1
|
Praveena G, Jayachandran A, Manda Venkata S, Asthana A. From bench to bedside: The evolution of extracellular vesicle diagnostics through microfluidic and paper-based technologies. Colloids Surf B Biointerfaces 2025; 252:114675. [PMID: 40222114 DOI: 10.1016/j.colsurfb.2025.114675] [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: 12/19/2024] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 04/15/2025]
Abstract
"Extracellular vesicles (EVs) have emerged as key mediators of intercellular communication and valuable biomarkers for various diseases. However, traditional EV isolation and detection methods often struggle with efficiency, scalability, and purity, limiting their clinical utility. Recent advances in microfluidic and paper-based technologies offer innovative solutions that enhance EV isolation and detection by reducing sample volume, accelerating processing times, and integrating multiple analytical steps into compact platforms. These technologies hold significant promise for advancing point-of-care diagnostics, enabling rapid disease detection, personalized treatment monitoring, and better patient outcomes. For example, early detection of cancer biomarkers through EVs can facilitate timely intervention, potentially improving survival rates, while rapid infectious disease diagnostics can support prompt treatment. Despite their potential, challenges such as standardization, scalability, and regulatory hurdles remain. This review discusses recent advancements in microfluidic and paper-based EV diagnostic technologies, their comparative advantages over traditional methods, and their transformative potential in clinical practice."
Collapse
Affiliation(s)
- Ganji Praveena
- Urvogelbio Private Limited, AHERF, Film Nagar, Hyderabad, Telangana 500033, India
| | - Arjun Jayachandran
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad (NIPER - Hyderabad), Balanagar, Hyderabad, Telangana 500037, India
| | - Sasidhar Manda Venkata
- Urvogelbio Private Limited, AHERF, Film Nagar, Hyderabad, Telangana 500033, India; Apollo Hospitals Educational and Research Foundation (AHERF), Cell and Molecular Biology Research Lab, Hyderabad, India.
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad (NIPER - Hyderabad), Balanagar, Hyderabad, Telangana 500037, India.
| |
Collapse
|
2
|
Huang Q, Wang J, Ning H, Liu W, Han X. Exosome isolation based on polyethylene glycol (PEG): a review. Mol Cell Biochem 2025; 480:2847-2861. [PMID: 39702782 DOI: 10.1007/s11010-024-05191-x] [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: 09/06/2024] [Accepted: 12/07/2024] [Indexed: 12/21/2024]
Abstract
Exosome acts as an outstanding biomarker for ongoing studies, diagnosis and prognosis of multiples diseases. Therefore, the call for economically and efficiently isolating a large number of exosomes is an active area of investigation. However, to date, the challenges including complex isolated procedure, uneconomical equipment, low protein content and distinct loss in the particle number of exosomes etc. still encounter in exosome isolation. Polyethylene glycol (PEG)-induced exosome isolation increasingly attracts wide attention of scientists. PEG precipitation reveals higher performance in the yield of exosomes among multiple common isolation techniques. PEG-based precipitation is a temporarily low-purity, but inexpensive, time-save, labor-less, convenient and high-yield technique to gain exosomes with high biological activities. Hence, the PEG-based exosome isolation approach wins the endorsement of experimental workers. Herein, we summary the existing knowledge on procedures of PEG-based exosomes separation from different biospecimens, the binding process of PEG to exosomes, some notices, demerits, merits of PEG-based exosome isolation, and at last the advantages by combining PEG-precipitation to other techniques for exosome isolation, with a view to eliciting profound insights for investigators who recruit PEG for exosome separation, and advancing references for the standardization of PEG-based exosome isolation in future.
Collapse
Affiliation(s)
- Qionglian Huang
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jue Wang
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanjuan Ning
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Liu
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
3
|
Cho O. Plasma exosomal miR-150-3p, NMT2, and PRDM1 as predictive biomarkers of acute tumor response in patients with cervical cancer undergoing chemoradiotherapy. Am J Cancer Res 2025; 15:546-558. [PMID: 40084359 PMCID: PMC11897638 DOI: 10.62347/spqy5709] [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: 12/25/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Locally advanced cervical cancer (LACC) is primarily treated with weekly cisplatin-based concurrent chemoradiotherapy (CCRT); however, predicting acute tumor response remains challenging. This study aimed to identify plasma exosomal microRNAs (miRNAs) and messenger RNAs (mRNAs) that could predict rapid tumor regression in patients with LACC undergoing CCRT. Overall, 41 patients with stage IB-IVB cervical cancer were included. All patients received CCRT, and plasma exosomal RNA samples were collected before treatment and 2 weeks after radiation therapy (RT). Acute tumor response (AR) was defined as the regression rate of tumor volume (TV) (cm3) measured at the fourth week of treatment compared with the initial TV (iTV). The log2 fold change of miRNA and mRNA was calculated by comparing RNA read counts before and after the second week of CCRT for each patient. A correlation matrix identified RNAs associated with AR. The selected RNAs were validated through linear regression and Wilcoxon rank-sum tests. Leave-one-out cross-validation was performed in subgroups based on iTV. miR-150-3p, NMT2, and PRDM1 were identified as key predictors of AR, demonstrating significant associations with immune-mediated tumor responses. A decrease in post-RT levels of these RNAs was significantly associated with poor AR, particularly in patients with large iTVs. The predictive model combining miR-150-3p, NMT2, and PRDM1 showed strong correlation with AR (R2 = 0.831, P < 0.0001) in the test dataset and was validated in an independent cohort (R2 = 0.496, P = 0.006). Cross-validation indicated the robustness of these biomarkers in predicting AR across varying TVs. These findings highlight the potential of plasma exosomal miR-150-3p, NMT2, and PRDM1 are promising biomarkers for predicting AR in patients with LACC undergoing CCRT. These findings could facilitate personalized RT strategies and improve patient outcomes. Further multicenter studies are warranted to validate these biomarkers in larger, diverse cohorts.
Collapse
Affiliation(s)
- Oyeon Cho
- Gynecologic Cancer Center, Department of Radiation Oncology, Ajou University School of Medicine 164 World Cup-ro, Yeongtong-gu, Suwon 16499, Korea
| |
Collapse
|
4
|
Mohamed AH, Abaza T, Youssef YA, Rady M, Fahmy SA, Kamel R, Hamdi N, Efthimiado E, Braoudaki M, Youness RA. Extracellular vesicles: from intracellular trafficking molecules to fully fortified delivery vehicles for cancer therapeutics. NANOSCALE ADVANCES 2025; 7:934-962. [PMID: 39823046 PMCID: PMC11733735 DOI: 10.1039/d4na00393d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) are emerging as viable tools in cancer treatment due to their ability to carry a wide range of theranostic activities. This review summarizes different forms of EVs such as exosomes, microvesicles, apoptotic bodies, and oncosomes. It also sheds the light onto isolation methodologies, characterization techniques and therapeutic applications of all discussed EVs. Evidence indicates that EVs are particularly effective in delivering chemotherapeutic medications, and immunomodulatory agents. However, the advancement of EV-based therapies into clinical practice is hindered by challenges including EVs heterogeneity, cargo loading efficiency, and in vivo stability. Overall, EVs have the potential to change cancer therapeutic paradigms. Continued research and development activities are critical for improving EV-based medications and increasing their therapeutic impact.
Collapse
Affiliation(s)
- Adham H Mohamed
- Department of Chemistry, Faculty of Science, Cairo University 12613 Giza Egypt
| | - Tasneem Abaza
- Biotechnology and Biomolecular Chemistry Program, Faculty of Science, Cairo University 12613 Giza Egypt
- Université Paris-Saclay, Université d'Evry Val D'Essonne 91000 Évry-Courcouronnes Île-de-France France
| | - Yomna A Youssef
- Department of Physiology, Faculty of Physical Therapy, German International University (GIU) 11835 Cairo Egypt
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
- Faculty of Biotechnology, German International University New Administrative Capital 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre 12622 Cairo Egypt
| | - Nabila Hamdi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
| | - Eleni Efthimiado
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens Athens Greece
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical, and Biological Science, School of Life and Medical Sciences, University of Hertfordshire Hatfield AL10 9AB UK
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| |
Collapse
|
5
|
Lorite P, Domínguez JN, Palomeque T, Torres MI. Extracellular Vesicles: Advanced Tools for Disease Diagnosis, Monitoring, and Therapies. Int J Mol Sci 2024; 26:189. [PMID: 39796048 PMCID: PMC11720073 DOI: 10.3390/ijms26010189] [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: 11/29/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membrane-encapsulated vesicles released by cells into the extracellular space. They play a crucial role in intercellular communication by transporting bioactive molecules such as proteins, lipids, and nucleic acids. EVs can be detected in body fluids, including blood plasma, urine, saliva, amniotic fluid, breast milk, and pleural ascites. The complexity and diversity of EVs require a robust and standardized approach. By adhering to standardized protocols and guidelines, researchers can ensure the consistency, purity, and reproducibility of isolated EVs, facilitating their use in diagnostics, therapies, and research. Exosomes and microvesicles represent an exciting frontier in modern medicine, with significant potential to transform the diagnosis and treatment of various diseases with an important role in personalized medicine and precision therapy. The primary objective of this review is to provide an updated analysis of the significance of EVs by highlighting their mechanisms of action and exploring their applications in the diagnosis and treatment of various diseases. Additionally, the review addresses the existing limitations and future potential of EVs, offering practical recommendations to resolve current challenges and enhance their viability for clinical use. This comprehensive approach aims to bridge the gap between EV research and its practical application in healthcare.
Collapse
Affiliation(s)
| | | | | | - María Isabel Torres
- Department of Experimental Biology, Faculty of Health Sciences, University of Jaén, 23071 Jaén, Spain; (P.L.); (J.N.D.); (T.P.)
| |
Collapse
|
6
|
Lim M, Shin H, Jeong H, Kwon Y, Kim M, Lee J, Park J. Diagnosis of Prostate Cancer Metastasis via Extracellular Vesicles Isolated Using Two-Phase Interface as Membrane-Less Filter. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404846. [PMID: 39403999 DOI: 10.1002/smll.202404846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/11/2024] [Indexed: 12/28/2024]
Abstract
Extracellular vesicles (EVs), nano-sized particles secreted by cells, are increasingly recognized as promising biomarkers. However, the isolation and purification of EVs need improvement, impeding their practical application. Aqueous two-phase systems (ATPS) offer a method to separate EVs with high purity and yield compared to other techniques, yet the unclear isolation mechanism limits efficiency. To elucidate the separation process and enhance ATPS-based EV isolation, Kramers' theory and Fick's law are employed. The simulations and experiments reveal that the liquid-liquid interface in ATPS acts as a size cut-off filter for EVs, functioning without a membrane. It is discovered that rapid transport of particles to the interface is crucial for fast isolation, but this transport in separated phases relies solely on diffusion, which slows the process. To address this, a vortex is introduced to enhance particle movement through convection, significantly improving efficiency. This method achieves over 80% recovery of EVs from blood plasma and removes more than 90% of low-density lipoprotein, high-density lipoprotein, and albumin within an hour. Applying this ATPS-based membrane-less filter to plasma from prostate cancer patients, concentrations of markers on EVs are quantified. Using machine learning, metastatic and non-metastatic prostate cancer are distinguished with greater accuracy than the traditional PSA-based method.
Collapse
Affiliation(s)
- Minyeob Lim
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hyunwoo Shin
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hwapyeong Jeong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Yongmin Kwon
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Meeyoung Kim
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jiyoul Lee
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jaesung Park
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
- Global Research Institute for Exosome (GRIE), Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| |
Collapse
|
7
|
Shama KA, Greenberg ZF, Tammame C, He M, Taylor BL. Diseased Tendon Models Demonstrate Influence of Extracellular Matrix Alterations on Extracellular Vesicle Profile. Bioengineering (Basel) 2024; 11:1019. [PMID: 39451395 PMCID: PMC11505312 DOI: 10.3390/bioengineering11101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Tendons enable movement through their highly aligned extracellular matrix (ECM), predominantly composed of collagen I. Tendinopathies disrupt the structural integrity of tendons by causing fragmentation of collagen fibers, disorganization of fiber bundles, and an increase in glycosaminoglycans and microvasculature, thereby driving the apparent biomechanical and regenerative capacity in patients. Moreover, the complex cellular communication within the tendon microenvironment ultimately dictates the fate between healthy and diseased tendon, wherein extracellular vesicles (EVs) may facilitate the tendon's fate by transporting biomolecules within the tissue. In this study, we aimed to elucidate how the EV functionality is altered in the context of tendon microenvironments by using polycaprolactone (PCL) electrospun scaffolds mimicking healthy and pathological tendon matrices. Scaffolds were characterized for fiber alignment, mechanical properties, and cellular activity. EVs were isolated and analyzed for concentration, heterogeneity, and protein content. Our results show that our mimicked healthy tendon led to an increase in EV secretion and baseline metabolic activity over the mimicked diseased tendon, where reduced EV secretion and a significant increase in metabolic activity over 5 days were observed. These findings suggest that scaffold mechanics may influence EV functionality, offering insights into tendon homeostasis. Future research should further investigate how EV cargo affects the tendon's microenvironment.
Collapse
Affiliation(s)
- Kariman A. Shama
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
| | | | - Chadine Tammame
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
| | - Mei He
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32603, USA;
| | - Brittany L. Taylor
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
| |
Collapse
|
8
|
Kim MY, Jo MS, Choi SG, Moon HW, Park J, Lee JY. Repeated Injections of Mesenchymal Stem Cell-Derived Exosomes Ameliorate Erectile Dysfunction in a Cavernous Nerve Injury Rat Model. World J Mens Health 2024; 42:787-796. [PMID: 38311373 PMCID: PMC11439812 DOI: 10.5534/wjmh.230218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/21/2023] [Indexed: 02/10/2024] Open
Abstract
PURPOSE To evaluate the therapeutic effect of repeated injections of mesenchymal stem cell (MSC)-derived exosomes on the erectile dysfunction (ED) of bilateral cavernous nerve injury (BCNI) rat model and to identify potential target genes of these injections. MATERIALS AND METHODS MSC-derived exosomes were isolated using an aqueous two-phase system. Rats were randomly assigned into four groups: Normal, BCNI, exosome once, and exosome-repeat groups. After four weeks, we measured the intracavernosal pressure (ICP)/mean arterial pressure (MAP) ratio to evaluate erectile function and examined cavernous nerve tissues for histological and molecular analyses. RNA sequencing in penile tissues was used to determine differentially expressed genes and was verified by quantitative polymerase chain reaction. Human umbilical vein endothelial cells (HUVECs) were used for in vitro studies to analyze biological roles. RESULTS The ICP/MAP ratios in the exosome-once and exosome-repeat groups were significantly increased compared to those in the BCNI group. Interestingly, the ICP/MAP ratio showed a greater increase in the exosome-repeat group, which also showed significantly increased smooth muscle/collagen ratio, α-smooth muscle actin and neuronal nitric oxide synthase expression, and cyclic guanosine monophosphate level compared to the BCNI and exosome-once groups. Three genes were significantly differentially expressed in the exosome group, among which Ras homolog family member B promoted cell proliferation and angiogenesis of HUVECs. CONCLUSIONS Repeated injections of MSC-derived exosomes can be effective in the treatment of rat models with ED induced by cavernous nerve injury.
Collapse
Affiliation(s)
- Mee Young Kim
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Prostate Institute, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Soo Jo
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sun Geum Choi
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Prostate Institute, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyong Woo Moon
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Prostate Institute, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jaesung Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Korea
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Prostate Institute, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
9
|
Sankaranarayanan J, Lee SC, Kim HK, Kang JY, Kuppa SS, Seon JK. Cinnamaldehyde-Treated Bone Marrow Mesenchymal-Stem-Cell-Derived Exosomes via Aqueous Two-Phase System Attenuate IL-1β-Induced Inflammation and Catabolism via Modulation of Proinflammatory Signaling Pathways. Int J Mol Sci 2024; 25:7263. [PMID: 39000370 PMCID: PMC11242605 DOI: 10.3390/ijms25137263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/21/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disorder that is distinguished by inflammation and chronic cartilage damage. Interleukin-1β (IL-1β) is a proinflammatory cytokine that plays an important role in the catabolic processes that underlie the pathogenesis of OA. In this study, we investigate the therapeutic efficacy of exosomes derived from untreated bone-marrow-derived mesenchymal stem cells (BMMSC-Exo) and those treated with cinnamaldehyde (BMMSC-CA-Exo) for preventing the in vitro catabolic effects of IL-1β on chondrocytes. We stimulated chondrocytes with IL-1β to mimic the inflammatory microenvironment of OA. We then treated these chondrocytes with BMMSC-Exo and BMMSC-CA-Exo isolated via an aqueous two-phase system and evaluated their effects on the key cellular processes using molecular techniques. Our findings revealed that treatment with BMMSC-Exo reduces the catabolic effects of IL-1β on chondrocytes and alleviates inflammation. However, further studies directly comparing treatments with BMMSC-Exo and BMMSC-CA-Exo are needed to determine if CA preconditioning can provide additional anti-inflammatory benefits to the exosomes beyond those of CA preconditioning or treatment with regular BMMSC-Exo. Through a comprehensive molecular analysis, we elucidated the regulatory mechanisms underlying this protective effect. We found a significant downregulation of proinflammatory signaling pathways in exosome-infected chondrocytes, suggesting the potential modulation of the NF-κB and MAPK signaling cascades. Furthermore, our study identified the molecular cargo of BMMSC-Exo and BMMSC-CA-Exo, determining the key molecules, such as anti-inflammatory cytokines and cartilage-associated factors, that may contribute to their acquisition of chondroprotective properties. In summary, BMMSC-Exo and BMMSC-CA-Exo exhibit the potential as therapeutic agents for OA by antagonizing the in vitro catabolic effects of IL-1β on chondrocytes. The regulation of the proinflammatory signaling pathways and bioactive molecules delivered by the exosomes suggests a multifaceted mechanism of action. These findings highlight the need for further investigation into exosome-based therapies for OA and joint-related diseases.
Collapse
Affiliation(s)
- Jaishree Sankaranarayanan
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.S.); (S.S.K.)
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Seok Cheol Lee
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Hyung Keun Kim
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Ju Yeon Kang
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Sree Samanvitha Kuppa
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.S.); (S.S.K.)
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Jong Keun Seon
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.S.); (S.S.K.)
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| |
Collapse
|
10
|
Sharma V, Joo JU, Mottafegh A, Kim DP. Continuous and autonomous-flow separation of laccase enzyme utilizing functionalized aqueous two-phase system with computer vision control. BIORESOURCE TECHNOLOGY 2024; 403:130888. [PMID: 38788804 DOI: 10.1016/j.biortech.2024.130888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Downstream processing of biomolecules, particularly therapeutic proteins and enzymes, presents a formidable challenge due to intricate unit operations and high costs. This study introduces a novel cysteine (cys) functionalized aqueous two-phase system (ATPS) utilizing polyethylene glycol (PEG) and potassium phosphate, referred as PEG-K3PO4/cys, for selective extraction of laccase from complex protein mixtures. A 3D-baffle micro-mixer and phase separator was meticulously designed and equipped with computer vision controller, to enable precise mixing and continuous phase separation under automated-flow. Microfluidic-assisted ATPS exhibits substantial increase in partition coefficient (Kflow = 16.3) and extraction efficiency (EEflow = 88 %) for laccase compared to conventional batch process. Integrated and continuous-flow process efficiently partitioned laccase, even in low concentrations and complex crude extracts. Circular dichroism spectra of laccase confirm structural stability of enzyme throughout the purification process. Eventually, continuous-flow microfluidic bioseparation is highly useful for seamless downstream processing of target biopharmaceuticals in integrated and autonomous manner.
Collapse
Affiliation(s)
- Vikas Sharma
- Center for Intelligent Microprocess of Pharmaceutical Synthesis, Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jeong-Un Joo
- Center for Intelligent Microprocess of Pharmaceutical Synthesis, Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Amirreza Mottafegh
- Center for Intelligent Microprocess of Pharmaceutical Synthesis, Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Dong-Pyo Kim
- Center for Intelligent Microprocess of Pharmaceutical Synthesis, Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| |
Collapse
|
11
|
Liu C, Yazdani N, Moran CS, Salomon C, Seneviratne CJ, Ivanovski S, Han P. Unveiling clinical applications of bacterial extracellular vesicles as natural nanomaterials in disease diagnosis and therapeutics. Acta Biomater 2024; 180:18-45. [PMID: 38641182 DOI: 10.1016/j.actbio.2024.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/03/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are naturally occurring bioactive membrane-bound nanoparticles released by both gram-negative and gram-positive bacterial species, exhibiting a multifaceted role in mediating host-microbe interactions across various physiological conditions. Increasing evidence supports BEVs as essential mediators of cell-to-cell communicaiton, influencing bacterial pathogenicity, disease mechanisms, and modulating the host immune response. However, the extent to which these BEV-mediated actions can be leveraged to predict disease onset, guide treatment strategies, and determine clinical outcomes remains uncertain, particularly in terms of their clinical translation potentials. This review briefly describes BEV biogenesis and their internalisation by recipient cells and summarises methods for isolation and characterization, essential for understanding their composition and cargo. Further, it discusses the potential of biofluid-associated BEVs as biomarkers for various diseases, spanning both cancer and non-cancerous conditions. Following this, we outline the ongoing human clinical trials of using BEVs for vaccine development. In addition to disease diagnostics, this review explores the emerging research of using natural or engineered BEVs as smart nanomaterials for applications in anti-cancer therapy and bone regeneration. This discussion extends to key factors for unlocking the clinical potential of BEVs, such as standardization of BEV isolation and characterisation, as well as other hurdles in translating these findings to the clinical setting. We propose that addressing these hurdles through collaborative research efforts and well-designed clinical trials holds the key to fully harnessing the clinical potential of BEVs. As this field advances, this review suggests that BEV-based nanomedicine has the potential to revolutionize disease management, paving the way for innovative diagnosis, therapeutics, and personalized medicine approaches. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) from both host cells and bacteria serve as multifunctional biomaterials and are emerging in the fields of biomedicine, bioengineering, and biomaterials. However, the majority of current studies focus on host-derived EVs, leaving a gap in comprehensive research on bacteria-derived EVs (BEVs). Although BEVs offer an attractive option as nanomaterials for drug delivery systems, their unique nanostructure and easy-to-modify functions make them a potential method for disease diagnosis and treatment as well as vaccine development. Our work among the pioneering studies investigating the potential of BEVs as natural nanobiomaterials plays a crucial role in both understanding the development of diseases and therapeutic interventions.
Collapse
Affiliation(s)
- Chun Liu
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Negar Yazdani
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Corey S Moran
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029 Australia
| | - Chaminda Jayampath Seneviratne
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Sašo Ivanovski
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia.
| | - Pingping Han
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia.
| |
Collapse
|
12
|
Naquin TD, Canning AJ, Gu Y, Chen J, Naquin CM, Xia J, Lu B, Yang S, Koroza A, Lin K, Wang HN, Jeck WR, Lee LP, Vo-Dinh T, Huang TJ. Acoustic separation and concentration of exosomes for nucleotide detection: ASCENDx. SCIENCE ADVANCES 2024; 10:eadm8597. [PMID: 38457504 PMCID: PMC10923504 DOI: 10.1126/sciadv.adm8597] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/05/2024] [Indexed: 03/10/2024]
Abstract
Efficient isolation and analysis of exosomal biomarkers hold transformative potential in biomedical applications. However, current methods are prone to contamination and require costly consumables, expensive equipment, and skilled personnel. Here, we introduce an innovative spaceship-like disc that allows Acoustic Separation and Concentration of Exosomes and Nucleotide Detection: ASCENDx. We created ASCENDx to use acoustically driven disc rotation on a spinning droplet to generate swift separation and concentration of exosomes from patient plasma samples. Integrated plasmonic nanostars on the ASCENDx disc enable label-free detection of enriched exosomes via surface-enhanced Raman scattering. Direct detection of circulating exosomal microRNA biomarkers from patient plasma samples by the ASCENDx platform facilitated a diagnostic assay for colorectal cancer with 95.8% sensitivity and 100% specificity. ASCENDx overcomes existing limitations in exosome-based molecular diagnostics and holds a powerful position for future biomedical research, precision medicine, and point-of-care medical diagnostics.
Collapse
Affiliation(s)
- Ty D. Naquin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Aidan J. Canning
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Yuyang Gu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Jianing Chen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Chloe M. Naquin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Jianping Xia
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Brandon Lu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Shujie Yang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Aleksandra Koroza
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Katherine Lin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Hsin-Neng Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - William R. Jeck
- Department of Pathology, Duke University Medical Center, Durham, NC 27708, USA
| | - Luke P. Lee
- Harvard Medical School, Harvard University; Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Bioengineering and Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| |
Collapse
|
13
|
Roszkowski S. Therapeutic potential of mesenchymal stem cell-derived exosomes for regenerative medicine applications. Clin Exp Med 2024; 24:46. [PMID: 38427086 PMCID: PMC10907468 DOI: 10.1007/s10238-023-01282-z] [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: 10/26/2023] [Accepted: 11/08/2023] [Indexed: 03/02/2024]
Abstract
Mesenchymal stem cell-derived exosomes have emerged as a promising cell-free therapy for tissue engineering. Compared to intact stem cells, exosomes have advantages like low immunogenicity and ability to carry regenerative cargo. This review examined the potential of exosomes to treat defects in skin, bone and cartilage. In preclinical models, exosomes improved wound healing, stimulated bone regeneration, and enabled cartilage repair by transferring proteins, mRNAs and microRNAs. Their effects were elicited by modulating inflammation, angiogenesis, cell proliferation and matrix synthesis. Exosomes represent a promising cell-free therapy for tissue engineering. However, challenges remain regarding scalable isolation, elucidating mechanisms, and translating this approach to human trials. Understanding these challenges will enable the successful clinical translation of exosomes for regenerative medicine applications.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Debowa St. 3, 85-626, Bydgoszcz, Poland.
| |
Collapse
|
14
|
Hormozi A, Hasanzadeh S, Ebrahimi F, Daei N, Hajimortezayi Z, Mehdizadeh A, Zamani M. Treatment with Exosomes Derived from Mesenchymal Stem Cells: A New Window of Healing Science in Regenerative Medicine. Curr Stem Cell Res Ther 2024; 19:879-893. [PMID: 37622719 DOI: 10.2174/1574888x18666230824165014] [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: 03/11/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 08/26/2023]
Abstract
Many studies have been conducted on the potential applications of mesenchymal stem cells (MSCs) over recent years due to their growing importance in regenerative medicine. Exosomes are considered cargos capable of transporting proteins, peptides, lipids, mRNAs, and growth factors. MSCsderived exosomes are also involved in the prevention or treatment of a variety of diseases, including cardiovascular diseases, neurological diseases, skin disorders, lung diseases, osteoarthritis, damaged tissue repair, and other diseases. This review attempted to summarize the importance of employing MSCs in regenerative medicine by gathering and evaluating information from current literature. The role of MSCs and the potential applications of MSCs-derived exosomes have also been discussed.
Collapse
Affiliation(s)
- Arezoo Hormozi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Sajedeh Hasanzadeh
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Faezeh Ebrahimi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Narges Daei
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zahra Hajimortezayi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Zamani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Simon L, Lapinte V, Morille M. Exploring the role of polymers to overcome ongoing challenges in the field of extracellular vesicles. J Extracell Vesicles 2023; 12:e12386. [PMID: 38050832 PMCID: PMC10696644 DOI: 10.1002/jev2.12386] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring nanoparticles released from all eucaryotic and procaryotic cells. While their role was formerly largely underestimated, EVs are now clearly established as key mediators of intercellular communication. Therefore, these vesicles constitute an attractive topic of study for both basic and applied research with great potential, for example, as a new class of biomarkers, as cell-free therapeutics or as drug delivery systems. However, the complexity and biological origin of EVs sometimes complicate their identification and therapeutic use. Thus, this rapidly expanding research field requires new methods and tools for the production, enrichment, detection, and therapeutic application of EVs. In this review, we have sought to explain how polymer materials actively contributed to overcome some of the limitations associated to EVs. Indeed, thanks to their infinite diversity of composition and properties, polymers can act through a variety of strategies and at different stages of EVs development. Overall, we would like to emphasize the importance of multidisciplinary research involving polymers to address persistent limitations in the field of EVs.
Collapse
Affiliation(s)
| | | | - Marie Morille
- ICGM, Univ Montpellier, CNRS, ENSCMMontpellierFrance
- Institut universitaire de France (IUF)ParisFrance
| |
Collapse
|
16
|
Arif S, Moulin VJ. Extracellular vesicles on the move: Traversing the complex matrix of tissues. Eur J Cell Biol 2023; 102:151372. [PMID: 37972445 DOI: 10.1016/j.ejcb.2023.151372] [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: 06/22/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Extracellular vesicles are small particles involved in intercellular signaling. They are produced by virtually all cell types, transport biological molecules, and are released into the extracellular space. Studies on extracellular vesicles have become more numerous in recent years, leading to promising research on their potential impact on health and disease. Despite significant progress in understanding the bioactivity of extracellular vesicles, most in vitro and in vivo studies overlook their transport through the extracellular matrix in tissues. The interaction or free diffusion of extracellular vesicles in their environment can provide valuable insights into their efficacy and function. Therefore, understanding the factors that influence the transport of extracellular vesicles in the extracellular matrix is essential for the development of new therapeutic approaches that involve the use of these extracellular vesicles. This review discusses the importance of the interaction between extracellular vesicles and the extracellular matrix and the different factors that influence their diffusion. In addition, we evaluate their role in tissue homeostasis, pathophysiology, and potential clinical applications. Understanding the complex interaction between extracellular vesicles and the extracellular matrix is critical in order to develop effective strategies to target specific cells and tissues in a wide range of clinical applications.
Collapse
Affiliation(s)
- Syrine Arif
- Faculté de Médecine, Université Laval, Quebec City, QC G1V 0A6, Canada; Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada; Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Véronique J Moulin
- Faculté de Médecine, Université Laval, Quebec City, QC G1V 0A6, Canada; Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada; Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
17
|
Zhang C, Zhou X, Wang D, Hao L, Zeng Z, Su L. Hydrogel-Loaded Exosomes: A Promising Therapeutic Strategy for Musculoskeletal Disorders. J Clin Pharm Ther 2023; 2023:1-36. [DOI: 10.1155/2023/1105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Clinical treatment strategies for musculoskeletal disorders have been a hot research topic. Accumulating evidence suggests that hydrogels loaded with MSC-derived EVs show great potential in improving musculoskeletal injuries. The ideal hydrogels should be capable of promoting the development of new tissues and simulating the characteristics of target tissues, with the properties matching the cell-matrix constituents of autologous tissues. Although there have been numerous reports of hydrogels loaded with MSC-derived EVs for the repair of musculoskeletal injuries, such as intervertebral disc injury, tendinopathy, bone fractures, and cartilage injuries, there are still many hurdles to overcome before the clinical application of modified hydrogels. In this review, we focus on the advantages of the isolation technique of EVs in combination with different types of hydrogels. In this context, the efficacy of hydrogels loaded with MSC-derived EVs in different musculoskeletal injuries is discussed in detail to provide a reference for the future application of hydrogels loaded with MSC-derived EVs in the clinical treatment of musculoskeletal injuries.
Collapse
Affiliation(s)
- Chunyu Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Li Hao
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Lei Su
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| |
Collapse
|
18
|
Kanao E, Ishida K, Mizuta R, Li Y, Imami K, Tanigawa T, Sasaki Y, Akiyoshi K, Adachi J, Otsuka K, Ishihama Y, Kubo T. Rapid and Highly Efficient Purification of Extracellular Vesicles Enabled by a TiO 2 Hybridized Spongy-like Polymer. Anal Chem 2023; 95:14502-14510. [PMID: 37703188 DOI: 10.1021/acs.analchem.3c03411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
We developed a novel purification medium of extracellular vesicles (EVs) by constructing a spongy-like monolithic polymer kneaded with TiO2 microparticles (TiO2-hybridized spongy monolith, TiO2-SPM). TiO2-SPM was applied in a solid-phase extraction format and enabled simple, rapid, and highly efficient purification of EVs. This is due to the high permeability caused by the continuous large flow-through pores of the monolithic skeleton (median pore size; 5.21 μm) and the specific interaction of embedded TiO2 with phospholipids of the lipid bilayers. Our method also excels in efficiency and comprehensiveness, collecting small EVs (SEVs) from the same volume of a cell culture medium 130.7 times more than typical ultracentrifugation and 4.3 times more than affinity purification targeting surface phosphatidylserine by magnetic beads. The purification method was completed within 1 h with simple operations and was directly applied to serum SEVs. Finally, we demonstrated flexibility toward the shape and size of our method by depleting EVs from fetal bovine serum (FBS), which is a necessary process to prevent contamination of culture cell-derived EVs with exogenous FBS-derived EVs. Our method will eliminate the tedious and difficult purification processes of EVs, providing a universal purification platform for EV-based drug discovery and pathological diagnosis.
Collapse
Affiliation(s)
- Eisuke Kanao
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 567-0085, Osaka, Japan
| | - Koki Ishida
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Ryosuke Mizuta
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yuka Li
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Koshi Imami
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Tetsuya Tanigawa
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Jun Adachi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 567-0085, Osaka, Japan
| | - Koji Otsuka
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 567-0085, Osaka, Japan
| | - Takuya Kubo
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| |
Collapse
|
19
|
El-Tanani M, Nsairat H, Matalka II, Aljabali AAA, Mishra V, Mishra Y, Naikoo GA, Chava SR, Charbe NB, Tambuwala MM. Impact of exosome therapy on pancreatic cancer and its progression. Med Oncol 2023; 40:225. [PMID: 37405480 PMCID: PMC10322774 DOI: 10.1007/s12032-023-02101-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/19/2023] [Indexed: 07/06/2023]
Abstract
Pancreatic cancer, one of the most aggressive tumors, has a dismal prognosis because of the low rates of early identification, fast progression, difficulties following surgery, and the ineffectiveness of current oncologic therapies. There are no imaging techniques or biomarkers that can accurately identify, categorize, or predict the biological behavior of this tumor. Exosomes are extracellular vesicles that play a crucial rule in the progression, metastasis, and chemoresistance of pancreatic cancer. They have been verified to be potential biomarkers for pancreatic cancer management. Studying the role of exosomes in pancreatic cancer is substantial. Exosomes are secreted by most eukaryotic cells and participated in intercellular communication. The components of exosomes, including proteins, DNA, mRNA, microRNA, long non-coding RNA, circular RNA, etc., play a crucial role in regulating tumor growth, metastasis, and angiogenesis in the process of cancer development, and can be used as a prognostic marker and/or grading basis for tumor patients. Hereby, in this concise review, we intend to summarize exosomes components and isolation, exosome secretion, function, importance of exosomes in the progression of pancreatic cancer and exosomal miRNAs as possible pancreatic cancer biomarkers. Finally, the application potential of exosomes in the treatment of pancreatic cancer, which provides theoretical supports for using exosomes to serve precise tumor treatment in the clinic, will be discussed.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan.
- Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, West Yorkshire, UK.
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid, 21163, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Gowhar A Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, PC 211, Salalah, Oman
| | | | - Nitin B Charbe
- Department of Pharmaceutics, College of Pharmacy, Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| |
Collapse
|
20
|
Fernandes LB, D'Souza JS, Prasad TSK, Ghag SB. Isolation and characterization of extracellular vesicles from Fusarium oxysporum f. sp. cubense, a banana wilt pathogen. Biochim Biophys Acta Gen Subj 2023; 1867:130382. [PMID: 37207907 DOI: 10.1016/j.bbagen.2023.130382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
Fusarium wilt of banana is a destructive widespread disease caused by Fusarium oxysporum f. sp. cubense (Foc) that ravaged banana plantations globally, incurring huge economic losses. Current knowledge demonstrates the involvement of several transcription factors, effector proteins, and small RNAs in the Foc-banana interaction. However, the precise mode of communication at the interface remains elusive. Cutting-edge research has emphasized the significance of extracellular vesicles (EVs) in trafficking the virulent factors modulating the host physiology and defence system. EVs are ubiquitous inter- and intra-cellular communicators across kingdoms. This study focuses on the isolation and characterization of Foc EVs from methods that make use of sodium acetate, polyethylene glycol, ethyl acetate, and high-speed centrifugation. Isolated EVs were microscopically visualized using Nile red staining. Further, the EVs were characterized using transmission electron microscopy, which revealed the presence of spherical, double-membrane, vesicular structures ranging in size from 50 to 200 nm (diameter). The size was also determined using the principle based on Dynamic Light Scattering. The Foc EVs contained proteins that were separated using SDS-PAGE and ranged between 10 and 315 kDa. Mass spectrometry analysis revealed the presence of EV-specific marker proteins, toxic peptides, and effectors. The Foc EVs were found to be cytotoxic, whose toxicity increased with EVs isolated from the co-culture preparation. Taken together, a better understanding of Foc EVs and their cargo will aid in deciphering the molecular crosstalk between banana and Foc.
Collapse
Affiliation(s)
- Lizelle B Fernandes
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai campus, Kalina, Santacruz (East), Mumbai 400098, India
| | - Jacinta S D'Souza
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai campus, Kalina, Santacruz (East), Mumbai 400098, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore 575018, India
| | - Siddhesh B Ghag
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai campus, Kalina, Santacruz (East), Mumbai 400098, India.
| |
Collapse
|
21
|
Kieda J, Appak-Baskoy S, Jeyhani M, Navi M, Chan KWY, Tsai SSH. Microfluidically-generated Encapsulated Spheroids (μ-GELS): An All-Aqueous Droplet Microfluidics Platform for Multicellular Spheroids Generation. ACS Biomater Sci Eng 2023; 9:1043-1052. [PMID: 36626575 DOI: 10.1021/acsbiomaterials.2c00963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Spheroids are three-dimensional clusters of cells that serve as in vitro tumor models to recapitulate in vivo morphology. A limitation of many existing on-chip platforms for spheroid formation is the use of cytotoxic organic solvents as the continuous phase in droplet generation processes. All-aqueous methods do not contain cytotoxic organic solvents but have so far been unable to achieve complete hydrogel gelation on chip. Here, we describe an enhanced droplet microfluidic platform that achieves on-chip gelation of all-aqueous hydrogel multicellular spheroids (MCSs). Specifically, we generate dextran-alginate droplets containing MCF-7 breast cancer cells, surrounded by polyethylene glycol, at a flow-focusing junction. Droplets then travel to a second flow-focusing junction where they interact with calcium chloride and gel on chip to form hydrogel MCSs. On-chip gelation of the MCSs is possible here because of an embedded capillary at the second junction that delays the droplet gelation, which prevents channel clogging problems that would otherwise exist. In drug-free experiments, we demonstrate that MCSs remain viable for 6 days. We also confirm the applicability of this system for cancer drug testing by observing that dose-dependent cell death is achievable using doxorubicin.
Collapse
Affiliation(s)
- Jennifer Kieda
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Sila Appak-Baskoy
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Chemistry and Biology, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| | - Morteza Jeyhani
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| | - Maryam Navi
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Katherine W Y Chan
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Scott S H Tsai
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| |
Collapse
|
22
|
Zhao L, Yu L, Wang X, He J, Zhu X, Zhang R, Yang A. Mechanisms of function and clinical potential of exosomes in esophageal squamous cell carcinoma. Cancer Lett 2023; 553:215993. [PMID: 36328162 DOI: 10.1016/j.canlet.2022.215993] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/05/2022] [Accepted: 10/27/2022] [Indexed: 11/20/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) remains one of the most lethal and widespread malignancies in China. Exosomes, a subset of tiny extracellular vesicles manufactured by all cells and present in all body fluids, contribute to intercellular communication and have become a focus of the search for new therapeutic strategies for cancer. A number of global analyses of exosome-mediated functions and regulatory mechanism in malignant diseases have recently been reported. There is extensive evidence that exosomes can be used as diagnostic and prognostic markers for cancer. However, our understanding of their clinical value and mechanisms of action in ESCC is still limited and has not been systematically reviewed. Here, we review current research specifically focused on the functions and mechanisms of action of ESCC tumor-derived exosomes and non-ESCC-derived exosomes in ESCC progression and describe opportunities and challenges in the clinical translation of exosomes.
Collapse
Affiliation(s)
- Lijun Zhao
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Lili Yu
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiangpeng Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jangtao He
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiaofei Zhu
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Rui Zhang
- The State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Angang Yang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China; The State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
Nicoletti A, Negri M, Paratore M, Vitale F, Ainora ME, Nista EC, Gasbarrini A, Zocco MA, Zileri Dal Verme L. Diagnostic and Prognostic Role of Extracellular Vesicles in Pancreatic Cancer: Current Evidence and Future Perspectives. Int J Mol Sci 2023; 24:885. [PMID: 36614326 PMCID: PMC9821035 DOI: 10.3390/ijms24010885] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive tumors, with a dismal prognosis due to poor detection rates at early stages, rapid progression, post-surgical complications, and limited effectiveness of conventional oncologic therapies. There are no consistently reliable biomarkers or imaging modalities to accurately diagnose, classify, and predict the biological behavior of this tumor. Therefore, it is imperative to develop new and improved strategies to detect pancreatic lesions in the early stages of cancerization with greater sensitivity and specificity. Extracellular vesicles, including exosome and microvesicles, are membrane-coated cellular products that are released in the outer environment. All cells produce extracellular vesicles; however, this process is enhanced by inflammation and tumorigenesis. Based on accumulating evidence, extracellular vesicles play a crucial role in pancreatic cancer progression and chemoresistance. Moreover, they may represent potential biomarkers and promising therapy targets. The aim of the present review is to review the current evidence on the role of extracellular vesicles in pancreatic cancer.
Collapse
|
24
|
Williams S, Fernandez-Rhodes M, Law A, Peacock B, Lewis MP, Davies OG. Comparison of extracellular vesicle isolation processes for therapeutic applications. J Tissue Eng 2023; 14:20417314231174609. [PMID: 37251735 PMCID: PMC10214056 DOI: 10.1177/20417314231174609] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/23/2023] [Indexed: 05/31/2023] Open
Abstract
While extracellular vesicles (EVs) continue to gain interest for therapeutic applications, their clinical translation is limited by a lack of optimal isolation methods. We sought to determine how universally applied isolation methods impact EV purity and yield. EVs were isolated by ultracentrifugation (UC), polyethylene glycol precipitation, Total Exosome Isolation Reagent, an aqueous two-phase system with and without repeat washes or size exclusion chromatography (SEC). EV-like particles could be detected for all isolation methods but varied in their purity and relative expression of surface markers (Alix, Annexin A2, CD9, CD63 and CD81). Assessments of sample purity were dependent on the specificity of characterisation method applied, with total particle counts and particle to protein (PtP) ratios often not aligning with quantitative measures of tetraspanin surface markers obtained using high-resolution nano-flow cytometry. While SEC resulted in the isolation of fewer particles with a relatively low PtP ratio (1.12 × 107 ± 1.43 × 106 vs highest recorded; ATPS/R 2.01 × 108 ± 1.15 × 109, p ⩽ 0.05), EVs isolated using this method displayed a comparatively high level of tetraspanin positivity (e.g. ExoELISA CD63⁺ particles; 1.36 × 1011 ± 1.18 × 1010 vs ATPS/R 2.58 × 1010 ± 1.92 × 109, p ⩽ 0.001). Results originating from an accompanying survey designed to evaluate pragmatic considerations surrounding method implementation (e.g. scalability and cost) identified that SEC and UC were favoured for overall efficiency. However, reservations were highlighted in the scalability of these methods, which could potentially hinder downstream therapeutic applications. In conclusion, variations in sample purity and yield were evident between isolation methods, while standard non-specific assessments of sample purity did not align with advanced quantitative high-resolution analysis of EV surface markers. Reproducible and specific assessments of EV purity will be critical for informing therapeutic studies.
Collapse
Affiliation(s)
- Soraya Williams
- School of Sport, Exercise and Health
Sciences, Loughborough University, Loughborough, UK
| | - Maria Fernandez-Rhodes
- School of Sport, Exercise and Health
Sciences, Loughborough University, Loughborough, UK
| | - Alice Law
- NanoFCM Co., Ltd, Medicity, Nottingham,
UK
| | | | - Mark P. Lewis
- School of Sport, Exercise and Health
Sciences, Loughborough University, Loughborough, UK
| | - Owen G. Davies
- School of Sport, Exercise and Health
Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
25
|
Daradmare S, Lee CS. Recent progress in the synthesis of all-aqueous two-phase droplets using microfluidic approaches. Colloids Surf B Biointerfaces 2022; 219:112795. [PMID: 36049253 DOI: 10.1016/j.colsurfb.2022.112795] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/10/2022] [Accepted: 08/21/2022] [Indexed: 12/21/2022]
Abstract
An aqueous two-phase system (ATPS) is a system with liquid-liquid phase separation and shows great potential for the extraction, separation, purification, and enrichment of proteins, membranes, viruses, enzymes, nucleic acids, and other biomolecules because of its simplicity, biocompatibility, and wide applicability [1-4]. The clear aqueous-aqueous interface of ATPSs is highly advantageous for their implementation, therefore making ATPSs a green alternative approach to replace conventional emulsion systems, such as water-in-oil droplets. All aqueous emulsions (water-in-water, w-in-w) hold great promise in the biomedical field as glucose sensors [5] and promising carriers for the encapsulation and release of various biomolecules and nonbiomolecules [6-10]. However, the ultralow interfacial tension between the two phases is a hurdle in generating w-in-w emulsion droplets. In the past, bulk emulsification and electrospray techniques were employed for the generation of w-in-w emulsion droplets and the fabrication of microparticles and microcapsules in the later stage. Bulk emulsification is a simple and low-cost technique; however, it generates polydisperse w-in-w emulsion droplets. Another technique, electrospray, involves easy experimental setups that can generate monodisperse but nonspherical w-in-w emulsion droplets. In comparison, microfluidic platforms provide monodisperse w-in-w emulsion droplets with spherical shapes, deal with the small volumes of solutions and short reaction times and achieve portability and versatility in their design through rapid prototyping. Owing to several advantages, microfluidic approaches have recently been introduced. To date, several different strategies have been explored to generate w-in-w emulsions and multiple w-in-w emulsions and to fabricate microparticles and microcapsules using conventional microfluidic devices. Although a few review articles on ATPSs emulsions have been published in the past, to date, few reviews have exclusively focused on the evolution of microfluidic-based ATPS droplets. The present review begins with a brief discussion of the history of ATPSs and their fundamentals, which is followed by an account chronicling the integration of microfluidic devices with ATPSs to generate w-in-w emulsion droplets. Furthermore, the stabilization strategies of w-in-w emulsion droplets and microfluidic fabrication of microparticles and microcapsules for modern applications, such as biomolecule encapsulation and spheroid construction, are discussed in detail in this review. We believe that the present review will provide useful information to not only new entrants in the microfluidic community wanting to appreciate the findings of the field but also existing researchers wanting to keep themselves updated on progress in the field.
Collapse
Affiliation(s)
- Sneha Daradmare
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Chang-Soo Lee
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
26
|
Yu J, Xie X, Ma Y, Yang Y, Wang C, Xia G, Ding X, Liu X. Effects and potential mechanism of Ca 2+/calmodulin‑dependent protein kinase II pathway inhibitor KN93 on the development of ovarian follicle. Int J Mol Med 2022; 50:121. [PMID: 35929517 PMCID: PMC9387563 DOI: 10.3892/ijmm.2022.5177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/11/2022] [Indexed: 11/09/2022] Open
Abstract
Adequate regulation of the speed of follicular development has been reported to prolong the reproductive life of the ovary. The aim of the present study was to assess the potential effects and mechanism of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) pathway on the development of ovarian follicle. In the present study, the expression of CaMKII was measured in the ovary of mice at different developmental stages by immunofluorescence, confirming that CaMKII has a role in follicular development. Subsequently, the 17.5 days post-coitus (dpc) embryonic ovaries were collected and cultured with KN93 for 4 days in vitro. It was revealed that KN93 inhibited the development of follicles, where it reduced the expression levels of oocyte and granulosa cell markers DEAD-box helicase 4 (DDX4) and forkhead box L2 (FOXL2). These results suggested that KN93 could delay follicular development. Proteomics technology was then used to find that 262 proteins of KN93 treated 17.5 dpc embryonic ovaries were significantly altered after in vitro culture. Bioinformatics analysis was used to analyze these altered proteins. In total, four important Kyoto Encyclopedia of Genes and Genome pathways, namely steroid biosynthesis, p53 signaling pathway and retinol metabolism and metabolic pathways, were particularly enriched. Further analysis revealed that the upregulated proteins NADP-dependent steroid dehydrogenase-like (Nsdhl), lanosterol synthase (Lss), farnesyl-diphosphate farnesyltransferase 1 (Fdft1), cytochrome P450 family 51 family A member 1 (Cyp51a1), hydroxymethylglutaryl-CoA synthase 1 (Hmgcs1), fatty acid synthase (Fasn) and dimethylallyltranstransferase (Fdps) were directly interacting with each other in the four enriched pathways. In summary, the potential mechanism of KN93 in slowing down follicular development most likely lies in its inhibitory effects on CaMKII, which upregulated the expression of Nsdhl, Lss, Fdft1, Cyp51a1, Hmgcs1, Fasn and Fdps. This downregulated the expression of oocyte and granulosa cell markers DDX4 and FOXL2 in the follicles, thereby delaying follicular development. Overall, these results provide novel insight into the potential mechanism by which KN93 and CaMKII can delay follicular development.
Collapse
Affiliation(s)
- Jianjie Yu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R. China
| | - Xianguo Xie
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R. China
| | - Yabo Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R. China
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R. China
| | - Chao Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R.China
| | - Guoliang Xia
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R. China
| | - Xiangbin Ding
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, P.R. China
| | - Xinfeng Liu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, College of Life Science, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region 750021, P.R. China
| |
Collapse
|
27
|
Kim H, Yi J, Yu J, Park J, Jang SK. A Simple and Effective Method to Concentrate Hepatitis C Virus: Aqueous Two-Phase System Allows Highly Efficient Enrichment of Enveloped Viruses. Viruses 2022; 14:v14091987. [PMID: 36146792 PMCID: PMC9503063 DOI: 10.3390/v14091987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
To investigate the proliferation cycle of a virus, virus-host interaction, and pathogenesis of a virus, virion particles must be concentrated from the media of virus cell culture or the sera of virus-infected patients. Ultracentrifugation of the culture media is a standard method for concentrating virion particles. However, this method is time-consuming and requires special equipment (ultracentrifuge). Moreover, a large number of infectious viruses are lost during enrichment. We developed a new method of hepatitis C virus (HCV) concentration to overcome the issues associated with traditional methods of virus concentration. We used an aqueous two-phase system (ATPS) to concentrate the virus. HCV, which causes various liver diseases, such as liver fibrosis, cirrhosis, and hepatocellular carcinoma, was used as a model virus to test the efficacy and reliability of the ATPS. The efficiency of HCV concentration by the ATPS was approximately three times higher than that by ultracentrifugation. Moreover, the infectivity of the concentrated HCV, which is a labile virus, remained the same after concentration of the virus by the ATPS. Considering the simplicity and effectiveness of the ATPS, it is the method of choice for concentrating viruses.
Collapse
Affiliation(s)
- Heesun Kim
- Molecular Virology Laboratory, POSTECH Biotech Center, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Johan Yi
- Nanoparticle and Vesicle Laboratory, Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Jinbae Yu
- Molecular Virology Laboratory, Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Jaesung Park
- Nanoparticle and Vesicle Laboratory, Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Korea
- Nanoparticle and Vesicle Laboratory, School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Korea
- Correspondence: (J.P.); (S.K.J.)
| | - Sung Key Jang
- Molecular Virology Laboratory, POSTECH Biotech Center, Pohang University of Science and Technology, Pohang 37673, Korea
- Molecular Virology Laboratory, Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
- Correspondence: (J.P.); (S.K.J.)
| |
Collapse
|
28
|
Elkhoury K, Chen M, Koçak P, Enciso-Martínez E, Bassous NJ, Lee MC, Byambaa B, Rezaei Z, Li Y, Urbina M, Gurian M, Sobahi N, Hussain MA, Sanchez-Gonzalez L, Leijten J, Hassan S, Arab-Tehrany E, Ward JE, Shin SR. Hybrid extracellular vesicles-liposome incorporated advanced bioink to deliver microRNA. Biofabrication 2022; 14:10.1088/1758-5090/ac8621. [PMID: 35917808 PMCID: PMC9594995 DOI: 10.1088/1758-5090/ac8621] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/02/2022] [Indexed: 11/12/2022]
Abstract
In additive manufacturing, bioink formulations govern strategies to engineer 3D living tissues that mimic the complex architectures and functions of native tissues for successful tissue regeneration. Conventional 3D-printed tissues are limited in their ability to alter the fate of laden cells. Specifically, the efficient delivery of gene expression regulators (i.e. microRNAs (miRNAs)) to cells in bioprinted tissues has remained largely elusive. In this study, we explored the inclusion of extracellular vesicles (EVs), naturally occurring nanovesicles (NVs), into bioinks to resolve this challenge. EVs show excellent biocompatibility, rapid endocytosis, and low immunogenicity, which lead to the efficient delivery of miRNAs without measurable cytotoxicity. EVs were fused with liposomes to prolong and control their release by altering their physical interaction with the bioink. Hybrid EVs-liposome (hEL) NVs were embedded in gelatin-based hydrogels to create bioinks that could efficiently encapsulate and deliver miRNAs at the target site in a controlled and sustained manner. The regulation of cells' gene expression in a 3D bioprinted matrix was achieved using the hELs-laden bioink as a precursor for excellent shape fidelity and high cell viability constructs. Novel regulatory factors-loaded bioinks will expedite the translation of new bioprinting applications in the tissue engineering field.
Collapse
Affiliation(s)
- Kamil Elkhoury
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- LIBio, Université de Lorraine, F-54000 Nancy, France
- These authors contributed equally to this work
| | - Mo Chen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- Department of Gynecology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai 200011, China
- These authors contributed equally to this work
| | - Polen Koçak
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- Department of Biomedical Engineering, Faculty of Engineering, İstinye University, 34396 Sariyer/Istanbul, Trukey
| | - Eduardo Enciso-Martínez
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Nicole Joy Bassous
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | | | - Zahra Rezaei
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Yang Li
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Mariely Urbina
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Melvin Gurian
- Department of Developmental BioEngineering, University of Twente, Enschede, Overijssel 7522 NB, The Netherlands
| | - Nebras Sobahi
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Mohammad Asif Hussain
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | | | - Jeroen Leijten
- Department of Developmental BioEngineering, University of Twente, Enschede, Overijssel 7522 NB, The Netherlands
| | - Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- Department of Biology, Khalifa University, 127788, Abu Dhabi, UAE Division of Genetics
| | | | - Jennifer Ellis Ward
- Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, 02115 USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| |
Collapse
|
29
|
Subudhi PD, Bihari C, Sarin SK, Baweja S. Emerging Role of Edible Exosomes-Like Nanoparticles (ELNs) as Hepatoprotective Agents. Nanotheranostics 2022; 6:365-375. [PMID: 35795340 PMCID: PMC9254361 DOI: 10.7150/ntno.70999] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/24/2022] [Indexed: 11/07/2022] Open
Abstract
Liver diseases are responsible for over 2 million deaths each year and the number is rapidly increasing. There is a strong link between edibles, gut microbiota, liver fat and the liver damage. There are very limited therapeutic options for treatment specifically for Alcoholic liver disease (ALD) and Non-Alcoholic liver disease (NAFLD). Recently, identified Edible Exosomes-like nanoparticles (ELNs) are plant derived membrane bound particles, released by microvesicular bodies for cellular communication and regulate immune responses against many pathogens. Many studies have identified their role as hepatoprotective agent as they carry bioactive material as cargoes which are transferred to recipient cells and affect various biological functions in liver. They are also known to carry specific miRNA, which increases the copy number of beneficial bacteria and the production of lactic acid metabolites in gut and hence restrains from liver injury through portal vein. Few in-vitro studies also have been reported about the anti-inflammatory, anti-oxidant and detoxification properties of ELNs which again protects the liver. The properties such as small size, biocompatibility, stability, low toxicity and non-immunogenicity make ELNs as a better therapeutic option. But, till now, studies on the effect of ELNs as therapeutics are still at its infancy yet promising. Here we discuss about the isolation, characterization, their role in maintaining the gut microbiome and liver homeostasis. Also, we give an outline about the latest advances in ELNs modifications, its biological effects, limitations and we propose the future prospective of ELNs as therapeutics.
Collapse
Affiliation(s)
- P Debishree Subudhi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, Delhi, India
| | - Chhagan Bihari
- Department of Pathology, Institute of Liver and Biliary Sciences, Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, Delhi, India
| | - Sukriti Baweja
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, Delhi, India
| |
Collapse
|
30
|
Kim G, Chen X, Yang Y. Pathogenic Extracellular Vesicle (EV) Signaling in Amyotrophic Lateral Sclerosis (ALS). Neurotherapeutics 2022; 19:1119-1132. [PMID: 35426061 PMCID: PMC9587178 DOI: 10.1007/s13311-022-01232-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs), once considered a pathway for cells to remove waste, have now emerged as an important mechanism for intercellular communication. EVs are particularly appealing in understanding the central nervous system (CNS) communication, given that there are very diverse cell types in the CNS and constant communications among various cells to respond to the frequently changing environment. While they are heterogeneous and new vesicles are continuously to be discovered, EVs are primarily classified as plasma membrane-derived microvesicles (MVs) and endosome-derived exosomes. Secretion of EVs has been shown from all CNS cell types in vitro and intercellular EV signaling has been implicated in neural development, axon integrity, neuron to glia communication, and propagation of protein aggregates formed by disease pathogenic proteins. However, significant hurdles remain to be tackled in understanding their physiological and pathological roles as well as how they can be developed as biomarkers or new therapeutics. Here we provide our summary on the known cell biology of EVs and discuss opportunities and challenges in understanding EV biology in the CNS and particularly their involvement in ALS pathogenesis.
Collapse
Affiliation(s)
- Gloria Kim
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Xuan Chen
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA.
- Graduate School of Biomedical Sciences, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA.
| |
Collapse
|
31
|
Yakubovich EI, Polischouk AG, Evtushenko VI. Principles and Problems of Exosome Isolation from Biological Fluids. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2022; 16:115-126. [PMID: 35730027 PMCID: PMC9202659 DOI: 10.1134/s1990747822030096] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/03/2022]
Abstract
Exosomes, the subclass of small membrane extracellular vesicles, have great diagnostic and therapeutic potential, but the lack of standardized methods for their efficient isolation and analysis limits the introduction of exosomal technologies into clinical practice. This review discusses the problems associated with the isolation of exosomes from biological fluids, as well as the principles of traditional and alternative methods of isolation. The aim of the presented review is to illustrate the variety of approaches based on the physical and biochemical properties of exosomes that can be used for exosome isolation. The advantages and disadvantages of different methods are discussed.
Collapse
Affiliation(s)
- E. I. Yakubovich
- Granov Russian Research Center for Radiology and Surgical Technologies, Ministry of Health of the Russian Federation, 197758 St. Petersburg, Russia
| | - A. G. Polischouk
- Granov Russian Research Center for Radiology and Surgical Technologies, Ministry of Health of the Russian Federation, 197758 St. Petersburg, Russia
| | - V. I. Evtushenko
- Granov Russian Research Center for Radiology and Surgical Technologies, Ministry of Health of the Russian Federation, 197758 St. Petersburg, Russia
| |
Collapse
|
32
|
Janouskova O, Herma R, Semeradtova A, Poustka D, Liegertova M, Malinska HA, Maly J. Conventional and Nonconventional Sources of Exosomes-Isolation Methods and Influence on Their Downstream Biomedical Application. Front Mol Biosci 2022; 9:846650. [PMID: 35586196 PMCID: PMC9110031 DOI: 10.3389/fmolb.2022.846650] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Despite extensive study of extracellular vesicles (EVs), specifically exosomes (EXs) as biomarkers, important modulators of physiological or pathological processes, or therapeutic agents, relatively little is known about nonconventional sources of EXs, such as invertebrate or plant EXs, and their uses. Likewise, there is no clear information on the overview of storage conditions and currently used isolation methods, including new ones, such as microfluidics, which fundamentally affect the characterization of EXs and their other biomedical applications. The purpose of this review is to briefly summarize conventional and nonconventional sources of EXs, storage conditions and typical isolation methods, widely used kits and new "smart" technologies with emphasis on the influence of isolation techniques on EX content, protein detection, RNA, mRNA and others. At the same time, attention is paid to a brief overview of the direction of biomedical application of EXs, especially in diagnostics, therapy, senescence and aging and, with regard to the current situation, in issues related to Covid-19.
Collapse
Affiliation(s)
- Olga Janouskova
- Centre of Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista University in Ústí Nad Labem, Ústí Nad Labem, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
33
|
Brown SV, Dewitt S, Clayton A, Waddington RJ. Identifying the Efficacy of Extracellular Vesicles in Osteogenic Differentiation: An EV-Lution in Regenerative Medicine. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.849724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have long been the focus for regenerative medicine and the restoration of damaged or aging cells throughout the body. However, the efficacy of MSCs in cell-based therapy still remains unpredictable and carries with it enumerable risks. It is estimated that only 3-10% of MSCs survive transplantation, and there remains undefined and highly variable heterogeneous biological potency within these administered cell populations. The mode of action points to secreted factors produced by MSCs rather than the reliance on engraftment. Hence harnessing such secreted elements as a replacement for live-cell therapies is attractive. Extracellular vesicles (EVs) are heterogenous lipid bounded structures, secreted by cells. They comprise a complex repertoire of molecules including RNA, proteins and other factors that facilitate cell-to-cell communication. Described as protected signaling centers, EVs can modify the cellular activity of recipient cells and are emerging as a credible alternative to cell-based therapies. EV therapeutics demonstrate beneficial roles for wound healing by preventing apoptosis, moderating immune responses, and stimulating angiogenesis, in addition to promoting cell proliferation and differentiation required for tissue matrix synthesis. Significantly, EVs maintain their signaling function following transplantation, circumventing the issues related to cell-based therapies. However, EV research is still in its infancy in terms of their utility as medicinal agents, with many questions still surrounding mechanistic understanding, optimal sourcing, and isolation of EVs for regenerative medicine. This review will consider the efficacy of using cell-derived EVs compared to traditional cell-based therapies for bone repair and regeneration. We discuss the factors to consider in developing productive lines of inquiry and establishment of standardized protocols so that EVs can be harnessed from optimal secretome production, to deliver reproducible and effective therapies.
Collapse
|
34
|
Juhl AD, Wüstner D. Pathways and Mechanisms of Cellular Cholesterol Efflux-Insight From Imaging. Front Cell Dev Biol 2022; 10:834408. [PMID: 35300409 PMCID: PMC8920967 DOI: 10.3389/fcell.2022.834408] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
Cholesterol is an essential molecule in cellular membranes, but too much cholesterol can be toxic. Therefore, mammalian cells have developed complex mechanisms to remove excess cholesterol. In this review article, we discuss what is known about such efflux pathways including a discussion of reverse cholesterol transport and formation of high-density lipoprotein, the function of ABC transporters and other sterol efflux proteins, and we highlight their role in human diseases. Attention is paid to the biophysical principles governing efflux of sterols from cells. We also discuss recent evidence for cholesterol efflux by the release of exosomes, microvesicles, and migrasomes. The role of the endo-lysosomal network, lipophagy, and selected lysosomal transporters, such as Niemann Pick type C proteins in cholesterol export from cells is elucidated. Since oxysterols are important regulators of cellular cholesterol efflux, their formation, trafficking, and secretion are described briefly. In addition to discussing results obtained with traditional biochemical methods, focus is on studies that use established and novel bioimaging approaches to obtain insight into cholesterol efflux pathways, including fluorescence and electron microscopy, atomic force microscopy, X-ray tomography as well as mass spectrometry imaging.
Collapse
Affiliation(s)
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
35
|
Crowe CD, Keating CD. Microfluidic Control of Coexisting Chemical Microenvironments within Multiphase Water-in-Fluorocarbon Droplets. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:1811-1820. [PMID: 35090115 DOI: 10.1021/acs.langmuir.1c02929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The use of aqueous polymer-based phase separation within water-in-oil emulsion droplets provides a powerful platform for exploring the impact of compartmentalization and preferential partitioning on biologically relevant solutes. By forming an emulsion, a bulk solution is converted into a large number of chemically isolated microscale droplets. Microfluidic techniques provide an additional level of control over the formation of such systems. This enables the selective production of multiphase droplets with desired solution compositions and specific characteristics, such as solute partitioning. Here, we demonstrate control over the chemical microenvironment by adjusting the composition to increase tie line length for poly(ethylene glycol) (PEG)-dextran aqueous two-phase systems (ATPS) encapsulated within multiphase water-in-fluorocarbon oil emulsion droplets. Through rational adjustment of microfluidic parameters alone, ATPS droplets containing differing compositions could be produced during the course of a single experiment, with the produced droplets demonstrating a controllable range of tie line lengths. This provided control over partitioning behavior for biologically relevant macromolecules such that the difference in local protein concentration between adjacent phases could be rationally tuned. This work illustrates a broadly applicable technique to rationally create emulsified multiphase aqueous systems of desired compositions through the adjustment of microfluidic parameters alone, allowing for easy and rapid screening of various chemical microenvironments.
Collapse
Affiliation(s)
- Charles D Crowe
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Christine D Keating
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
36
|
Pereira Neves H, Max Dias Ferreira G, Max Dias Ferreira G, Rodrigues de Lemos L, Dias Rodrigues G, Albis Leão V, Barbosa Mageste A. Liquid-liquid extraction of rare earth elements using systems that are more environmentally friendly: Advances, challenges and perspectives. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2021.120064] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Kim J, Cho S, Park Y, Lee J, Park J. Evaluation of micro-RNA in extracellular vesicles from blood of patients with prostate cancer. PLoS One 2021; 16:e0262017. [PMID: 34972164 PMCID: PMC8719659 DOI: 10.1371/journal.pone.0262017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) contain various types of molecules including micro-RNAs, so isolating EVs can be an effective way to analyze and diagnose diseases. A lot of micro-RNAs have been known in relation to prostate cancer (PCa), and we evaluate miR-21, miR-141, and miR-221 in EVs and compare them with prostate-specific antigen (PSA). EVs were isolated from plasma of 38 patients with prostate cancer and 8 patients with benign prostatic hyperplasia (BPH), using a method that showed the highest recovery of RNA. Isolation of EVs concentrated micro-RNAs, reducing the cycle threshold (Ct) value of RT-qPCR amplification of micro-RNA such as miR-16 by 5.12 and miR-191 by 4.65, compared to the values before EV isolation. Normalization of target micro-RNAs was done using miR-191. For miR-221, the mean expression level of patients with localized PCa was significantly higher than that of the control group, having 33.45 times higher expression than the control group (p < 0.01). Area under curve (AUC) between BPH and PCa for miR-221 was 0.98 (p < 0.0001), which was better than AUC for prostate-specific antigen (PSA) level in serum for the same patients. The levels of miR-21 and miR-141 in EVs did not show significant changes in patients with PCa compared to the control group in this study. This study suggests isolating EVs can be a helpful approach in analyzing micro-RNAs with regard to disease.
Collapse
Affiliation(s)
- Jiyoon Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
| | - Siwoo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
| | - Yonghyun Park
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jiyoul Lee
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jaesung Park
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
| |
Collapse
|
38
|
Ren Y, Ge K, Sun D, Hong Z, Jia C, Hu H, Shao F, Yao B. Rapid enrichment and sensitive detection of extracellular vesicles through measuring the phospholipids and transmembrane protein in a microfluidic chip. Biosens Bioelectron 2021; 199:113870. [PMID: 34915212 DOI: 10.1016/j.bios.2021.113870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 11/02/2022]
Abstract
Extracellular vesicles (EVs) have attracted tremendous attention in recent years and quantification of EVs is a key issue in the evaluation of vesicle-based diagnostics and therapeutic development, but it's quite challenging to determine whether higher protein expression signals are due to larger vesicle amount or higher protein content within each vesicle. To solve this problem, herein, we proposed a strategy based on staining phospholipid bilayers of EVs with lipophilic dyes to evaluate their lipid amount, which was subsequently normalized as an internal standard for studying the expression of transmembrane protein (i.e., CD63) on EVs in different samples. In addition, a microfluidic platform based on electrophoresis technology was invented to effectively enrich and detect EVs. Small fluorescent labeling molecules (i.e., uncombined aptamers) were on-chip removed from EVs without pre-separation via ultracentrifugation or ultrafiltration which were indispensable in nanoparticle tracking analysis (NTA) and flow cytometry techniques and the performance of this assay is comparable to NTA. Finally, it was found obvious difference in the expression of CD63 on EVs before and after normalization based on lipid amount in plasma samples. This method is expected to provide more accurate information when comparing the expression levels of EVs biomarkers in different samples.
Collapse
Affiliation(s)
- Yongan Ren
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Ke Ge
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, The Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, China
| | - Danyang Sun
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Zichen Hong
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Changku Jia
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, The Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, China
| | - Huan Hu
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, China
| | - Fangwei Shao
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou, China.
| |
Collapse
|
39
|
Chen C, Wang J, Sun M, Li J, Wang HMD. Toward the next-generation phyto-nanomedicines: cell-derived nanovesicles (CDNs) for natural product delivery. Biomed Pharmacother 2021; 145:112416. [PMID: 34781147 DOI: 10.1016/j.biopha.2021.112416] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 02/08/2023] Open
Abstract
Phytochemicals are plant-derived bioactive compounds, which have been widely used for therapeutic purposes. Due to the poor water-solubility, low bioavailability and non-specific targeting characteristic, diverse classes of nanocarriers are utilized for encapsulation and delivery of bio-effective agents. Cell-derived nanovesicles (CDNs), known for exosomes or extracellular vesicles (EVs), are biological nanoparticles with multiple functions. Compared to the artificial counterpart, CDNs hold great potential in drug delivery given the higher stability, superior biocompatibility and the lager capability of encapsulating bioactive molecules. Here, we provide a bench-to-bedside review of CDNs-based nanoplatform, including the bio-origin, preparation, characterization and functionalization. Beyond that, the focus is laid on the therapeutic effect of CDNs-mediated drug delivery for natural products. The state-of-art development as well as some pre-clinical applications of using CDNs for disease treatment is also summarized. It is highly expected that the continuing development of CDNs-based delivery systems will further promote the clinical utilization and translation of phyto-nanomedicines.
Collapse
Affiliation(s)
- Chaoxiang Chen
- College of Food and Biological Engineering, Jimei University, China
| | - Jialin Wang
- College of Food and Biological Engineering, Jimei University, China
| | - Mengdi Sun
- College of Food and Biological Engineering, Jimei University, China
| | - Jian Li
- College of Food and Biological Engineering, Jimei University, China.
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
40
|
Aqueous Two-Phase System–Ion Chromatography for Determination of Thiocyanate in Raw Milk. SEPARATIONS 2021. [DOI: 10.3390/separations8110212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Thiocyanate could effectively inhibit bacteria in milk and extend the shelf life of milk. However, excessive addition will lead to health risks. Therefore, the determination of thiocyanate in raw milk has received a lot of attention, but the determination could be interfered with by other components in raw milk and the pre-treatment of raw milk is complex. In this study, a new pretreatment method combined with ion chromatography (IC) for rapid and sensitive determination of thiocyanate is proposed. An acetonitrile/(NH4)2SO4 aqueous two-phase system (ATPS) was developed for the separation and enrichment of thiocyanate in raw milk. Response surface methodology was performed to optimize the extraction conditions and an efficient pretreatment were obtained using ATPS composed of 42% acetonitrile (w/w) and 16% (NH4)2SO4 (w/w), with the pH 4.7, and the recovery of thiocyanate reached 107.24 ± 0.5%, and the enrichment ratio was 10.74 ± 0.03. IC was used to establish a thiocyanate enrichment method. The linear range was from 0.05 to 15 mg/L and R2 = 0.998, the limit of detection (LOD) was 0.2 μg/L, the limit of quantification (LQD) was 0.6 μg/L. Hence, it is feasible to combine ATPS with IC for the enrichment and determination of thiocyanate in raw milk.
Collapse
|
41
|
Qin B, Hu XM, Su ZH, Zeng XB, Ma HY, Xiong K. Tissue-derived extracellular vesicles: Research progress from isolation to application. Pathol Res Pract 2021; 226:153604. [PMID: 34500372 DOI: 10.1016/j.prp.2021.153604] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are the structures that all cells release into the environment. They are separated by a lipid bilayer and contain the cellular components that release them. To date, most studies have been performed on EVs derived from cell supernatants or different body fluids, while the number of studies on EV isolation directly from tissues is still limited. Studies of EV isolation directly from tissues may provide us with better information. This review summarizes the role of EV in the extracellular matrix, the protocol for isolation of EV in the tissue interstitium, and the application of the protocol in different tissues.
Collapse
Affiliation(s)
- Bo Qin
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District Guilin North Road No.16, Huangshi 435003, China
| | - Xi-Min Hu
- Clinical Medicine Eight-year Program, 02 Class, 17 Grade, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Zhen-Hong Su
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District Guilin North Road No.16, Huangshi 435003, China
| | - Xiao-Bo Zeng
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District Guilin North Road No.16, Huangshi 435003, China
| | - Hong-Ying Ma
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Xialu District Guilin North Road No.16, Huangshi 435003, China
| | - Kun Xiong
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Ophthalmology, Changsha, Hunan 410008, China.
| |
Collapse
|
42
|
Min L, Wang B, Bao H, Li X, Zhao L, Meng J, Wang S. Advanced Nanotechnologies for Extracellular Vesicle-Based Liquid Biopsy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102789. [PMID: 34463056 PMCID: PMC8529441 DOI: 10.1002/advs.202102789] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 05/09/2023]
Abstract
Extracellular vesicles (EVs) are emerging as a new source of biomarkers in liquid biopsy because of their wide presence in most body fluids and their ability to load cargoes from disease-related cells. Owing to the crucial role of EVs in disease diagnosis and treatment, significant efforts have been made to isolate, detect, and analyze EVs with high efficiency. A recent overview of advanced EV detection nanotechnologies is discussed here. First, several key challenges in EV-based liquid biopsies are introduced. Then, the related pivotal advances in nanotechnologies for EV isolation based on physical features, chemical affinity, and the combination of nanostructures and chemical affinity are summarized. Next, a summary of high-sensitivity sensors for EV detection and advanced approaches for single EV detection are provided. Later, EV analysis is introduced in practical clinical scenarios, and the application of machine learning in this field is highlighted. Finally, future opportunities for the development of next-generation nanotechnologies for EV detection are presented.
Collapse
Affiliation(s)
- Li Min
- Department of GastroenterologyBeijing Friendship HospitalCapital Medical UniversityNational Clinical Research Center for Digestive DiseasesBeijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijing100050P. R. China
| | - Binshuai Wang
- Department of UrologyPeking University Third HospitalBeijing100191P. R. China
| | - Han Bao
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Xinran Li
- Department of UrologyPeking University Third HospitalBeijing100191P. R. China
| | - Libo Zhao
- Echo Biotech Co., Ltd.Beijing102206P. R. China
| | - Jingxin Meng
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shutao Wang
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
43
|
Kim CJ, Dong L, Amend SR, Cho YK, Pienta KJ. The role of liquid biopsies in prostate cancer management. LAB ON A CHIP 2021; 21:3263-3288. [PMID: 34346466 DOI: 10.1039/d1lc00485a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Liquid biopsy has emerged as a complement to invasive tissue biopsy to guide cancer diagnosis and treatment. The common liquid biopsy biomarkers are circulating tumor cells (CTCs), extracellular vesicles (EVs), and circulating tumor DNA (ctDNA). Each biomarker provides specific information based on its intrinsic characteristics. Prostate cancer is the second most common cancer in males worldwide. In men with low-grade localized prostate cancer, the disease can often be managed by active surveillance. For men who require treatment, the 5-year survival rate of localized prostate cancer is the highest among all cancer types, but the metastatic disease remains incurable. Metastatic prostate cancer invariably progresses to involve multiple bone sites and develops into a castration-resistant disease that leads to cancer death. The need to appropriately diagnose and guide the serial treatment of men with prostate cancer has led to the implementation of many studies to apply liquid biopsies to prostate cancer management. This review describes recent advancements in isolation and detection technology and the strength and weaknesses of the three circulating biomarkers. The clinical studies based on liquid biopsy results are summarized to depict the future perspective in the role of liquid biopsy on prostate cancer management.
Collapse
Affiliation(s)
- Chi-Ju Kim
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | | | |
Collapse
|
44
|
Ahmed T, Yamanishi C, Kojima T, Takayama S. Aqueous Two-Phase Systems and Microfluidics for Microscale Assays and Analytical Measurements. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2021; 14:231-255. [PMID: 33950741 DOI: 10.1146/annurev-anchem-091520-101759] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Phase separation is a common occurrence in nature. Synthetic and natural polymers, salts, ionic liquids, surfactants, and biomacromolecules phase separate in water, resulting in an aqueous two-phase system (ATPS). This review discusses the properties, handling, and uses of ATPSs. These systems have been used for protein, nucleic acid, virus, and cell purification and have in recent years found new uses for small organics, polysaccharides, extracellular vesicles, and biopharmaceuticals. Analytical biochemistry applications such as quantifying protein-protein binding, probing for conformational changes, or monitoring enzyme activity have been performed with ATPSs. Not only are ATPSs biocompatible, they also retain their properties at the microscale, enabling miniaturization experiments such as droplet microfluidics, bacterial quorum sensing, multiplexed and point-of-care immunoassays, and cell patterning. ATPSs include coacervates and may find wider interest in the context of intracellular phase separation and origin of life. Recent advances in fundamental understanding and in commercial application are also considered.
Collapse
Affiliation(s)
- Tasdiq Ahmed
- Walter H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA;
| | - Cameron Yamanishi
- Walter H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA;
| | - Taisuke Kojima
- Walter H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA;
| | - Shuichi Takayama
- Walter H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA;
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
45
|
Kim J, Shim JS, Han BH, Kim HJ, Park J, Cho IJ, Kang SG, Kang JY, Bong KW, Choi N. Hydrogel-based hybridization chain reaction (HCR) for detection of urinary exosomal miRNAs as a diagnostic tool of prostate cancer. Biosens Bioelectron 2021; 192:113504. [PMID: 34298498 DOI: 10.1016/j.bios.2021.113504] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
Although urinary exosomal microRNAs (miRNAs) have recently emerged as potential biomarkers, clinical applications are still limited due to their low concentration in small volumes of clinical samples. Therefore, the development of a non-invasive, specific diagnostic tool, along with profiling exosomal miRNA markers from urine, remains a significant challenge. Here, we present hydrogel-based hybridization chain reaction (HCR) for multiplex signal amplification to detect urinary exosomal miRNAs from human clinical samples. We succeeded in identifying small amounts (~amol) of exosomal miRNAs from 600 μL of urine with up to ~35-fold amplification and enhanced detection limits by over an order of magnitude for two miRNA biomarker candidates, hsa-miR-6090 and hsa-miR-3665. Furthermore, we proposed ratiometric analysis without requiring normalization to a reference miRNA and validated the clinical diagnostic potential toward differentiating prostate cancer patients from healthy controls. Our hydrogel-based HCR could serve as a new diagnostic platform for a non-invasive liquid biopsy before burdensome tissue biopsy of various diseases, including prostate cancer screening, complementing the PSA test.
Collapse
Affiliation(s)
- Junbeom Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea; Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, South Korea
| | - Ji Sung Shim
- Department of Urology, Korea University College of Medicine, Seoul, 02841, South Korea
| | - Bo Hoon Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea; School of Mechanical Engineering, Korea University, Seoul, 02841, South Korea
| | - Hye Jin Kim
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Jaesung Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea; School of Interdisciplinary Bioscience and Bioengineering. Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Il-Joo Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, South Korea; School of Electrical and Electronics Engineering, Yonsei University, Seoul, 03722, South Korea; Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, South Korea
| | - Sung Gu Kang
- Department of Urology, Korea University College of Medicine, Seoul, 02841, South Korea
| | - Ji Yoon Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, South Korea
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, South Korea.
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
46
|
Villarreal-Leal RA, Cooke JP, Corradetti B. Biomimetic and immunomodulatory therapeutics as an alternative to natural exosomes for vascular and cardiac applications. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 35:102385. [PMID: 33774130 PMCID: PMC8238887 DOI: 10.1016/j.nano.2021.102385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/21/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Inflammation is a central mechanism in cardiovascular diseases (CVD), where sustained oxidative stress and immune responses contribute to cardiac remodeling and impairment. Exosomes are extracellular vesicles released by cells to communicate with their surroundings and to modulate the tissue microenvironment. Recent evidence indicates their potential as cell-free immunomodulatory therapeutics for CVD, preventing cell death and fibrosis while inducing wound healing and angiogenesis. Biomimetic exosomes are semi-synthetic particles engineered using essential moieties present in natural exosomes (lipids, RNA, proteins) to reproduce their therapeutic effects while improving on scalability and standardization due to the ample range of moieties available to produce them. In this review, we provide an up-to-date description of the use of exosomes for CVD and offer our vision on the areas of opportunity for the development of biomimetic strategies. We also discuss the current limitations to overcome in the process towards their translation into clinic.
Collapse
Affiliation(s)
- Ramiro A Villarreal-Leal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
| | - John P Cooke
- RNA Therapeutics Program, Department of Cardiovascular Sciences (R.S., J.P.C.), Houston Methodist Research Institute, TX, USA; Houston Methodist DeBakey Heart and Vascular Center (J.P.C.), Houston Methodist Hospital, TX, USA
| | - Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Center of NanoHealth, Swansea University Medical School, Swansea, UK.
| |
Collapse
|
47
|
Chen Q, Zhang Y, Chen H, Liu J, Liu J. Enhancing the Sensitivity of DNA and Aptamer Probes in the Dextran/PEG Aqueous Two-Phase System. Anal Chem 2021; 93:8577-8584. [PMID: 34101437 DOI: 10.1021/acs.analchem.1c01419] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Increasing the local concentration of DNA-based probes is a convenient way to improve the sensitivity of biosensors. Instead of using organic solvents or ionic liquids that phase-separate with water based on hydrophobic interactions, we herein studied a classic aqueous two-phase system (ATPS) comprising polyethylene glycol (PEG) and dextran. Polymers of higher molecular weights and higher concentrations favored phase separation. DNA oligonucleotides are selectively enriched in the dextran-rich phase unless the pH was increased to 12. A higher volume ratio of PEG-to-dextran and a higher concentration of PEG also enrich more DNA probes in the dextran-rich phase. The partition efficiency of the T15 DNA was enriched around seven times in the dextran phase when the volume ratio of dextran and PEG reached 1:10. The detection of limit improved by 3.6-fold in a molecular beacon-based DNA detection system with the ATPS. The ATPS also increased the sensitivity for the detection of Hg2+ and adenosine triphosphate, although these target molecules alone distributed equally in the two phases. This work demonstrates a simple method using water soluble polymers to improve biosensors.
Collapse
Affiliation(s)
- Qiaoshu Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China.,Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Yanwen Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Hui Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Jianbo Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
48
|
Al Naem M, Bourebaba L, Kucharczyk K, Röcken M, Marycz K. Therapeutic mesenchymal stromal stem cells: Isolation, characterization and role in equine regenerative medicine and metabolic disorders. Stem Cell Rev Rep 2021; 16:301-322. [PMID: 31797146 DOI: 10.1007/s12015-019-09932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a popular treatment modality in equine orthopaedics. Regenerative therapies are especially interesting for pathologies like complicated tendinopathies of the distal limb, osteoarthritis, osteochondritis dissecans (OCD) and more recently metabolic disorders. Main sources for MSC harvesting in the horse are bone marrow, adipose tissue and umbilical cord blood. While the acquisition of umbilical cord blood is fairly easy and non-invasive, extraction of bone marrow and adipose tissue requires more invasive techniques. Characterization of the stem cells as a result of any isolation method, is also a crucial step for the confirmation of the cells' stemness properties; thus, three main characteristics must be fulfilled by these cells, namely: adherence, expression of a series of well-defined differentiation clusters as well as pluripotency. EVs, resulting from the paracrine action of MSCs, also play a key role in the therapeutic mechanisms mediated by stem cells; MSC-EVs are thus largely implicated in the regulation of proliferation, maturation, polarization and migration of various target cells. Evidence that EVs alone represent a complex network 0involving different soluble factors and could then reflect biophysical characteristics of parent cells has fuelled the importance of developing highly specific techniques for their isolation and analysis. All these aspects related to the functional and technical understanding of MSCs will be discussed and summarized in this review.
Collapse
Affiliation(s)
- Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kucharczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
49
|
Liu J, Yoo J, Ho JY, Jung Y, Lee S, Hur SY, Choi YJ. Plasma-derived exosomal miR-4732-5p is a promising noninvasive diagnostic biomarker for epithelial ovarian cancer. J Ovarian Res 2021; 14:59. [PMID: 33910598 PMCID: PMC8082916 DOI: 10.1186/s13048-021-00814-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Exosomal miRNAs regulate gene expression and play important roles in several diseases. We used exosomal miRNA profiling to investigate diagnostic biomarkers of epithelial ovarian cancer (EOC). METHODS In total, 55 individuals were enrolled, comprising healthy (n = 21) and EOC subjects (n = 34). Small mRNA (smRNA) sequencing and real-time PCR (RT-PCR) were performed to identify potential biomarkers. Receiver operating characteristic (ROC) curves were conducted to determine biomarker sensitivity and specificity. RESULTS Using smRNA sequencing, we identified seven up-regulated (miR-4732-5p, miR-877-5p, miR-574-3p, let-7a-5p, let-7b-5p, let-7c-5p, and let-7f-5p) and two down-regulated miRNAs (miR-1273f and miR-342-3p) in EOC patients when compared with healthy subjects. Of these, miR-4732-5p and miR-1273f were the most up-regulated and down-regulated respectively, therefore they were selected for RT-PCR analysis. Plasma derived exosomal miR-4732-5p had an area under the ROC curve of 0.889, with 85.7% sensitivity and 82.4% specificity in distinguishing EOC patients from healthy subjects (p<0.0001) and could be a potential biomarker for monitoring the EOC progression from early stage to late stage (p = 0.018). CONCLUSIONS Plasma derived exosomal miR-4732-5p may be a promising candidate biomarker for diagnosing EOC.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jigeun Yoo
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yoon Ho
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yuyeon Jung
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sanha Lee
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soo Young Hur
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youn Jin Choi
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Lee J, Kwon Y, Jung J, Shin H, Park J. Immunostaining Extracellular Vesicles Based on an Aqueous Two-Phase System: For Analysis of Tetraspanins. ACS APPLIED BIO MATERIALS 2021; 4:3294-3303. [PMID: 35014416 DOI: 10.1021/acsabm.0c01625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Immunostaining of extracellular vesicles (EVs) has become necessary for the characterization of EV subtypes, clarification of the EV biogenesis/cellular uptake pathway, drug delivery, etc. Immunostained EVs must be in suspension for further downstream analyses or uses. However, conventional EV immunostaining methods yielding EVs in suspension lack either sufficient recovery or staining specificity because of the washing steps. In this study, we have devised and tested a method to wash immunostained EVs with successive aqueous two-phase system (ATPS) separations. The ATPS is a liquid-liquid extraction procedure that ensures a gentle separation of target molecules. The ATPS has been successfully employed to separate EVs from other impurities with high yield and high purity. Immunostained EVs were washed with the ATPS and compared with other immunostaining methods to confirm the proposed method's high EV recovery and staining accuracy. According to the result, the ATPS-based EV immunostaining method required as low as ∼1 μg without compromise of accuracy and recovery.
Collapse
Affiliation(s)
- Jingeol Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk 790-784, Republic of Korea
| | - Yongmin Kwon
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk 790-784, Republic of Korea
| | - Jaehun Jung
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk 790-784, Republic of Korea
| | - Hyunwoo Shin
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk 790-784, Republic of Korea
| | - Jaesung Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk 790-784, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeong-buk 790-784, Republic of Korea
| |
Collapse
|