery to specific cells, minimizing accumulation in healthy organs and reducing the side effects. This is a rapidly growing area in drug delivery research and this review aims to comprehensively discuss the recent advances in the field. AREA COVERED Recent studies have presented compelling evidence supporting the application of exosomes as efficient delivery vehicles that escape endosome trapping, achieving effective in vivo delivery in animal models. This review provides a systemic discussion on the exosome-based delivery technology, with topics covering exosome purification, surface modification, and targeted delivery of various cargos ranging from siRNAs, miRNAs, and proteins, to small molecule drugs. EXPERT OPINION Exosome-based gene and drug delivery has low toxicity and low immunogenicity. Surface modifications of the exosomes can effectively avoid endosome trapping and increase delivery efficiency. This exciting technology can be applied to improve the treatments for a wide variety of diseases.
Collapse
Affiliation(s)
- Sundus Jabeen Amina
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Tasmia Azam
- School of Chemical and Materials Engineering (SCME), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Fatima Dagher
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Bin Guo
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| |
Collapse
|
12
|
Cui Z, Amevor FK, Zhao X, Mou C, Pang J, Peng X, Liu A, Lan X, Liu L. Potential therapeutic effects of milk-derived exosomes on intestinal diseases. J Nanobiotechnology 2023; 21:496. [PMID: 38115131 PMCID: PMC10731872 DOI: 10.1186/s12951-023-02176-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 12/21/2023] Open
Abstract
Exosomes are extracellular vesicles with the diameter of 30 ~ 150 nm, and are widely involved in intercellular communication, disease diagnosis and drug delivery carriers for targeted disease therapy. Therapeutic application of exosomes as drug carriers is limited due to the lack of sources and methods for obtaining adequate exosomes. Milk contains abundant exosomes, several studies have shown that milk-derived exosomes play crucial roles in preventing and treating intestinal diseases. In this review, we summarized the biogenesis, secretion and structure, current novel methods used for the extraction and identification of exosomes, as well as discussed the role of milk-derived exosomes in treating intestinal diseases, such as inflammatory bowel disease, necrotizing enterocolitis, colorectal cancer, and intestinal ischemia and reperfusion injury by regulating intestinal immune homeostasis, restoring gut microbiota composition and improving intestinal structure and integrity, alleviating conditions such as oxidative stress, cell apoptosis and inflammation, and reducing mitochondrial reactive oxygen species (ROS) and lysosome accumulation in both humans and animals. In addition, we discussed future prospects for the standardization of milk exosome production platform to obtain higher concentration and purity, and complete exosomes derived from milk. Several in vivo clinical studies are needed to establish milk-derived exosomes as an effective and efficient drug delivery system, and promote its application in the treatment of various diseases in both humans and animals.
Collapse
Affiliation(s)
- Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, P. R. China
| | - Xingtao Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Sichuan, P. R. China
| | - Chunyan Mou
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Jiaman Pang
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xie Peng
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Anfang Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xi Lan
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
- College of Animal Science and Technology, Chongqing Key Laboratory of Forage & Herbivore, Chongqing Engineering Research Center for Herbivores Resource Protection and Utilization, Southwest University, Beibei, Chongqing, 400715, P. R. China.
| |
Collapse
|
13
|
Cheng Z, Shang J, Wang H, Yu L, Yuan Z, Zhang Y, Du Y, Tian J. Molecular imaging-guided extracellular vesicle-based drug delivery for precise cancer management: Current status and future perspectives. J Control Release 2023; 362:97-120. [PMID: 37625599 DOI: 10.1016/j.jconrel.2023.08.040] [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: 05/18/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Extracellular vesicles (EVs), the mediators of intercellular communication, have attracted the attention of researchers for the important roles they play in cancer treatment. Compared with other inorganic nano-materials, EVs possess the advantages of higher biocompatibility, better physiochemical stability, easier surface modification, and excellent biosafety. They can be used as an advanced drug delivery system with an improved therapeutic index for various therapeutic agents. Engineered EV-based imaging and therapeutic agents (engineered EVs) have emerged as useful tools in targeted cancer diagnosis and therapy. Non-invasive tracing of engineered EVs contributes to a better evaluation of their functions in cancer progression, in vivo dynamic biodistribution, therapeutic response, and drug-loading efficiency. Recent advances in real-time molecular imaging (MI), and innovative EV labeling strategies have led to the development of novel tools that can evaluate the pharmacokinetics of engineered EVs in cancer management, which may accelerate further clinical translation of novel EV-based drug delivery platforms. Herein, we review the latest advances in EVs, their characteristics, and current examples of EV-based targeted drug delivery for cancer. Then, we discuss the prominent applications of MI for tracing both natural and engineered EVs. Finally, we discuss the current challenges and considerations of EVs in targeted cancer treatment and the limitations of different MI modalities. In the coming decades, EV-based therapeutic applications for cancer with improved drug loading and targeting abilities will be developed, and better anti-cancer effects of drug delivery nanoplatform will be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jihuan Shang
- School of Clinical Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Huarong Wang
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China
| | - Leyi Yu
- Beijing Haidian Hospital, Beijing 100080, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China.
| | - Yinlong Zhang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, China; Science and Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
14
|
Liu R, Jia R, Wang D, Mirkin MV. Elucidating the Shape of Current Transients in Electrochemical Resistive-Pulse Sensing of Single Liposomes. Anal Chem 2023; 95:13756-13761. [PMID: 37676905 DOI: 10.1021/acs.analchem.3c02476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Electrochemical resistive-pulse (ERP) sensing with conductive carbon nanopipettes (CNPs) has recently been developed and employed for the detection of single liposomes and biological vesicles, and for the analysis of redox molecules contained in such vesicles. However, the origins of different shapes of current transients produced by the translocation of single vesicles through the CNP remain poorly understood. Herein, we report extensive finite-element simulations of both portions of an ERP transient, the current blockage by a vesicle approaching and passing through the pipet orifice and the faradaic current spike due to oxidation/reduction of redox species released from a vesicle on the carbon surface, for different values of parameters defining the geometry and dynamics of the vesicle/CNP system. The effects of the pipet geometry, surface charge, transport, vesicle trajectory, and collision location on the shape of current transients are investigated. The possibility of quantitative analysis of experimental ERP transients produced by translocations of liposomes and extracellular vesicles by fitting them to simulated curves is demonstrated. The developed theory can enable a more reliable interpretation of complicated ERP signals and characterization of the size and contents of single biological and artificial vesicles.
Collapse
Affiliation(s)
- Rujia Liu
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Rui Jia
- Department of Chemistry and Biochemistry, Queens College - CUNY, Flushing, New York 11367, United States
- The Graduate Center of City University of New York, New York, New York 10016, United States
| | - Dengchao Wang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Michael V Mirkin
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
- The Graduate Center of City University of New York, New York, New York 10016, United States
| |
Collapse
|
15
|
Back SJ, Kim W, Kim DY, Kim SJ, Hwang SR, Jung GB. Rapid and simple isolation and detection of exosomes using CaTiO 3:Eu 3+@Fe 3O 4 multifunctional nanocomposites. Anal Biochem 2023; 673:115161. [PMID: 37201773 DOI: 10.1016/j.ab.2023.115161] [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: 02/01/2023] [Revised: 03/27/2023] [Accepted: 04/18/2023] [Indexed: 05/20/2023]
Abstract
Exosomes are potential biomarkers for disease diagnosis and treatment, as well as drug carriers. However, as their isolation and detection remain critical issues, convenient, rapid, low-cost, and effective methods are necessary. In this study, we present a rapid and simple method for directly capturing and analyzing exosomes from complex cell culture media using CaTiO3:Eu3+@Fe3O4 multifunctional nanocomposites. The CaTiO3:Eu3+@Fe3O4 nanocomposites were prepared by high-energy ball-milling and used to isolate exosomes by binding CaTiO3:Eu3+@Fe3O4 nanocomposites and the hydrophilic phosphate head of the exosome phospholipids. Notably, the developed CaTiO3:Eu3+@Fe3O4 multifunctional nanocomposites achieved results comparable with those of commercially available TiO2 and were separated using a magnet within 10 min. Moreover, we report a surface-enhanced Raman scattering (SERS)-based immunoassay for detecting the exosome biomarker CD81. Gold nanorods (Au NRs) were modified with detection antibodies, and antibody-conjugated Au NRs were labeled with 3, 3, diethylthiatricarbocyanine iodide (DTTC) as the SERS tags. A method combining magnetic separation and SERS was developed to detect exosomal biomarker CD81. The results of this study demonstrate the feasibility of this new technique as a useful tool for exosome isolation and detection.
Collapse
Affiliation(s)
- Sung Jin Back
- Department of Physics Education, Chosun University, Gwangju, 61452, Republic of Korea
| | - Woong Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
| | - Da Young Kim
- Department of Physics Education, Chosun University, Gwangju, 61452, Republic of Korea
| | - Seok-Jun Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Gyeong Bok Jung
- Department of Physics Education, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
16
|
Zhang E, Phan P, Zhao Z. Cellular nanovesicles for therapeutic immunomodulation: A perspective on engineering strategies and new advances. Acta Pharm Sin B 2023; 13:1789-1827. [PMID: 37250173 PMCID: PMC10213819 DOI: 10.1016/j.apsb.2022.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023] Open
Abstract
Cellular nanovesicles which are referred to as cell-derived, nanosized lipid bilayer structures, have emerged as a promising platform for regulating immune responses. Owing to their outstanding advantages such as high biocompatibility, prominent structural stability, and high loading capacity, cellular nanovesicles are suitable for delivering various immunomodulatory molecules, such as small molecules, nucleic acids, peptides, and proteins. Immunomodulation induced by cellular nanovesicles has been exploited to modulate immune cell behaviors, which is considered as a novel cell-free immunotherapeutic strategy for the prevention and treatment of diverse diseases. Here we review emerging concepts and new advances in leveraging cellular nanovesicles to activate or suppress immune responses, with the aim to explicate their applications for immunomodulation. We overview the general considerations and principles for the design of engineered cellular nanovesicles with tailored immunomodulatory activities. We also discuss new advances in engineering cellular nanovesicles as immunotherapies for treating major diseases.
Collapse
Affiliation(s)
- Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
- Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Comparative analysis of magnetically activated cell sorting and ultracentrifugation methods for exosome isolation. PLoS One 2023; 18:e0282238. [PMID: 36854030 PMCID: PMC9974127 DOI: 10.1371/journal.pone.0282238] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Mesenchymal stem cell-derived exosomes regulate cell migration, proliferation, differentiation, and synthesis of the extracellular matrix, giving great potential for the treatment of different diseases. The ultracentrifugation method is the gold standard method for exosome isolation due to the simple protocol, and high yield, but presents low purity and requires specialized equipment. Amelioration of technical optimization is required for quick and reliable confinement of exosomes to translate them to the clinic as cell therapeutics In this study, we hypothesized that magnetically activated cell sorting may provide, an effective, reliable, and rapid tool for exosome isolation when compared to ultracentrifugation. We, therefore, aimed to compare the efficiency of magnetically activated cell sorting and ultracentrifugation for human mesenchymal stem cell-derived exosome isolation from culture media by protein quantification, surface biomarker, size, number, and morphological analysis. Magnetically activated cell sorting provided a higher purity and amount of exosomes that carry visible magnetic beads when compared to ultracentrifugation. The particle number of the magnetically activated cell sorting group was higher than the ultracentrifugation. In conclusion, magnetically activated cell sorting presents a quick, and reliable method to collect and present human mesenchymal stem cell exosomes to clinics at high purity for potential cellular therapeutic approaches. The novel isolation and purification method may be extended to different clinical protocols using different autogenic or allogeneic cell sources.
Collapse
|
18
|
Xu X, Xiang Y, Yang Y, Liu K, Cui Z, Tong X, Chen J, Hou F, Luo Z. The application of tumor cell-derived vesicles in oncology therapy. Clin Transl Oncol 2023; 25:364-374. [PMID: 36207510 DOI: 10.1007/s12094-022-02966-w] [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: 07/28/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Tumor cell-derived vesicles are released by tumor cells, have a phospholipid bilayer, and are widely distributed in various biological fluids. In recent years, it has been found that tumor cell-derived vesicles contain proteins, metabolites and nucleic acids and can be delivered to recipient cells to perform their physiological functions, such as mediating specific intercellular communication, activating or inhibiting signaling pathways, participating in regulating the modulation of tumor microenvironment and influencing tumor development, which can be used for early detection and diagnosis of cancer. In addition, tumor cell-derived vesicles exhibit multiple properties in tumor therapeutic applications and may serve as a new class of delivery systems. In this review, we elaborate on the application of tumor cell-derived vesicles in oncology therapy.
Collapse
Affiliation(s)
- Ximei Xu
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China.
| | - Yin Xiang
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Yang Yang
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Kai Liu
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Zhiwei Cui
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Xiaodong Tong
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Junliang Chen
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Fang Hou
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Zhiqiang Luo
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| |
Collapse
|
19
|
Progress of Endogenous and Exogenous Nanoparticles for Cancer Therapy and Diagnostics. Genes (Basel) 2023; 14:genes14020259. [PMID: 36833186 PMCID: PMC9957423 DOI: 10.3390/genes14020259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
The focus of this brief review is to describe the application of nanoparticles, including endogenous nanoparticles (e.g., extracellular vesicles, EVs, and virus capsids) and exogenous nanoparticles (e.g., organic and inorganic materials) in cancer therapy and diagnostics. In this review, we mainly focused on EVs, where a recent study demonstrated that EVs secreted from cancer cells are associated with malignant alterations in cancer. EVs are expected to be used for cancer diagnostics by analyzing their informative cargo. Exogenous nanoparticles are also used in cancer diagnostics as imaging probes because they can be easily functionalized. Nanoparticles are promising targets for drug delivery system (DDS) development and have recently been actively studied. In this review, we introduce nanoparticles as a powerful tool in the field of cancer therapy and diagnostics and discuss issues and future prospects.
Collapse
|
20
|
Nejabati HR, Roshangar L, Nouri M. Follicular fluid extracellular vesicle miRNAs and ovarian aging. Clin Chim Acta 2023; 538:29-35. [PMID: 36368351 DOI: 10.1016/j.cca.2022.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
The decrease in the reproductive potential due to aging occurs as a gradual decline in the quantity and quality of the ovarian reserve, a phenomenon associated with risk of miscarriage, pregnancy loss, low ovarian stimulation, and oocyte abnormalities, such as chromosomal aneuploidies. Numerous studies have shown that the fertility potential of older women is decreased by changes to the cellular composition of the follicles. Additionally, a unique method of cellular communication has been identified which involves the release of extracellular vesicles (EVs) in various body fluids including follicular fluid (FF). The changing composition of EVs especially non-coding RNAs, such as miRNAs has been documented across a broad range of cell types during aging. Accordingly, alterations of miRNA cargo within FF-derived EVs due to increased age may serve as a potential predictor of oocyte quality. In this review we examine the relationship between FF EV miRNAs and ovarian aging.
Collapse
Affiliation(s)
- Hamid Reza Nejabati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Alptekin A, Parvin M, Chowdhury HI, Rashid MH, Arbab AS. Engineered exosomes for studies in tumor immunology. Immunol Rev 2022; 312:76-102. [PMID: 35808839 DOI: 10.1111/imr.13107] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Exosomes are a type of extracellular vesicle (EV) with diameters of 30-150 nm secreted by most of the cells into the extracellular spaces and can alter the microenvironment through cell-to-cell interactions by fusion with the plasma membrane and subsequent endocytosis and release of the cargo. Because of their biocompatibility, low toxicity and immunogenicity, permeability (even through the blood-brain barrier (BBB)), stability in biological fluids, and ability to accumulate in the lesions with higher specificity, investigators have started making designer's exosomes or engineered exosomes to carry biologically active protein on the surface or inside the exosomes as well as using exosomes to carry drugs, micro RNA, and other products to the site of interest. In this review, we have discussed biogenesis, markers, and contents of various exosomes including exosomes of immune cells. We have also discussed the current methods of making engineered and designer's exosomes as well as the use of engineered exosomes targeting different immune cells in the tumors, stroke, as well as at peripheral blood. Genetic engineering and customizing exosomes create an unlimited opportunity to use in diagnosis and treatment. Very little use has been discovered, and we are far away to reach its limits.
Collapse
Affiliation(s)
- Ahmet Alptekin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Mahrima Parvin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | | | | | - Ali S Arbab
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
22
|
Kwon Y, Park J. Methods to analyze extracellular vesicles at single particle level. MICRO AND NANO SYSTEMS LETTERS 2022. [DOI: 10.1186/s40486-022-00156-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
AbstractExtracellular vesicles (EVs) are nano-sized vesicles derived from cells that transport biomaterials between cells through biofluids. Due to their biological role and components, they are considered as potential drug carriers and for diagnostic applications. Today's advanced nanotechnology enables single-particle-level analysis that was difficult in the past due to its small size below the diffraction limit. Single EV analysis reveals the heterogeneity of EVs, which could not be discovered by various ensemble analysis methods. Understanding the characteristics of single EVs enables more advanced pathological and biological researches. This review focuses on the advanced techniques employed for EV analysis at the single particle level and describes the principles of each technique.
Collapse
|
23
|
Xie Y, Xu X, Lin J, Xu Y, Wang J, Ren Y, Wu A. Effective Separation of Cancer-Derived Exosomes in Biological Samples for Liquid Biopsy: Classic Strategies and Innovative Development. GLOBAL CHALLENGES (HOBOKEN, NJ) 2022; 6:2100131. [PMID: 36176940 PMCID: PMC9463520 DOI: 10.1002/gch2.202100131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/28/2022] [Indexed: 05/26/2023]
Abstract
Liquid biopsy has remarkably facilitated clinical diagnosis and surveillance of cancer via employing a non-invasive way to detect cancer-derived components, such as circulating tumor DNA and circulating tumor cells from biological fluid samples. The cancer-derived exosomes, which are nano-sized vesicles secreted by cancer cells have been investigated in liquid biopsy as their important roles in intracellular communication and disease development have been revealed. Given the challenges posed by the complicated humoral microenvironment, which contains a variety of different cells and macromolecular substances in addition to the exosomes, it has attracted a large amount of attention to effectively isolate exosomes from collected samples. In this review, the authors aim to analyze classic strategies for separation of cancer-derived exosomes, giving an extensive discussion of advantages and limitations of these methods. Furthermore, the innovative multi-strategy methods to realize efficient isolation of cancer-derived exosomes in practical applications are also presented. Additionally, the possible development trends of exosome separation in to the future is discussed in this review.
Collapse
Affiliation(s)
- Yujiao Xie
- Cixi Institute of Biomedical EngineeringInternational Cooperation Base of Biomedical MaterialsTechnology and ApplicationChinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and EngineeringCASNingbo315201P. R. China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhou516000P. R. China
- Research Group for Fluids and Thermal EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
- Department of MechanicalMaterials and Manufacturing EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
| | - Xiawei Xu
- Cixi Institute of Biomedical EngineeringInternational Cooperation Base of Biomedical MaterialsTechnology and ApplicationChinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and EngineeringCASNingbo315201P. R. China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhou516000P. R. China
- Research Group for Fluids and Thermal EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
- Department of MechanicalMaterials and Manufacturing EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
| | - Jie Lin
- Cixi Institute of Biomedical EngineeringInternational Cooperation Base of Biomedical MaterialsTechnology and ApplicationChinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and EngineeringCASNingbo315201P. R. China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhou516000P. R. China
| | - Yanping Xu
- Cixi Institute of Biomedical EngineeringInternational Cooperation Base of Biomedical MaterialsTechnology and ApplicationChinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and EngineeringCASNingbo315201P. R. China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhou516000P. R. China
| | - Jing Wang
- Department of Electrical and Electronic EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
- Key Laboratory of More Electric Aircraft Technology of Zhejiang ProvinceUniversity of Nottingham Ningbo ChinaNingbo315100China
- Nottingham Ningbo China Beacons of Excellence Research and Innovation InstituteNingbo315040China
| | - Yong Ren
- Research Group for Fluids and Thermal EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
- Department of MechanicalMaterials and Manufacturing EngineeringUniversity of Nottingham Ningbo ChinaNingbo315100China
- Nottingham Ningbo China Beacons of Excellence Research and Innovation InstituteNingbo315040China
- Key Laboratory of Carbonaceous Wastes Processing and Process Intensification Research of Zhejiang ProvinceUniversity of Nottingham Ningbo ChinaNingbo315100China
| | - Aiguo Wu
- Cixi Institute of Biomedical EngineeringInternational Cooperation Base of Biomedical MaterialsTechnology and ApplicationChinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical MaterialsNingbo Institute of Materials Technology and EngineeringCASNingbo315201P. R. China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhou516000P. R. China
| |
Collapse
|
24
|
Habibian A, Soleimanjahi H, Hashemi SM, Babashah S. Characterization and Comparison of Mesenchymal Stem Cell-Derived Exosome Isolation Methods using Culture Supernatant. ARCHIVES OF RAZI INSTITUTE 2022; 77:1383-1388. [PMID: 36883158 PMCID: PMC9985774 DOI: 10.22092/ari.2021.356141.1790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/26/2021] [Indexed: 03/09/2023]
Abstract
Exosomes are extracellular endosomal nanoparticles, which are formed under complex processes during the formation of multivesicular bodies. They are also achieved from conditioned media of a variety of cell types, especially mesenchymal stem cells (MSCs). Exosomes can modulate intracellular physiological actions via signaling molecules on the surface or secretion of components to the extracellular spaces. Furthermore, they are potentially used as crucial agents for cell-free therapy; however, their isolation and characterization can be challenging. In the current study, two methods of exosome isolation have been characterized and compared using a culture media of adipose-derived mesenchymal stem cells, namely ultracentrifugation and a commercial kit; moreover, the efficiency of these two methods was highlighted in this study. Two different isolation methods of exosomes from MSCs were used to compare the efficiency of exosomes. For both isolation methods, transmission electron microscopy, dynamic light scattering (DLS), and bicinchoninic acid (BCA) assay have been performed. The electron microscopy and DLS indicated the presence of exosomes. Moreover, the kit and ultracentrifugation isolates contained approximately comparable amounts of protein measured by the BCA. Overall, the two isolation methods had similar performances. Although ultracentrifugation is used as a gold standard for exosome isolation, the commercial kit has some advantages and can be applied alternatively according to its cost-effectiveness and time-saving properties.
Collapse
Affiliation(s)
- A Habibian
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - H Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - S M Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - S Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
25
|
Mousavi SM, Amin Mahdian SM, Ebrahimi MS, Taghizadieh M, Vosough M, Sadri Nahand J, Hosseindoost S, Vousooghi N, Javar HA, Larijani B, Hadjighassem MR, Rahimian N, Hamblin MR, Mirzaei H. Microfluidics for detection of exosomes and microRNAs in cancer: State of the art. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:758-791. [PMID: 35664698 PMCID: PMC9130092 DOI: 10.1016/j.omtn.2022.04.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Exosomes are small extracellular vesicles with sizes ranging from 30-150 nanometers that contain proteins, lipids, mRNAs, microRNAs, and double-stranded DNA derived from the cells of origin. Exosomes can be taken up by target cells, acting as a means of cell-to-cell communication. The discovery of these vesicles in body fluids and their participation in cell communication has led to major breakthroughs in diagnosis, prognosis, and treatment of several conditions (e.g., cancer). However, conventional isolation and evaluation of exosomes and their microRNA content suffers from high cost, lengthy processes, difficult standardization, low purity, and poor yield. The emergence of microfluidics devices with increased efficiency in sieving, trapping, and immunological separation of small volumes could provide improved detection and monitoring of exosomes involved in cancer. Microfluidics techniques hold promise for advances in development of diagnostic and prognostic devices. This review covers ongoing research on microfluidics devices for detection of microRNAs and exosomes as biomarkers and their translation to point-of-care and clinical applications.
Collapse
Affiliation(s)
- Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeid Ebrahimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women’s Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Akbari Javar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
26
|
Gurunathan S, Kang MH, Song H, Kim NH, Kim JH. The role of extracellular vesicles in animal reproduction and diseases. J Anim Sci Biotechnol 2022; 13:62. [PMID: 35681164 PMCID: PMC9185900 DOI: 10.1186/s40104-022-00715-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/05/2022] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are nanosized membrane-enclosed compartments that serve as messengers in cell-to-cell communication, both in normal physiology and in pathological conditions. EVs can transfer functional proteins and genetic information to alter the phenotype and function of recipient cells, which undergo different changes that positively affect their structural and functional integrity. Biological fluids are enriched with several subpopulations of EVs, including exosomes, microvesicles (MVs), and apoptotic bodies carrying several cargoes, such as lipids, proteins, and nucleic acids. EVs associated with the reproductive system are actively involved in the regulation of different physiological events, including gamete maturation, fertilization, and embryo and fetal development. EVs can influence follicle development, oocyte maturation, embryo production, and endometrial-conceptus communication. EVs loaded with cargoes are used to diagnose various diseases, including pregnancy disorders; however, these are dependent on the type of cell of origin and pathological characteristics. EV-derived microRNAs (miRNAs) and proteins in the placenta regulate inflammatory responses and trophoblast invasion through intercellular delivery in the placental microenvironment. This review presents evidence regarding the types of extracellular vesicles, and general aspects of isolation, purification, and characterization of EVs, particularly from various types of embryos. Further, we discuss EVs as mediators and messengers in reproductive biology, the effects of EVs on placentation and pregnancy disorders, the role of EVs in animal reproduction, in the male reproductive system, and mother and embryo cross-communication. In addition, we emphasize the role of microRNAs in embryo implantation and the role of EVs in reproductive and therapeutic medicine. Finally, we discuss the future perspectives of EVs in reproductive biology.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Nam Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
27
|
Loch-Neckel G, Matos AT, Vaz AR, Brites D. Challenges in the Development of Drug Delivery Systems Based on Small Extracellular Vesicles for Therapy of Brain Diseases. Front Pharmacol 2022; 13:839790. [PMID: 35422699 PMCID: PMC9002061 DOI: 10.3389/fphar.2022.839790] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Small extracellular vesicles (sEVs) have ∼30–200 nm diameter size and may act as carriers of different cargoes, depending on the cell of origin or on the physiological/pathological condition. As endogenous nanovesicles, sEVs are important in intercellular communication and have many of the desirable features of an ideal drug delivery system. sEVs are naturally biocompatible, with superior targeting capability, safety profile, nanometric size, and can be loaded with both lipophilic and hydrophilic agents. Because of their biochemical and physical properties, sEVs are considered a promising strategy over other delivery vehicles in the central nervous system (CNS) since they freely cross the blood-brain barrier and they can be directed to specific nerve cells, potentiating a more precise targeting of their cargo. In addition, sEVs remain stable in the peripheral circulation, making them attractive nanocarrier systems to promote neuroregeneration. This review focuses on the recent progress in methods for manufacturing, isolating, and engineering sEVs that can be used as a therapeutic strategy to overcome neurodegeneration associated with pathologies of the CNS, with particular emphasis on Alzheimer’s, Parkinson’s, and amyotrophic lateral sclerosis diseases, as well as on brain tumors.
Collapse
Affiliation(s)
- Gecioni Loch-Neckel
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Teresa Matos
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Rita Vaz
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.,Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.,Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
28
|
Wongkaewkhiaw S, Wongrakpanich A, Krobthong S, Saengsawang W, Chairoungdua A, Boonmuen N. Induction of apoptosis in human colorectal cancer cells by nanovesicles from fingerroot (Boesenbergia rotunda (L.) Mansf.). PLoS One 2022; 17:e0266044. [PMID: 35377896 PMCID: PMC8979466 DOI: 10.1371/journal.pone.0266044] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 03/12/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is the leading cause of cancer-related deaths worldwide, warranting the urgent need for a new treatment option. Plant-derived nanovesicles containing bioactive compounds represent new therapeutic avenues due to their unique characteristics as natural nanocarriers for bioactive molecules with therapeutic effects. Recent evidence has revealed potential anticancer activity of bioactive compounds from Boesenbergia rotunda (L.) Mansf. (fingerroot). However, the effect and the underlying mechanisms of fingerroot-derived nanovesicles (FDNVs) against colorectal cancer are still unknown. We isolated the nanovesicles from fingerroot and demonstrated their anticancer activity against two colorectal cancer cell lines, HT-29 and HCT116. The IC50 values were 63.9 ± 2.4, 57.8 ± 4.1, 47.8 ± 7.6 μg/ml for HT-29 cells and 57.7 ± 6.6, 47.2 ± 5.2, 34 ± 2.9 μg/ml for HCT116 cells at 24, 48, and 72 h, respectively. Interestingly, FDNVs were not toxic to a normal colon epithelial cell line, CCD 841 CoN. FDNVs exhibited selective uptake by the colorectal cancer cell lines but not the normal colon epithelial cell line. Moreover, dose- and time-dependent FDNV-induced apoptosis was only observed in the colorectal cancer cell lines. In addition, reactive oxygen species levels were substantially increased in colorectal cancer cells, but total glutathione decreased after treatment with FDNVs. Our results show that FDNVs exhibited selective anticancer activity in colorectal cancer cell lines via the disruption of intracellular redox homeostasis and induction of apoptosis, suggesting the utility of FDNVs as a novel intervention for colorectal cancer patients.
Collapse
Affiliation(s)
| | | | - Sucheewin Krobthong
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Witchuda Saengsawang
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology, OPS, MHESI, Bangkok, Thailand
| | - Nittaya Boonmuen
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
N V Lakshmi Kavya A, Subramanian S, Ramakrishna S. Therapeutic applications of exosomes in various diseases: A review. BIOMATERIALS ADVANCES 2022; 134:112579. [PMID: 35525729 DOI: 10.1016/j.msec.2021.112579] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 06/14/2023]
Abstract
Exosomes (30-150 nm in diameter) a subset of extracellular vesicles, secreted by mostly all cells, have been gaining enormous recognition from the last decade. In recent times, several studies have included exosomes to design novel therapeutic applications along with their contribution to diagnostic evaluations and pathophysiological processes. Based on cell origin, they show diverse functions and characteristics. This article is classified into several sections that include exosomes biogenesis, isolation methods, and application as therapeutic tools, commercialized exosome products, clinical trials, benefits, and challenges faced in the progress of exosome-dependent therapeutics. This work aims to give a thorough review of the numerous studies where exosomes act as therapeutic tools in the treatment of various disorders including heart, kidney, liver, and lung illnesses. The clinical trials involving exosomes, their advantages, and hazards, and difficulties involved during storage and large-scale production, applications of nanotechnology in exosome research while applying for therapeutic applications, and future directions are summarized.
Collapse
Affiliation(s)
| | - Sundarrajan Subramanian
- Center for Nanofibers and Nanotechnology Lab, Mechanical Engineering, National University of Singapore, Blk E3 05-12, 2 Engineering Drive 3, Singapore 117581, Singapore.
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology Lab, Mechanical Engineering, National University of Singapore, Blk E3 05-12, 2 Engineering Drive 3, Singapore 117581, Singapore.
| |
Collapse
|
30
|
Inci F. Benchmarking a Microfluidic-Based Filtration for Isolating Biological Particles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:1897-1909. [PMID: 35041413 DOI: 10.1021/acs.langmuir.1c03119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Isolating particles from complex fluids is a crucial approach in multiple fields including biomedicine. In particular, biological matrices contain a myriad of distinct particles with different sizes and structures. Extracellular vesicles (EVs), for instance, are nanosized particles carrying vital information from donor to recipient cells, and they have garnered significant impact on disease diagnostics, drug delivery, and theranostics applications. Among all the EV types, exosome particles are one of the smallest entities, sizing from 30 to 100 nm. Separating such small substances from a complex media such as tissue culture and serum is still one of the most challenging steps in this field. Membrane filtration is one of the convenient approaches for these operations; yet clogging, low-recovery, and high fouling are still major obstacles. In this study, we design a two-filter-integrated microfluidic device focusing on dead-end and cross-flow processes at the same time, thereby minimizing any interfering factors on the recovery. The design of this platform is also numerically assessed to understand pressure-drop and flow rate effects over the procedure. As a model, we isolate exosome particles from human embryonic kidney cells cultured in different conditions, which also mimic complex fluids such as serum. Moreover, by altering the flow direction, we refresh the membranes for minimizing clogging issues and benchmark the platform performance for multitime use. By comprehensively analyzing the design and operation parameters of this platform, we address the aforementioned existing barriers in the recovery, clogging, and fouling factors, thereby achieving the use of a microfluidic device multiple times for bio-nanoparticle isolation without any notable issues.
Collapse
Affiliation(s)
- Fatih Inci
- UNAM - National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey
- Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| |
Collapse
|
31
|
Aljohani ASM, Abdellatif AAH, Rasheed Z, Abdulmonem WA. Gold-Nanoparticle-Conjugated Citrate Inhibits Tumor Necrosis Factor- α Expression via Suppression of Nuclear Factor Kappa B (NF- κB) Activation in Breast Cancer Cells. J Biomed Nanotechnol 2022; 18:581-588. [PMID: 35484745 DOI: 10.1166/jbn.2022.3266] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
One of the leading causes of death worldwide is cancer. Excessive production of tumor necrosis factor (TNF)-α is known to activate nuclear transcription factor (NF)-κB, which plays a lethal role in the onset of multiple disorders including cancer. In this study, we aimed to determine the therapeutic role of novel gold nanoparticles conjugated with citrate (AuNPs-CIT) on the elevated expression of TNF-α in breast cancer cells. AuNPs-CIT were synthesized by the citrate-reduction method and were characterized by UV-VIS spectroscopic analysis, zeta-potential analysis, and size analysis. The potential of these newly generated AuNPs-CIT particles was tested on phorbol 12-myristate 13-acetate (PMA)-stimulated cancer cells. Our data showed that the AuNPs-CIT were spherical, with a mean size of 21.3±0.65 nm and a stabilized zetapotential at -41.4±0.98 mV. These newly generated AuNPs-CIT nanoparticles inhibited PMA-induced activation and nuclear translocation of NF-κB p65 in MCF-7 cells. They also have the tendency to block TNF-α expression in stimulated cancer cells. In conclusion, AuNPs-CIT inhibits PMA-induced TNF-α mRNA and protein expression via deactivation of NF-κB signaling in breast cancer cells. These findings suggest that AuNPs-CIT might be useful in cancer treatment.
Collapse
Affiliation(s)
- Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Zafar Rasheed
- Department of Medical Biochemistry, College of Medicine, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
Perpetuo L, Ferreira R, Thongboonkerd V, Guedes S, Amado F, Vitorino R. Urinary exosomes: Diagnostic impact with a bioinformatic approach. Adv Clin Chem 2022; 111:69-99. [DOI: 10.1016/bs.acc.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
33
|
Onoshima D, Baba Y. Cancer diagnosis and analysis devices based on multimolecular crowding. Chem Commun (Camb) 2021; 57:13655-13661. [PMID: 34854439 DOI: 10.1039/d1cc05556a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The study of the multimolecular crowding around cancer cells has opened up the possibility of developing new devices for cancer diagnosis and analysis through the measurement of intercellular communication related to cell proliferation and invasive metastasis associated with cancer malignancy. In particular, cells and extracellular vesicles that flow into the bloodstream contain metabolites and secreted products of the cancer microenvironment. These are positioned as targets for the development of new devices for the understanding and application of multimolecular crowding around cancer cells. Examples include the separation analysis of cancer cells in blood for the next generation of less invasive testing techniques, and mapping analysis using Raman scattering to detect cancer cells without staining. Another example is the evaluation of the relationship between exosomes and cancer traits for the exploration of new anti-cancer drugs, and the commercialization of exosome separation devices for ultra-early cancer diagnosis. The development of nanobiodevice engineering, which applies multimolecular crowding to conventional nanobioscience, is expected to contribute to the diagnosis and analysis of various diseases in the future.
Collapse
Affiliation(s)
- Daisuke Onoshima
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan. .,Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
34
|
Tan YJ, Wong BYX, Vaidyanathan R, Sreejith S, Chia SY, Kandiah N, Ng ASL, Zeng L. Altered Cerebrospinal Fluid Exosomal microRNA Levels in Young-Onset Alzheimer's Disease and Frontotemporal Dementia. J Alzheimers Dis Rep 2021; 5:805-813. [PMID: 34870106 PMCID: PMC8609483 DOI: 10.3233/adr-210311] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2021] [Indexed: 12/11/2022] Open
Abstract
Background: micro-RNAs (miRNAs) are stable, small, non-coding RNAs enriched in exosomes. Their variation in levels according to different disease etiologies have made them a promising diagnostic biomarker for neurodegenerative diseases such as Alzheimer’s disease (AD). Altered expression of miR-320a, miR-328-3p, and miR-204-5p have been reported in AD and frontotemporal dementia (FTD). Objective: To determine their reliability, we aimed to examine the expression of three exosomal miRNAs isolated from cerebrospinal fluid (CSF) of patients with young-onset AD and FTD (< 65 years), correlating with core AD biomarkers and cognitive scores. Methods: Exosomes were first isolated from CSF samples of 48 subjects (8 controls, 28 AD, and 12 FTD), followed by RNA extraction and quantitative PCR to measure the expression of miR-320a, miR-328-3p, and miR-204-5p. Results: Expression of all three markers (miR-320a (p = 0.005), miR-328-3p (p = 0.049), and miR-204-5p (p = 0.036)) were significantly lower in AD versus controls. miR-320a was reduced in FTD versus controls (p = 0.049) and miR-328-3p was lower in AD versus FTD (p = 0.054). Notably, lower miR-328-3p levels could differentiate AD from FTD and controls with an AUC of 0.702, 95% CI: 0.534– 0.870, and showed significant correlation with lower CSF Aβ42 levels (r = 0.359, p = 0.029). Pathway enrichment analysis identified potential targets of miR-328-3p implicated in the AMPK signaling pathway linked to amyloid-β and tau metabolism in AD. Conclusion: Overall, we demonstrated miR-320a and miR-204-5p as reliable biomarkers for AD and FTD and report miR-328-3p as a novel AD biomarker.
Collapse
Affiliation(s)
- Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Benjamin Y X Wong
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | | | - Sivaramapanicker Sreejith
- Biomedical Institute for Global Health Research & Technology (BIGHEART), National University of Singapore, Singapore
| | - Sook Yoong Chia
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore
| | - Nagaendran Kandiah
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore.,Neuroscience and Behavioural Disorders Unit, Duke-NUS Medical School, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| | - Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore.,Neuroscience and Behavioural Disorders Unit, Duke-NUS Medical School, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore.,Neuroscience and Behavioural Disorders Unit, Duke-NUS Medical School, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| |
Collapse
|
35
|
Dosing extracellular vesicles. Adv Drug Deliv Rev 2021; 178:113961. [PMID: 34481030 DOI: 10.1016/j.addr.2021.113961] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/19/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are natural nanoparticles containing biologically active molecules. They are important mediators of intercellular communication and can be exploited therapeutically by various bioengineering approaches. To accurately determine the therapeutic potential of EVs in pre-clinical and clinical settings, dependable dosing strategies are of utmost importance. However, the field suffers from inconsistencies comprising all areas of EV production and characterisation. Therefore, a standardised and well-defined process in EV quantification, key to reliable therapeutic EV dosing, remains to be established. Here, we examined 64 pre-clinical studies for EV-based therapeutics with respect to their applied EV dosing strategies. We identified variations in effective dosing strategies irrespective of the applied EV purification method and cell source. Moreover, we found dose discrepancies depending on the disease model, where EV doses were selected without accounting for published EV pharmacokinetics or biodistribution patterns. We therefore propose to focus on qualitative aspects when dosing EV-based therapeutics, such as the potency of the therapeutic cargo entity. This will ensure batch-to-batch reliability and enhance reproducibility between applications. Furthermore, it will allow for the successful benchmarking of EV-based therapeutics compared to other nanoparticle drug delivery systems, such as viral vector-based or lipid-based nanoparticle approaches.
Collapse
|
36
|
Jang B, Chung H, Jung H, Song HK, Park E, Choi HS, Jung K, Choe H, Yang S, Oh ES. Extracellular Vesicles from Korean Codium fragile and Sargassum fusiforme Negatively Regulate Melanin Synthesis. Mol Cells 2021; 44:736-745. [PMID: 34650007 PMCID: PMC8560586 DOI: 10.14348/molcells.2021.2167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 07/27/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Although various marine ingredients have been exploited for the development of cosmetic products, no previous study has examined the potential of seaweed extracellular vesicles (EV) in such applications. Our results revealed that EV from Codium fragile and Sargassum fusiforme effectively decreased α-MSH-mediated melanin synthesis in MNT-1 human melanoma cells, associated with downregulation of MITF (microphthalmia-associated transcription factor), tyrosinase and TRP1 (tyrosinase-related proteins 1). The most effective inhibitory concentrations of EV were 250 μg/ml for S. fusiforme and 25 μg/ml for C. fragile, without affecting the viability of MNT-1 cells. Both EV reduced melanin synthesis in the epidermal basal layer of a three-dimensional model of human epidermis. Moreover, the application of the prototype cream containing C. fragile EV (final 5 μg/ml) yielded 1.31% improvement in skin brightness in a clinical trial. Together, these results suggest that EV from C. fragile and S. fusiforme reduce melanin synthesis and may be potential therapeutic and/or supplementary whitening agents.
Collapse
Affiliation(s)
- Bohee Jang
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Heesung Chung
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Hyejung Jung
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Hyun-Kuk Song
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Eunhye Park
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Hack Sun Choi
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea
| | | | - Han Choe
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | | | - Eok-Soo Oh
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
37
|
Scaled preparation of extracellular vesicles from conditioned media. Adv Drug Deliv Rev 2021; 177:113940. [PMID: 34419502 DOI: 10.1016/j.addr.2021.113940] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/13/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) especially of mesenchymal stem/stomal cells (MSCs) are increasingly considered as biotherapeutic agents for a variety of different diseases. For translating them effectively into the clinics, scalable production processes fulfilling good manufacturing practice (GMP) are needed. Like for other biotherapeutic agents, the manufacturing of EV products can be subdivided in the upstream and downstream processing and the subsequent quality control, each of them containing several unit operations. During upstream processing (USP), cells are isolated, stored (cell banking) and expanded; furthermore, EV-containing conditioned media are produced. During downstream processing (DSP), conditioned media (CM) are processed to obtain concentrated and purified EV products. CM are either stored until DSP or are directly processed. As first unit operation in DSP, clarification removes remaining cells, debris and other larger impurities. The key operations of each EV DSP is volume-reduction combined with purification of the concentrated EVs. Most of the EV preparation methods used in conventional research labs including differential centrifugation procedures are limited in their scalability. Consequently, it is a major challenge in the therapeutic EV field to identify appropriate EV concentration and purification methods allowing scale up. As EVs share several features with enveloped viruses, that are used for more than two decades in the clinics now, several principles can be adopted to EV manufacturing. Here, we introduce and discuss volume reducing and purification methods frequently used for viruses and analyze their value for the manufacturing of EV-based therapeutics.
Collapse
|
38
|
Fortunato D, Mladenović D, Criscuoli M, Loria F, Veiman KL, Zocco D, Koort K, Zarovni N. Opportunities and Pitfalls of Fluorescent Labeling Methodologies for Extracellular Vesicle Profiling on High-Resolution Single-Particle Platforms. Int J Mol Sci 2021; 22:10510. [PMID: 34638850 PMCID: PMC8508895 DOI: 10.3390/ijms221910510] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
The relevance of extracellular vesicles (EVs) has grown exponentially, together with innovative basic research branches that feed medical and bioengineering applications. Such attraction has been fostered by the biological roles of EVs, as they carry biomolecules from any cell type to trigger systemic paracrine signaling or to dispose metabolism products. To fulfill their roles, EVs are transported through circulating biofluids, which can be exploited for the administration of therapeutic nanostructures or collected to intercept relevant EV-contained biomarkers. Despite their potential, EVs are ubiquitous and considerably heterogeneous. Therefore, it is fundamental to profile and identify subpopulations of interest. In this study, we optimized EV-labeling protocols on two different high-resolution single-particle platforms, the NanoFCM NanoAnalyzer (nFCM) and Particle Metrix ZetaView Fluorescence Nanoparticle Tracking Analyzer (F-NTA). In addition to the information obtained by particles' scattered light, purified and non-purified EVs from different cell sources were fluorescently stained with combinations of specific dyes and antibodies to facilitate their identification and characterization. Despite the validity and compatibility of EV-labeling strategies, they should be optimized for each platform. Since EVs can be easily confounded with similar-sized nanoparticles, it is imperative to control instrument settings and the specificity of staining protocols in order to conduct a rigorous and informative analysis.
Collapse
Affiliation(s)
| | - Danilo Mladenović
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
- School of Natural Sciences and Health, Tallinn University, 10120 Tallinn, Estonia;
| | | | - Francesca Loria
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| | - Kadi-Liis Veiman
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| | - Davide Zocco
- Exosomics SpA, 53100 Siena, Italy; (D.F.); (M.C.); (D.Z.)
- Cell and Gene Therapy Research and Development, Lonza Inc., Rockville, MD 20850, USA
| | - Kairi Koort
- School of Natural Sciences and Health, Tallinn University, 10120 Tallinn, Estonia;
| | - Natasa Zarovni
- Exosomics SpA, 53100 Siena, Italy; (D.F.); (M.C.); (D.Z.)
- HansaBioMed Life Sciences Ltd., 12618 Tallinn, Estonia; (D.M.); (F.L.); (K.-L.V.)
| |
Collapse
|
39
|
Alzhrani GN, Alanazi ST, Alsharif SY, Albalawi AM, Alsharif AA, Abdel-Maksoud MS, Elsherbiny N. Exosomes: Isolation, characterization, and biomedical applications. Cell Biol Int 2021; 45:1807-1831. [PMID: 33913604 DOI: 10.1002/cbin.11620] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 12/13/2022]
Abstract
Exosomes are nano-sized bioactive vesicles of 30-150 nm in diameter. They are secreted by exocytosis of nearly all type of cells in to the extracellular fluid. Thereby, they can be found in many biological fluids. Exosomes regulate intracellular communication between cells via delivery of their cargo which include lipids, proteins, and nucleic acid. Many desirable features of exosomes made them promising candidates in several therapeutic applications. In this review, we discuss the use of exosomes as diagnostic tools and their possible biomedical applications. Additionally, current techniques used for isolation, purification, and characterization of exosomes from both biological fluids and in vitro cell cultures were discussed.
Collapse
Affiliation(s)
- Ghadi N Alzhrani
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Sarah T Alanazi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Sumayyah Y Alsharif
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Amani M Albalawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Anwar A Alsharif
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohamed S Abdel-Maksoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
40
|
Shirejini SZ, Inci F. The Yin and Yang of exosome isolation methods: conventional practice, microfluidics, and commercial kits. Biotechnol Adv 2021; 54:107814. [PMID: 34389465 DOI: 10.1016/j.biotechadv.2021.107814] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 08/08/2021] [Accepted: 08/08/2021] [Indexed: 12/18/2022]
Abstract
Exosomes are a subset of extracellular vesicles released from various cells, and they can be found in different bodily fluids. Exosomes are used as biomarkers to diagnose many diseases and to monitor therapy efficiency as they represent the status and origin of the cell, which they are released from. Considering that they co-exist in bodily fluids with other types of particles, their isolation still remains challenging since conventional separation methods are time-consuming, user-dependent, and result in low isolation yield. This review summarizes the conventional strategies and microfluidic-based methods for exosome isolation along with their strengths and limitations. Microfluidic devices emerge as a promising approach to overcome the limitations of the conventional methods due to their inherent characteristics, such as the need for minute sample volume and rapid operation, in order to isolate exosomes with a high yield and a high purity in a short amount of time, which make them unprecedented tools for molecular biology and clinical applications. This review elaborates on the existing microfluidic-based exosome isolation methods and denotes their benefits and drawbacks. Herein, we also introduce various commercially available platforms and kits for exosome isolation along with their working principles.
Collapse
Affiliation(s)
- Saeedreza Zeibi Shirejini
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey.
| |
Collapse
|
41
|
Pan D, Li Y, Yang F, Lv Z, Zhu S, Shao Y, Huang Y, Ning G, Feng S. Increasing toll-like receptor 2 on astrocytes induced by Schwann cell-derived exosomes promotes recovery by inhibiting CSPGs deposition after spinal cord injury. J Neuroinflammation 2021; 18:172. [PMID: 34372877 PMCID: PMC8353762 DOI: 10.1186/s12974-021-02215-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 07/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background Traumatic spinal cord injury (SCI) is a severely disabling disease that leads to loss of sensation, motor, and autonomic function. As exosomes have great potential in diagnosis, prognosis, and treatment of SCI because of their ability to easily cross the blood–brain barrier, the function of Schwann cell-derived exosomes (SCDEs) is still largely unknown. Methods A T10 spinal cord contusion was established in adult female mice. SCDEs were injected into the tail veins of mice three times a week for 4 weeks after the induction of SCI, and the control group was injected with PBS. High-resolution transmission electron microscope and western blot were used to characterize the SCDEs. Toll-like receptor 2 (TLR2) expression on astrocytes, chondroitin sulfate proteoglycans (CSPGs) deposition and neurological function recovery were measured in the spinal cord tissues of each group by immunofluorescence staining of TLR2, GFAP, CS56, 5-HT, and β-III-tublin, respectively. TLR2f/f mice were crossed to the GFAP-Cre strain to generate astrocyte specific TLR2 knockout mice (TLR2−/−). Finally, western blot analysis was used to determine the expression of signaling proteins and IKKβ inhibitor SC-514 was used to validate the involved signaling pathway. Results Here, we found that TLR2 increased significantly on astrocytes post-SCI. SCDEs treatment can promote functional recovery and induce the expression of TLR2 on astrocytes accompanied with decreased CSPGs deposition. The specific knockout of TLR2 on astrocytes abolished the decreasing CSPGs deposition and neurological functional recovery post-SCI. In addition, the signaling pathway of NF-κB/PI3K involved in the TLR2 activation was validated by western blot. Furthermore, IKKβ inhibitor SC-514 was also used to validate this signaling pathway. Conclusion Thus, our results uncovered that SCDEs can promote functional recovery of mice post-SCI by decreasing the CSPGs deposition via increasing the TLR2 expression on astrocytes through NF-κB/PI3K signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02215-x.
Collapse
Affiliation(s)
- Dayu Pan
- Department, of Orthopedics, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, People's Republic of China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yongjin Li
- Department, of Orthopedics, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, People's Republic of China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Fuhan Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
| | - Zenghui Lv
- Department, of Orthopedics, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, People's Republic of China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Shibo Zhu
- Department, of Orthopedics, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, People's Republic of China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yixin Shao
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ying Huang
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guangzhi Ning
- Department, of Orthopedics, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, People's Republic of China. .,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| | - Shiqing Feng
- Department, of Orthopedics, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, People's Republic of China. .,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| |
Collapse
|
42
|
Yasuno G, Koide H, Oku N, Asai T. Influence of Purification Process on the Function of Synthetic Polymer Nanoparticles. Chem Pharm Bull (Tokyo) 2021; 69:773-780. [PMID: 34334521 DOI: 10.1248/cpb.c21-00273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multifunctional synthetic polymers can bind to target molecules and are therefore widely investigated in diagnostics, drug delivery carriers, and separation carriers. Because these polymers are synthesized from nonbiological components, purification processes (e.g., chromatography, dialysis, extraction, and centrifugation) must be conducted after the synthesis. Although several purification methods are used for polymer purification, few reports have revealed the influence of purification process on the functions of polymer. In this study, we demonstrated that the characteristics, function, and stability of synthetic polymer depend on the purification process. N-Isopropylacrylamide-based polymer nanoparticles (NPs) and melittin (i.e., honey bee venom) were used as a model of synthetic polymer and target toxic peptide, respectively. Synthesized NPs were purified by dialysis in methanol, acetone precipitation, or centrifugation. NPs purified by dialysis in ultrapure water were used as control NPs. Then, NP size, surface charge, toxin neutralization effect, and stability were determined. NP size did not considerably change by purification with centrifugation; however, it decreased by purification using dialysis in methanol and acetone precipitation compared with that of control NPs. The ζ-potential of NPs changed after each purification process compared with that of control NPs. The melittin neutralization efficiency of NPs depended on the purification process; i.e., it decreased by acetone precipitation and increased by dialysis in methanol and centrifugation compared with that of control NPs. Of note, the addition of methanol and acetone decreased NP stability. These studies implied the importance of considering the effect of the purification method on synthetic polymer function.
Collapse
Affiliation(s)
- Go Yasuno
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences
| | - Hiroyuki Koide
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences
| | - Naoto Oku
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences.,Faculty of Pharma-Science, Teikyo University
| | - Tomohiro Asai
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences
| |
Collapse
|
43
|
Abstract
Extracellular Vesicles (EVs) gained significant interest within the last decade as a new source of biomarkers for the early detection of diseases and a promising tool for therapeutic applications. In this work, we present Extracellular Vesicles Quantitative Capillary Electrophoresis (EVqCE) to measure an average mass of RNA in EVs, determine EV concentrations and the degree of EV degradation after sample handling. We used EVqCE to analyze EVs isolated from conditioned media of three cancer cell lines. EVqCE employs capillary zone electrophoresis with laser-induced fluorescent detection to separate intact EVs from free nucleic acids. After lysis of EVs with a detergent, the encapsulated nucleic acids are released. Therefore, the initial concentration of intact EVs is calculated based on a nucleic acid peak gain. EVqCE works in a dynamic range of EV concentrations from 108 to 1010 particles/mL. The quantification process can be completed in less than one hour and requires minimum optimization. Furthermore, the average mass of RNA was found to be in the range of 200–400 ag per particle, noting that more aggressive cancer cells have less RNA in EVs (200 ag per particle) than non-aggressive cancer cells (350 ag per particle). EVqCE works well for the degradation analysis of EVs. Sonication for 10 min at 40 kHz caused 85% degradation of EVs, 10 freeze-thaw cycles (from −80 °C to 22 °C) produced 40%, 14-day storage at 4 °C made 32%, and vortexing for 5 min caused 5% degradation. Presently, EVqCE cannot separate and distinguish individual EV populations (exosomes, microvesicles, apoptotic bodies) from each other. Still, it is tolerant to the presence of non-EV particles, protein-lipid complexes, and protein aggregates.
Collapse
|
44
|
Raghav A, Tripathi P, Mishra BK, Jeong GB, Banday S, Gautam KA, Mateen QN, Singh P, Singh M, Singla A, Ahmad J. Mesenchymal Stromal Cell-Derived Tailored Exosomes Treat Bacteria-Associated Diabetes Foot Ulcers: A Customized Approach From Bench to Bed. Front Microbiol 2021; 12:712588. [PMID: 34385994 PMCID: PMC8354005 DOI: 10.3389/fmicb.2021.712588] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022] Open
Abstract
Exosomes are nano-vesicles of endosomal origin inherited with characteristics of drug delivery and cargo loading. Exosomes offer a diverse range of opportunities that can be exploited in the treatment of various diseases post-functionalization. This membrane engineering is recently being used in the management of bacteria-associated diabetic foot ulcers (DFUs). Diabetes mellitus (DM) is among the most crippling disease of society with a large share of its imposing economic burden. DM in a chronic state is associated with the development of micro- and macrovascular complications. DFU is among the diabetic microvascular complications with the consequent occurrence of diabetic peripheral neuropathy. Mesenchymal stromal cell (MSC)-derived exosomes post-tailoring hold promise to accelerate the diabetic wound repair in DFU associated with bacterial inhabitant. These exosomes promote the antibacterial properties with regenerative activity by loading bioactive molecules like growth factors, nucleic acids, and proteins, and non-bioactive substances like antibiotics. Functionalization of MSC-derived exosomes is mediated by various physical, chemical, and biological processes that effectively load the desired cargo into the exosomes for targeted delivery at specific bacterial DFUs and wound. The present study focused on the application of the cargo-loaded exosomes in the treatment of DFU and also emphasizes the different approaches for loading the desired cargo/drug inside exosomes. However, more studies and clinical trials are needed in the domain to explore this membrane engineering.
Collapse
Affiliation(s)
- Alok Raghav
- Multidisciplinary Research Unit, Department of Health Research, MoHFW, GSVM Medical College, Kanpur, India
| | | | | | - Goo-Bo Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Gachon University, Incheon, South Korea
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Kirti Amresh Gautam
- Multidisciplinary Research Unit, Department of Health Research, MoHFW, GSVM Medical College, Kanpur, India
| | - Qazi Noorul Mateen
- Department of Biochemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Prem Singh
- Department of Medicine, GSVM Medical College, Kanpur, India
| | - Manish Singh
- Department of Neurosurgery, GSVM Medical College, Kanpur, India
| | - Akhil Singla
- Department of Medicine, Maharishi Markandeshwar College and Hospital, Maharishi Markandeshwar University, Solan, India
| | - Jamal Ahmad
- Faculty of Medicine, Rajiv Gandhi Centre for Diabetes and Endocrinology, JN Medical College, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
45
|
Exosome/Liposome-like Nanoparticles: New Carriers for CRISPR Genome Editing in Plants. Int J Mol Sci 2021; 22:ijms22147456. [PMID: 34299081 PMCID: PMC8304373 DOI: 10.3390/ijms22147456] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Rapid developments in the field of plant genome editing using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) systems necessitate more detailed consideration of the delivery of the CRISPR system into plants. Successful and safe editing of plant genomes is partly based on efficient delivery of the CRISPR system. Along with the use of plasmids and viral vectors as cargo material for genome editing, non-viral vectors have also been considered for delivery purposes. These non-viral vectors can be made of a variety of materials, including inorganic nanoparticles, carbon nanotubes, liposomes, and protein- and peptide-based nanoparticles, as well as nanoscale polymeric materials. They have a decreased immune response, an advantage over viral vectors, and offer additional flexibility in their design, allowing them to be functionalized and targeted to specific sites in a biological system with low cytotoxicity. This review is dedicated to describing the delivery methods of CRISPR system into plants with emphasis on the use of non-viral vectors.
Collapse
|
46
|
Mastoridis S, Londoño MC, Kurt A, Kodela E, Crespo E, Mason J, Bestard O, Martínez-Llordella M, Sánchez-Fueyo A. Impact of donor extracellular vesicle release on recipient cell "cross-dressing" following clinical liver and kidney transplantation. Am J Transplant 2021; 21:2387-2398. [PMID: 32515541 DOI: 10.1111/ajt.16123] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/26/2020] [Accepted: 05/30/2020] [Indexed: 01/25/2023]
Abstract
In several murine models of transplantation, the "cross-dressing" of recipient antigen presenting cells (APCs) with intact donor major histocompatibility complex (MHC) derived from allograft-released small extracellular vesicles (sEVs) has been recently described as a key mechanism in eliciting and sustaining alloimmune responses. Investigation of these processes in clinical organ transplantation has, however, been hampered by the lack of sensitivity of conventional instruments and assays. We have employed advanced imaging flow cytometry (iFCM) to explore the kinetics of allograft sEV release and the extent to which donor sEVs might induce cross-dressing following liver and kidney transplantation. We report for the first time that recipient APC cross-dressing can be transiently detected in the circulation shortly after liver, but not kidney, transplantation in association with the release of HLA-bearing allograft-derived sEVs. In liver transplant recipients the majority of circulating cells exhibiting donor HLA are indeed cross-dressed cells and not passenger leukocytes. In keeping with experimental animal data, the downstream functional consequences of the transfer of circulating sEVs harvested from human transplant recipients varies depending on the type of transplant and time posttransplant. sEVs released shortly after liver, but not kidney, transplantation exhibit immunoinhibitory effects that could influence liver allograft immunogenicity.
Collapse
Affiliation(s)
- Sotiris Mastoridis
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - María-Carlota Londoño
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom.,Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Ada Kurt
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Elisavet Kodela
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Elena Crespo
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - John Mason
- Department of Physiology, Anatomy & Genetics, University of Oxford, United Kingdom
| | - Oriol Bestard
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Marc Martínez-Llordella
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Alberto Sánchez-Fueyo
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| |
Collapse
|
47
|
Singh K, Nalabotala R, Koo KM, Bose S, Nayak R, Shiddiky MJA. Separation of distinct exosome subpopulations: isolation and characterization approaches and their associated challenges. Analyst 2021; 146:3731-3749. [PMID: 33988193 DOI: 10.1039/d1an00024a] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exosomes are nano-sized extracellular vesicles that serve as a communications system between cells and have shown tremendous promise as liquid biopsy biomarkers in diagnostic, prognostic, and even therapeutic use in different human diseases. Due to the natural heterogeneity of exosomes, there is a need to separate exosomes into distinct biophysical and/or biochemical subpopulations to enable full interrogation of exosome biology and function prior to the possibility of clinical translation. Currently, there exists a multitude of different exosome isolation and characterization approaches which can, in limited capacity, separate exosomes based on biophysical and/or biochemical characteristics. While notable reviews in recent years have reviewed these approaches for bulk exosome sorting, we herein present a comprehensive overview of various conventional technologies and modern microfluidic and nanotechnological advancements towards isolation and characterization of exosome subpopulations. The benefits and limitations of these different technologies to improve their use for distinct exosome subpopulations in clinical practices are also discussed. Furthermore, an overview of the most commonly encountered technical and biological challenges for effective separation of exosome subpopulations is presented.
Collapse
Affiliation(s)
- Karishma Singh
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida 201301, UP, India.
| | - Ruchika Nalabotala
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida 201301, UP, India.
| | - Kevin M Koo
- The University of Queensland Centre for Clinical Research (UQCCR), Herston, QLD 4029, Australia.
| | - Sudeep Bose
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida 201301, UP, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida 201301, UP, India.
| | - Muhammad J A Shiddiky
- School of Environment and Natural Sciences and Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia.
| |
Collapse
|
48
|
Sakamoto S, Mallah D, Medeiros DJ, Dohi E, Imai T, Rose IVL, Matoba K, Zhu X, Kamiya A, Kano SI. Alterations in circulating extracellular vesicles underlie social stress-induced behaviors in mice. FEBS Open Bio 2021; 11:2678-2692. [PMID: 34043886 PMCID: PMC8487053 DOI: 10.1002/2211-5463.13204] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022] Open
Abstract
Chronic stress induces peripheral and intracerebral immune changes and inflammation, contributing to neuropathology and behavioral abnormalities relevant to psychiatric disorders such as depression. Although the pathological implication of many peripheral factors such as pro‐inflammatory cytokines, hormones, and macrophages has been demonstrated, the roles of circulating extracellular vesicles (EVs) for chronic stress mechanisms remain poorly investigated. Here, we report that chronic social defeat stress (CSDS)‐induced social avoidance phenotype, assessed by a previously untested three‐chamber social approach test, can be distinguished by multiple pro‐inflammatory cytokines and EV‐associated molecular signatures in the blood. We found that the expression patterns of miRNAs distinguished the CSDS‐susceptible mice from the CSDS‐resilient mice. Social avoidance behavior scores were also estimated with good accuracy by the expression patterns of multiple EV‐associated miRNAs. We also demonstrated that EVs enriched from the CSDS‐susceptible mouse sera upregulated the production of pro‐inflammatory cytokines in the LPS‐stimulated microglia‐like cell lines. Our results indicate the role of circulating EVs and associated miRNAs in CSDS susceptibility, which may be related to pro‐inflammatory mechanisms underlying stress‐induced neurobehavioral outcomes.
Collapse
Affiliation(s)
- Shinji Sakamoto
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dania Mallah
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Destynie J Medeiros
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Eisuke Dohi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Takashi Imai
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Indigo V L Rose
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ken Matoba
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shin-Ichi Kano
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| |
Collapse
|
49
|
Nakajima K, Nakatsuka R, Tsuji T, Doi K, Kawano S. Synchronized resistive-pulse analysis with flow visualization for single micro- and nanoscale objects driven by optical vortex in double orifice. Sci Rep 2021; 11:9323. [PMID: 33927219 PMCID: PMC8085213 DOI: 10.1038/s41598-021-87822-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/15/2021] [Indexed: 11/09/2022] Open
Abstract
Resistive-pulse analysis is a powerful tool for identifying micro- and nanoscale objects. For low-concentration specimens, the pulse responses are rare, and it is difficult to obtain a sufficient number of electrical waveforms to clearly characterize the targets and reduce noise. In this study, we conducted a periodic resistive-pulse analysis using an optical vortex and a double orifice, which repetitively senses a single micro- or nanoscale target particle with a diameter ranging from 700 nm to 2 \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\mu$$\end{document}μm. The periodic motion results in the accumulation of a sufficient number of waveforms within a short period. Acquired pulses show periodic ionic-current drops associated with the translocation events through each orifice. Furthermore, a transparent fluidic device allows us to synchronously average the waveforms by the microscopic observation of the translocation events and improve the signal-to-noise ratio. By this method, we succeed in distinguishing single particle diameters. Additionally, the results of measured signals and the simultaneous high-speed observations are used to quantitatively and systematically discuss the effect of the complex fluid flow in the orifices on the amplitude of the resistive pulse. The synchronized resistive-pulse analysis by the optical vortex with the flow visualization improves the pulse-acquisition rate for a single specific particle and accuracy of the analysis, refining the micro- and nanoscale object identification.
Collapse
Affiliation(s)
- Kichitaro Nakajima
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, 560-8531, Japan
| | - Ryoji Nakatsuka
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, 560-8531, Japan
| | - Tetsuro Tsuji
- Graduate School of Informatics, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kentaro Doi
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, 560-8531, Japan
| | - Satoyuki Kawano
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, 560-8531, Japan.
| |
Collapse
|
50
|
Yan H, Li Y, Cheng S, Zeng Y. Advances in Analytical Technologies for Extracellular Vesicles. Anal Chem 2021; 93:4739-4774. [PMID: 33635060 DOI: 10.1021/acs.analchem.1c00693] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- He Yan
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Yutao Li
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Shibo Cheng
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Yong Zeng
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States.,University of Florida Health Cancer Center, Gainesville, Florida 32610, United States
| |
Collapse
|