1
|
Talatapeh SP, Rezaie J, Nejati V. Extracellular Vesicle-based Delivery of Paclitaxel to Lung Cancer Cells: Uptake, Anticancer Effects, Autophagy and Mitophagy Pathways. Arch Med Res 2025; 56:103194. [PMID: 39922153 DOI: 10.1016/j.arcmed.2025.103194] [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: 09/08/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Due to their unique properties, extracellular vesicles (EVs) are promising nanocarriers for exogenous drug delivery. AIM We prepared a drug delivery system based on large EVs (LEVs) containing paclitaxel (PTX) (LEVs-PTX) to investigate anticancer effects on lung cancer cells with a focus on autophagy. METHODS LEVs-PTX were isolated from lung cancer cells by ultracentrifugation and characterized using different techniques. Rhodamine B dye (Rh B) was used to label LEVs-PTX for cell tracking. MTT assay was performed to investigate the cellular toxicity of PTX and LEVs-PTX for 24 h and 48 h. The uptake of LEVs-PTX was monitored by immunofluorescence microscopy in breast and lung cancer cells. A colorimetric assay was performed to evaluate apoptosis, while Western blotting assays were used to investigate autophagy proteins. Real-time PCR was used to measure mitophagy genes. RESULTS Characterization techniques showed that LEVs were isolated and loaded with PTX. Rh B labeled LEVs, which was confirmed by a fluorescence spectrophotometer. Immunofluorescence microscopy showed that the lung and breast cancer cells had captured LEVs. Cell viability was decreased in LEVs-PTX cells which coincided with an increase in caspase-3 activity in LEVs-PTX cells. The Beclin-1 protein level and LC3 II/I ratio decreased, while the P62 protein level was increased in LEVs-PTX cells. The mitophagy genes such as Pink-1 and Parkin were upregulated in LEVs-PTX cells. CONCLUSION The data show that LEVs-PTX induced apoptosis, which inhibited the autophagy pathway and increased mitophagy markers, suggesting damage to cell organelles through intracellular delivery of PTX.
Collapse
Affiliation(s)
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia, Iran.
| | - Vahid Nejati
- Department of Biology, Urmia University, Urmia, Iran
| |
Collapse
|
2
|
Cheravi M, Baharara J, Yaghmaei P, Roudbari NH. Differentiation of Human Adipose-derived Stem Cells to Exosome-affected Neural-like Cells Extracted from Human Cerebrospinal Fluid Using Bioprinting Process. Curr Stem Cell Res Ther 2024; 19:1042-1054. [PMID: 37957915 DOI: 10.2174/011574888x270145231102062259] [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: 07/26/2023] [Revised: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Advancement in tissue engineering has provided novel solutions for creating scaffolds as well as applying induction factors in the differentiation of stem cells. The present research aimed to investigate the differentiation of human adipose-derived mesenchymal stem cells to neural-like cells using the novel bioprinting method, as well as the effect of cerebrospinal fluid exosomes. METHODS In the present study, the extent of neuronal proliferation and differentiation of adipose- derived stem cells were explored using the MTT method, immunocytochemistry, and real-- time PCR in the scaffolds created by the bioprinting process. Furthermore, in order to investigate the veracity of the identity of the CSF (Cerebrospinal fluid) derived exosomes, after the isolation of exosomes, dynamic light scattering (DLS), scanning electron microscopy (SEM), and atomic force microscopy (AFM) techniques were used. RESULTS MTT findings indicated survivability and proliferation of cells in the scaffolds created by the bioprinting process during a 14-day period. The results obtained from real-time PCR showed that the level of MAP2 gene (Microtubule Associated Protein 2) expression increased on days 7 and 14, while the expression of the Nestin gene (intermediate filament protein) significantly decreased compared to the control. The investigation to confirm the identity of exosomes indicated that the CSF-derived exosomes had a spherical shape with a 40-100 nm size. CONCLUSION CSF-derived exosomes can contribute to the neuronal differentiation of adipose- derived stem cells in alginate hydrogel scaffolds created by the bioprinting process.
Collapse
Affiliation(s)
- Mojtaba Cheravi
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Javad Baharara
- Department of Biology and Research Center for Animal Development Applied Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nasim Hayati Roudbari
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Gao H, Zeng Y, Huang X, A L, Liang Q, Xie J, Lin X, Gong J, Fan X, Zou T, Xu H. Extracellular vesicles from organoid-derived human retinal progenitor cells prevent lipid overload-induced retinal pigment epithelium injury by regulating fatty acid metabolism. J Extracell Vesicles 2024; 13:e12401. [PMID: 38151470 PMCID: PMC10752800 DOI: 10.1002/jev2.12401] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/31/2023] [Accepted: 12/02/2023] [Indexed: 12/29/2023] Open
Abstract
Retinal degeneration (RD), a group of diseases leading to irreversible vision loss, is characterised by retinal pigment epithelium (RPE) or retinal neuron damage and loss. With fewer risks of immune rejection and tumorigenesis, stem cell-secreted extracellular vesicles (EVs) offer a new cell-free therapeutic paradigm for RD, which remains to be investigated. Human retinal organoid-derived retinal progenitor cells (hERO-RPCs) are an easily accessible and advanced cell source for RD treatment. However, hERO-RPCs-derived EVs require further characterisation. Here, we compared the characteristics of EVs from hERO-RPCs (hRPC-EVs) with those of human embryonic stem cell (hESC)-derived EVs (hESC-EVs) as controls. Based on in-depth proteomic analysis, we revealed remarkable differences between hRPC-EVs and hESC-EVs. A comparison between EVs and their respective cells of origin demonstrated that the protein loading of hRPC-EVs was more selective than that of hESC-EVs. In particular, hESC-EVs were enriched with proteins related to angiogenesis and cell cycle, whereas hRPC-EVs were enriched with proteins associated with immune modulation and retinal development. More importantly, compared with that of hESC-EVs, hRPC-EVs exhibited a lower correlation with cell proliferation and a unique capacity to regulate lipid metabolism. It was further confirmed that hRPC-EVs potentially eliminated lipid deposits, inhibited lipotoxicity and oxidative stress, and enhanced phagocytosis and survival of oleic acid-treated ARPE-19 cells. Mechanistically, hRPC-EVs are integrated into the mitochondrial network of oleic acid-treated ARPE-19 cells, and increased the level of mitochondrial fatty acid β-oxidation-related proteins. Thus, organoid-derived hRPC-EVs represent a promising source of cell-free therapy for RD, especially for blinding diseases related to abnormal lipid metabolism in RPE cells.
Collapse
Affiliation(s)
- Hui Gao
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| | - Yuxiao Zeng
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| | - Xiaona Huang
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| | - Luodan A
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| | - Qingle Liang
- Department of Clinical Laboratory Medicine, First Affiliated HospitalThird Military Medical University (Army Medical University)ChongqingChina
| | - Jing Xie
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| | - Xi Lin
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| | - Jing Gong
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of PsychologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Ting Zou
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
- Department of OphthalmologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
- Key Lab of Visual Damage and Regeneration & Restoration of ChongqingChongqingChina
| |
Collapse
|
4
|
Abudurexiti M, Zhao Y, Wang X, Han L, Liu T, Wang C, Yuan Z. Bio-Inspired Nanocarriers Derived from Stem Cells and Their Extracellular Vesicles for Targeted Drug Delivery. Pharmaceutics 2023; 15:2011. [PMID: 37514197 PMCID: PMC10386614 DOI: 10.3390/pharmaceutics15072011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
With their seemingly limitless capacity for self-improvement, stem cells have a wide range of potential uses in the medical field. Stem-cell-secreted extracellular vesicles (EVs), as paracrine components of stem cells, are natural nanoscale particles that transport a variety of biological molecules and facilitate cell-to-cell communication which have been also widely used for targeted drug delivery. These nanocarriers exhibit inherent advantages, such as strong cell or tissue targeting and low immunogenicity, which synthetic nanocarriers lack. However, despite the tremendous therapeutic potential of stem cells and EVs, their further clinical application is still limited by low yield and a lack of standardized isolation and purification protocols. In recent years, inspired by the concept of biomimetics, a new approach to biomimetic nanocarriers for drug delivery has been developed through combining nanotechnology and bioengineering. This article reviews the application of biomimetic nanocarriers derived from stem cells and their EVs in targeted drug delivery and discusses their advantages and challenges in order to stimulate future research.
Collapse
Affiliation(s)
- Munire Abudurexiti
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| | - Yue Zhao
- Department of Pharmacy, Sichuan Tianfu New Area People’s Hospital, Chengdu 610213, China;
| | - Xiaoling Wang
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia;
| | - Chengwei Wang
- Division of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhixiang Yuan
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| |
Collapse
|
5
|
Matos BMD, Stimamiglio MA, Correa A, Robert AW. Human pluripotent stem cell-derived extracellular vesicles: From now to the future. World J Stem Cells 2023; 15:453-465. [PMID: 37342215 PMCID: PMC10277970 DOI: 10.4252/wjsc.v15.i5.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/14/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
Extracellular vesicles (EVs) are nanometric particles that enclose cell-derived bioactive molecules in a lipid bilayer and serve as intercellular communication tools. Accordingly, in various biological contexts, EVs are reported to engage in immune modulation, senescence, and cell proliferation and differentiation. Therefore, EVs could be key elements for potential off-the-shelf cell-free therapy. Little has been studied regarding EVs derived from human pluripotent stem cells (hPSC-EVs), even though hPSCs offer good opportunities for induction of tissue regeneration and unlimited proliferative ability. In this review article, we provide an overview of studies using hPSC-EVs, focusing on identifying the conditions in which the cells are cultivated for the isolation of EVs, how they are characterized, and applications already demonstrated. The topics reported in this article highlight the incipient status of the studies in the field and the significance of hPSC-EVs’ prospective applications as PSC-derived cell-free therapy products.
Collapse
Affiliation(s)
- Bruno Moises de Matos
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| | | | - Alejandro Correa
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| | - Anny Waloski Robert
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| |
Collapse
|
6
|
Yedavilli S, Singh AD, Singh D, Samal R. Nano-Messengers of the Heart: Promising Theranostic Candidates for Cardiovascular Maladies. Front Physiol 2022; 13:895322. [PMID: 35899033 PMCID: PMC9313536 DOI: 10.3389/fphys.2022.895322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Till date, cardiovascular diseases remain a leading cause of morbidity and mortality across the globe. Several commonly used treatment methods are unable to offer safety from future complications and longevity to the patients. Therefore, better and more effective treatment measures are needed. A potential cutting-edge technology comprises stem cell-derived exosomes. These nanobodies secreted by cells are intended to transfer molecular cargo to other cells for the establishment of intercellular communication and homeostasis. They carry DNA, RNA, lipids, and proteins; many of these molecules are of diagnostic and therapeutic potential. Several stem cell exosomal derivatives have been found to mimic the cardioprotective attributes of their parent stem cells, thus holding the potential to act analogous to stem cell therapies. Their translational value remains high as they have minimal immunogenicity, toxicity, and teratogenicity. The current review highlights the potential of various stem cell exosomes in cardiac repair, emphasizing the recent advancements made in the development of cell-free therapeutics, particularly as biomarkers and as carriers of therapeutic molecules. With the use of genetic engineering and biomimetics, the field of exosome research for heart treatment is expected to solve various theranostic requirements in the field paving its way to the clinics.
Collapse
Affiliation(s)
- Sneha Yedavilli
- Department of Life Science, Central University of Karnataka, Kalaburagi, India
| | | | - Damini Singh
- Environmental Pollution Analysis Lab, Bhiwadi, India
| | - Rasmita Samal
- Department of Life Science, Central University of Karnataka, Kalaburagi, India
- *Correspondence: Rasmita Samal,
| |
Collapse
|
7
|
Veizades S, Tso A, Nguyen PK. Infection, inflammation and thrombosis: a review of potential mechanisms mediating arterial thrombosis associated with influenza and severe acute respiratory syndrome coronavirus 2. Biol Chem 2021; 403:231-241. [PMID: 34957734 DOI: 10.1515/hsz-2021-0348] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/07/2021] [Indexed: 12/30/2022]
Abstract
Thrombosis has long been reported as a potentially deadly complication of respiratory viral infections and has recently received much attention during the global coronavirus disease 2019 pandemic. Increased risk of myocardial infarction has been reported during active infections with respiratory viruses, including influenza and severe acute respiratory syndrome coronavirus 2, which persists even after the virus has cleared. These clinical observations suggest an ongoing interaction between these respiratory viruses with the host's coagulation and immune systems that is initiated at the time of infection but may continue long after the virus has been cleared. In this review, we discuss the epidemiology of viral-associated myocardial infarction, highlight recent clinical studies supporting a causal connection, and detail how the virus' interaction with the host's coagulation and immune systems can potentially mediate arterial thrombosis.
Collapse
Affiliation(s)
- Stefan Veizades
- Department of Medicine (Cardiovascular Medicine), Stanford University, Stanford, CA 94305, USA.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA.,Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Alexandria Tso
- Department of Medicine (Cardiovascular Medicine), Stanford University, Stanford, CA 94305, USA.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Patricia K Nguyen
- Department of Medicine (Cardiovascular Medicine), Stanford University, Stanford, CA 94305, USA.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Cui L, Saeed Y, Li H, Yang J. Regenerative medicine and traumatic brain injury: from stem cell to cell-free therapeutic strategies. Regen Med 2021; 17:37-53. [PMID: 34905963 DOI: 10.2217/rme-2021-0069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious health concern, yet there is a lack of standardized treatment to combat its long-lasting effects. The objective of the present study was to provide an overview of the limitation of conventional stem-cell therapy in the treatment of TBI and to discuss the application of novel acellular therapies and their advanced strategies to enhance the efficacy of stem cells derived therapies in the light of published study data. Moreover, we also discussed the factor to optimize the therapeutic efficiency of stem cell-derived acellular therapy by overcoming the challenges for its clinical translation. Hence, we concluded that acellular therapy possesses the potential to bring a breakthrough in the field of regenerative medicine to treat TBI.
Collapse
Affiliation(s)
- Lianxu Cui
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Yasmeen Saeed
- Guangdong VitaLife Biotechnology Co., LTD, 61 Xiannan Road, Nanhai District, Foshan, Guangdong, 528200, PR China
| | - Haomin Li
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Jingli Yang
- School of medicine, Foshan University, 18 Jiangwan Road, Foshan, Guangdong, 528000, PR China
| |
Collapse
|
9
|
Takahashi T, Schleimer RP. Epithelial-Cell-Derived Extracellular Vesicles in Pathophysiology of Epithelial Injury and Repair in Chronic Rhinosinusitis: Connecting Immunology in Research Lab to Biomarkers in Clinics. Int J Mol Sci 2021; 22:11709. [PMID: 34769139 PMCID: PMC8583779 DOI: 10.3390/ijms222111709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Epithelial barrier disruption and failure of epithelial repair by aberrant epithelial-mesenchymal transition (EMT)-induced basal cells observed in nasal mucosa of chronic rhinosinusitis (CRS) are speculated to play important roles in disease pathophysiology. Microparticles (MPs) are a type of extracellular vesicle (EV) released by budding or shedding from the plasma membrane of activated or apoptotic cells. MPs are detected in nasal lavage fluids (NLFs) and are now receiving attention as potential biomarkers to evaluate the degree of activation of immune cells and injury of structural cells in nasal mucosa of subjects with sinus disease. There are three types of epithelial-cell-derived MPs, which are defined by the expression of different epithelial specific markers on their surface: EpCAM, E-cadherin, and integrin β6 (ITGB6). When these markers are on MPs that are also carrying canonical EMT/mesenchymal markers (Snail (SNAI1); Slug (SNAI2); alpha-smooth muscle actin (αSMA, ACTA2)) or pro- and anti-coagulant molecules (tissue factor (TF); tissue plasminogen activator (tPA); plasminogen activator inhibitor-1 (PAI-1)), they provide insight as to the roles of epithelial activation for EMT or regulation of coagulation in the underlying disease. In this review, we discuss the potential of epithelial MPs as research tools to evaluate status of nasal mucosae of CRS patients in the lab, as well as biomarkers for management and treatment of CRS in the clinic.
Collapse
Affiliation(s)
- Toru Takahashi
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Robert P Schleimer
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
10
|
Heirani-Tabasi A, Hosseinzadeh S, Rabbani S, Ahmadi Tafti SH, Jamshidi K, Soufizomorrod M, Soleimani M. Cartilage tissue engineering by co-transplantation of chondrocyte extracellular vesicles and mesenchymal stem cells, entrapped in chitosan-hyaluronic acid hydrogel. Biomed Mater 2021; 16. [PMID: 34144542 DOI: 10.1088/1748-605x/ac0cbf] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) on injectable hydrogels are mostly used to regenerate articular cartilage, which would have a variety of outcomes. Chondrocyte extracellular vesicles (EVs) have attracted many attentions for their chondrogenic differentiation capacity; however, the roles of EVs in both chondrogenic differentiation of MSCs and cartilage regeneration are poorly understood yet. In the current study, to investigate the differentiation effects of human articular chondrocyte EVs on adipose-derived MSCs, they were cultured in injectable chitosan-hyaluronic acid (CS-HA) hydrogel and then treated with chondrocyte EVs for 21 days. The continuous treatment of EVs performed on MSCs increased chondrogenic genes' expressions ofSOX9andCOL2A1and induced expression of Col II protein. In addition, glycosaminoglycans secretion was detected in the EV-treated MSCs after about 14 days. The therapeutic efficiency of this hydrogel and EVs was studied in a rabbit osteochondral defect model. MRI results revealed that the cartilage regeneration capacity of EV-treated MSCs with CS-HA hydrogel was greater than the untreated MSCs or the EV-treated MSCs without hydrogel. Moreover, histological results showed hyaline-like cartilage in the CS-HA/MSC and CS-HA/EV/MSC groups in the cartilage defect sites. These findings suggested that the chondrocyte-EVs and CS-HA hydrogel could provide the preferable niche for chondrogenic differentiation of MSCs and cartilage regeneration in osteoarthritis cartilage injuries.
Collapse
Affiliation(s)
- Asieh Heirani-Tabasi
- Department of Cell Therapy and Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases, Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases, Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Khodamorad Jamshidi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Soufizomorrod
- Department of Cell Therapy and Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Cell Therapy and Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Abstract
The role of stem cells in augmenting reparative processes in the heart after ischemic injury has been successfully demonstrated in small and large animal models. However, the outcomes of cell therapy in clinical trials have been somewhat variable, with overall effects of autologous stem cell therapies demonstrating a modest improvement in cardiac structure and function. How stem cells repair the heart after cardiac injury is still not well understood. Most recent studies suggest that adult derived stem cells act primarily through paracrine signaling to exert beneficial effects, including modulation of immune response, stimulation of new blood vessel formation, or by inducing mature myocytes to transiently reenter the cell cycle, rather than robust direct differentiation of the transplanted cells into myocytes. In addition, data from multiple laboratory results confirmed clearance of stem cells themselves within a few days still leading to functional benefits further confirming the role of paracrine signaling in augmenting cardiac reparative processes rather than direct differentiation of cells. These findings rapidly evolved the field of extracellular vesicles specifically microvesicles (MVs) as they are active hubs of autocrine, paracrine, and endocrine signaling targeting different biological processes. The beneficial effects seen after stem cell transplantation could be linked to the cardioprotective factors packaged in the MVs secreted from stem cells. Therefore, stem cell MVs provide a new avenue for the treatment of cardiovascular disease through a multitude of mechanisms including cellular communication within the stem cell niches, delivery of genetic information, regulation of the immune system in the heart, and stimulation of angiogenesis which will be discussed in this review.
Collapse
|
12
|
Aquino JB, Sierra R, Montaldo LA. Diverse cellular origins of adult blood vascular endothelial cells. Dev Biol 2021; 477:117-132. [PMID: 34048734 DOI: 10.1016/j.ydbio.2021.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
During embryonic stages, vascular endothelial cells (ECs) originate from the mesoderm, at specific extraembryonic and embryonic regions, through a process called vasculogenesis. In the adult, EC renewal/replacement mostly depend on local resident ECs or endothelial progenitor cells (EPCs). Nevertheless, contribution from circulating ECs/EPCs was also reported. In addition, cells lacking from EC/EPC markers with in vitro extended plasticity were shown to originate endothelial-like cells (ELCs). Most of these cells consist of mesenchymal stromal progenitors, which would eventually get mobilized from the bone marrow after injury. Based on that, current knowledge on different mouse and human bone marrow stromal cell (BM-SC) subpopulations, able to contribute with mesenchymal stromal/stem cells (MSCs), is herein reviewed. Such analyses underline an unexpected heterogeneity among sinusoidal LepR+ stromal/CAR cells. For instance, in a recent report a subgroup of LepR+ stromal/CAR progenitors, which express GLAST and is traced in Wnt1Cre;R26RTom mice, was found to contribute with ELCs in vivo. These GLAST + Wnt1+ BM-SCs were shown to get mobilized to the peripheral blood and to contribute with liver regeneration. Other sources of ELCs, such as adipose, neural and dental pulp tissues, were also published. Finally, mechanisms likely involved in the enhanced cellular plasticity properties of bone marrow/adipose tissue stromal cells, able to originate ELCs, are assessed. In the future, strategies to analyze the in vivo expression profile of stromal cells, with MSC properties, in combination with screening of active genomic regions at the single cell-level, during early postnatal development and/or after injury, will likely help understanding properties of these ELC sources.
Collapse
Affiliation(s)
- Jorge B Aquino
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina.
| | - Romina Sierra
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| | - Laura A Montaldo
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| |
Collapse
|
13
|
Youn YJ, Shrestha S, Lee YB, Kim JK, Lee JH, Hur K, Mali NM, Nam SW, Kim SH, Lee S, Song DK, Jin HK, Bae JS, Hong CW. Neutrophil-derived trail is a proinflammatory subtype of neutrophil-derived extracellular vesicles. Am J Cancer Res 2021; 11:2770-2787. [PMID: 33456572 PMCID: PMC7806483 DOI: 10.7150/thno.51756] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/12/2020] [Indexed: 12/26/2022] Open
Abstract
Aims: Extracellular vesicles (EVs) are membrane-derived vesicles that mediate intercellular communications. Neutrophils produce different subtypes of EVs during inflammatory responses. Neutrophil-derived trails (NDTRs) are generated by neutrophils migrating toward inflammatory foci, whereas neutrophil-derived microvesicles (NDMVs) are thought to be generated by neutrophils that have arrived at the inflammatory foci. However, the physical and functional characteristics of neutrophil-derived EVs are incompletely understood. In this study, we aimed to investigate the differences between NDTRs and NDMVs. Methods: The generation of neutrophil-derived EVs were visualized by live-cell fluorescence images and the physical characteristics were further analyzed using nanotracking analysis assay, scanning electron microscopic analysis, and marker expressions. Functional characteristics of neutrophil-derived EVs were analyzed using assays for bactericidal activity, monocyte chemotaxis, phenotype polarization of macrophages, and miRNA sequencing. Finally, the effects of neutrophil-derived EVs on the acute and chronic inflammation were examined in vivo. Results: Both EVs share similar characteristics including stimulators, surface marker expression, bactericidal activity, and chemoattractive effect on monocytes via MCP-1. However, the integrin-mediated physical interaction was required for generation of NDTRs whereas NDMV generation was dependent on PI3K pathway. Interestingly, NDTRs contained proinflammatory miRNAs such as miR-1260, miR-1285, miR-4454, and miR-7975, while NDMVs contained anti-inflammatory miRNAs such as miR-126, miR-150, and miR-451a. Although both EVs were easily uptaken by monocytes, NDTRs enhanced proinflammatory macrophage polarization whereas NDMVs induced anti-inflammatory macrophage polarization. Moreover, NDTRs showed protective effects against lethality in a murine sepsis model and pathological changes in a murine chronic colitis model. Conclusion: These results suggest that NDTR is a proinflammatory subtype of neutrophil-derived EVs distinguished from NDMV.
Collapse
|
14
|
Induced Pluripotent Stem Cells: Hope in the Treatment of Diseases, including Muscular Dystrophies. Int J Mol Sci 2020; 21:ijms21155467. [PMID: 32751747 PMCID: PMC7432218 DOI: 10.3390/ijms21155467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem (iPS) cells are laboratory-produced cells that combine the biological advantages of somatic adult and stem cells for cell-based therapy. The reprogramming of cells, such as fibroblasts, to an embryonic stem cell-like state is done by the ectopic expression of transcription factors responsible for generating embryonic stem cell properties. These primary factors are octamer-binding transcription factor 4 (Oct3/4), sex-determining region Y-box 2 (Sox2), Krüppel-like factor 4 (Klf4), and the proto-oncogene protein homolog of avian myelocytomatosis (c-Myc). The somatic cells can be easily obtained from the patient who will be subjected to cellular therapy and be reprogrammed to acquire the necessary high plasticity of embryonic stem cells. These cells have no ethical limitations involved, as in the case of embryonic stem cells, and display minimal immunological rejection risks after transplant. Currently, several clinical trials are in progress, most of them in phase I or II. Still, some inherent risks, such as chromosomal instability, insertional tumors, and teratoma formation, must be overcome to reach full clinical translation. However, with the clinical trials and extensive basic research studying the biology of these cells, a promising future for human cell-based therapies using iPS cells seems to be increasingly clear and close.
Collapse
|
15
|
He N, Zhang Y, Zhang S, Wang D, Ye H. Exosomes: Cell-Free Therapy for Cardiovascular Diseases. J Cardiovasc Transl Res 2020; 13:713-721. [PMID: 32333198 DOI: 10.1007/s12265-020-09966-7] [Citation(s) in RCA: 25] [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: 11/07/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases (CVDs) are an important cause of death and disease worldwide. Because injured cardiac tissue cannot be repaired itself, it is urgent to develop other alternate therapies. Stem cells can be differentiated into cardiomyocytes, endothelial cells, and vascular smooth muscle cells for the treatment of CVDs. Therefore, cell therapy has recently been considered a viable treatment option that can significantly improve cardiac function. Nonetheless, implanted stem cells rarely survive in the recipient heart, suggesting that the benefits of stem cell therapy may involve other mechanisms. Exosomes derived from stem cells have a myocardial protection function after myocardial injury, and may be a promising and effective therapy for CVDs. Here, we discuss the application and mechanism of exosomes derived from stem cells in the diagnosis and treatment of CVDs and provide evidence for the application of exosomes in CVDs. Graphical Abstract.
Collapse
Affiliation(s)
- Nana He
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Yuelin Zhang
- Department of Medicine, University of Ningbo, Ningbo, China
| | - Shun Zhang
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Dongjuan Wang
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China
| | - Honghua Ye
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
16
|
Mighty J, Zhou J, Benito-Martin A, Sauma S, Hanna S, Onwumere O, Shi C, Muntzel M, Sauane M, Young M, Molina H, Cox D, Redenti S. Analysis of Adult Neural Retina Extracellular Vesicle Release, RNA Transport and Proteomic Cargo. Invest Ophthalmol Vis Sci 2020; 61:30. [PMID: 32084266 PMCID: PMC7326611 DOI: 10.1167/iovs.61.2.30] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose Extracellular vesicles (EVs) contain RNA and protein cargo reflective of the genotype and phenotype of the releasing cell of origin. Adult neural retina EV release, RNA transfer, and proteomic cargo are the focus of this study. Methods Adult wild-type mouse retinae were cultured and released EV diameters and concentrations quantified using Nanosight. Immunogold transmission electron microscopy (TEM) was used to image EV ultrastructure and marker protein localization. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to analyze retinal cell transcripts present in EVs. Super-resolution microscopy was used to image fluorescent (green) RNA and (red) lipid membrane labeled EVs, released by adult retina, and internalized by isolated retinal cells. Mass spectrometry was used to characterize the proteomes of adult retina and EVs. Results Adult neural retina released EVs at a rate of 1.42 +/- 0.08 × 108/mL over 5 days, with diameters ranging from 30 to 910 nm. The canonical EV markers CD63 and Tsg101 localized to retinal EVs. Adult retinal and neuronal mRNA species present in both retina and EVs included rhodopsin and the neuronal nuclei marker NeuN. Fluorescently labeled RNA in retinal cells was enclosed in EVs, transported to, and uptaken by co-cultured adult retinal cells. Proteomic analysis revealed 1696 protein species detected only in retinal cells, 957 species shared between retina and EVs, and 82 detected only in EVs. Conclusions The adult neural retina constitutively releases EVs with molecular cargo capable of intercellular transport and predicted involvement in biological processes including retinal physiology, mRNA processing, and transcription regulation within the retinal microenvironment.
Collapse
|
17
|
Jeske R, Bejoy J, Marzano M, Li Y. Human Pluripotent Stem Cell-Derived Extracellular Vesicles: Characteristics and Applications. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:129-144. [PMID: 31847715 PMCID: PMC7187972 DOI: 10.1089/ten.teb.2019.0252] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are found to play an important role in various biological processes and maintaining tissue homeostasis. Because of the protective effects, stem cell-derived EVs can be used to reduce oxidative stress and apoptosis in the recipient cells. In addition, EVs/exosomes have been used as directional communication tools between stem cells and parenchymal cells, giving them the ability to serve as biomarkers. Likewise, altered EVs/exosomes can be utilized for drug delivery by loading with proteins, small interfering RNAs, and viral vectors, in particular, because EVs/exosomes are able to cross the blood-brain barrier. In this review article, the properties of human induced pluripotent stem cell (iPSC)-derived EVs are discussed. The biogenesis, that is, how EVs originate in the endosomal compartment or from the cell layer of microvesicles, EV composition, the available methods of purification, and characterizations of EVs/exosomes are summarized. In particular, EVs/exosomes derived from iPSCs of different lineage specifications and the applications of these stem cell-derived exosomes in neurological diseases are discussed. Impact statement In this review, we summarized the work related to extracellular vesicles (EVs) derived from human pluripotent stem cells (hPSCs). In particular, EVs/exosomes derived from hPSCs of different lineage specifications and the applications of these stem cell-derived exosomes in neurological diseases are discussed. The results highlight the important role of cell-cell interactions in neural cellular phenotype and neurodegeneration. The findings reported in this article are significant for pluripotent stem cell-derived cell-free products toward applications in stem cell-based therapies.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| |
Collapse
|
18
|
Xie M, Xiong W, She Z, Wen Z, Abdirahman AS, Wan W, Wen C. Immunoregulatory Effects of Stem Cell-Derived Extracellular Vesicles on Immune Cells. Front Immunol 2020; 11:13. [PMID: 32117221 PMCID: PMC7026133 DOI: 10.3389/fimmu.2020.00013] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022] Open
Abstract
Recent investigations on the regulatory action of extracellular vesicles (EVs) on immune cells in vitro and in vivo have sparked interest on the subject. As commonly known, EVs are subcellular components secreted by a paracellular mechanism and are essentially a group of nanoparticles containing exosomes, microvesicles, and apoptotic bodies. They are double-layer membrane-bound vesicles enriched with proteins, nucleic acids, and other active compounds. EVs are recognized as a novel apparatus for intercellular communication that acts through delivery of signal molecules. EVs are secreted by almost all cell types, including stem/progenitor cells. The EVs derived from stem/progenitor cells are analogous to the parental cells and inhibit or enhance immune response. This review aims to provide its readers a comprehensive overview of the possible mechanisms underlying the immunomodulatory effects exerted by stem/progenitor cell-derived EVs upon natural killer (NK) cells, dendritic cells (DCs), monocytes/macrophages, microglia, T cells, and B cells.
Collapse
Affiliation(s)
- Min Xie
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiong
- Department of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, Chengdu, China
| | - Zhou She
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zaichi Wen
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Amin Sheikh Abdirahman
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wuqing Wan
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chuan Wen
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Lee YXF, Johansson H, Wood MJA, El Andaloussi S. Considerations and Implications in the Purification of Extracellular Vesicles - A Cautionary Tale. Front Neurosci 2019; 13:1067. [PMID: 31680809 PMCID: PMC6813730 DOI: 10.3389/fnins.2019.01067] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/24/2019] [Indexed: 12/29/2022] Open
Abstract
Extracellular vesicles (EVs) are nano-sized particles constitutively released from cells into all biological fluids. Interestingly, these vesicles contain genetic cargoes including proteins, RNA and bioactive lipids that can be functionally delivered and affect recipient cells. As a result, there is growing interest in studying EVs in pathological conditions, including central nervous system (CNS)-related diseases, as EVs may be used for diagnostic purposes or as therapeutic agents. However, one major bottleneck is the need for better EV purification strategies when considering complex biological sources such as serum/protein-rich media or plasma. In this study, we have performed a systematic comparison study between the current gold-standard method: ultracentrifugation, to an alternative: size-exclusion chromatography (LC), using induced pluripotent stem cell (iPSC) derived complex media as a model system. We demonstrate that LC allows for derivation of purer EVs from iPSCs, which was previously impossible with the original UC method. Importantly, our study further highlights the various drawbacks when using the conventional UC approach that lead to misinterpretation of EV data. Lastly, we describe novel data on our iPSC-EVs; how they could relate to stem cell biology and discuss their potential use as EV therapeutics for CNS diseases.
Collapse
Affiliation(s)
- Yi Xin Fiona Lee
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Henrik Johansson
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Yao X, Wei W, Wang X, Chenglin L, Björklund M, Ouyang H. Stem cell derived exosomes: microRNA therapy for age-related musculoskeletal disorders. Biomaterials 2019; 224:119492. [PMID: 31557588 DOI: 10.1016/j.biomaterials.2019.119492] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Age-associated musculoskeletal disorders (MSDs) have been historically overlooked by mainstream biopharmaceutical researchers. However, it has now been recognized that stem and progenitor cells confer innate healing capacity for the musculoskeletal system. Current evidence indicates that exosomes are particularly important in this process as they can mediate sequential and reciprocal interactions between cells to initiate and enhance healing. The present review focuses on stem cells (SCs) derived exosomes as a regenerative therapy for treatment of musculoskeletal disorders. We discuss mechanisms involving exosome-mediated transfer of RNAs and how these have been demonstrated in vitro and in vivo to affect signal transduction pathways in target cells. We envision that standardized protocols for stem cell culture as well as for the isolation and characterization of exosomes enable GMP-compliant large-scale production of SCs-derived exosomes. Hence, potential new treatment for age-related degenerative diseases can be seen in the horizon.
Collapse
Affiliation(s)
- Xudong Yao
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Wei
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaozhao Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Chenglin
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mikael Björklund
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, China
| | - Hongwei Ouyang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
21
|
Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids. Cells 2019; 8:cells8090993. [PMID: 31466320 PMCID: PMC6770916 DOI: 10.3390/cells8090993] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) contribute to a variety of signaling processes and the overall physiological and pathological states of stem cells and tissues. Human induced pluripotent stem cells (hiPSCs) have unique characteristics that can mimic embryonic tissue development. There is growing interest in the use of EVs derived from hiPSCs as therapeutics, biomarkers, and drug delivery vehicles. However, little is known about the characteristics of EVs secreted by hiPSCs and paracrine signaling during tissue morphogenesis and lineage specification. Methods: In this study, the physical and biological properties of EVs isolated from hiPSC-derived neural progenitors (ectoderm), hiPSC-derived cardiac cells (mesoderm), and the undifferentiated hiPSCs (healthy iPSK3 and Alzheimer’s-associated SY-UBH lines) were analyzed. Results: Nanoparticle tracking analysis and electron microscopy results indicate that hiPSC-derived EVs have an average size of 100–250 nm. Immunoblot analyses confirmed the enrichment of exosomal markers Alix, CD63, TSG101, and Hsc70 in the purified EV preparations. MicroRNAs including miR-133, miR-155, miR-221, and miR-34a were differently expressed in the EVs isolated from distinct hiPSC lineages. Treatment of cortical spheroids with hiPSC-EVs in vitro resulted in enhanced cell proliferation (indicated by BrdU+ cells) and axonal growth (indicated by β-tubulin III staining). Furthermore, hiPSC-derived EVs exhibited neural protective abilities in Aβ42 oligomer-treated cultures, enhancing cell viability and reducing oxidative stress. Our results demonstrate that the paracrine signaling provided by tissue context-dependent EVs derived from hiPSCs elicit distinct responses to impact the physiological state of cortical spheroids. Overall, this study advances our understanding of cell‒cell communication in the stem cell microenvironment and provides possible therapeutic options for treating neural degeneration.
Collapse
|
22
|
Osier N, Motamedi V, Edwards K, Puccio A, Diaz-Arrastia R, Kenney K, Gill J. Exosomes in Acquired Neurological Disorders: New Insights into Pathophysiology and Treatment. Mol Neurobiol 2018; 55:9280-9293. [PMID: 29663285 DOI: 10.1007/s12035-018-1054-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 03/29/2018] [Indexed: 01/08/2023]
Abstract
Exosomes are endogenous nanovesicles that play critical roles in intercellular signaling by conveying functional genetic information and proteins between cells. Exosomes readily cross the blood-brain barrier and have promise as therapeutic delivery vehicles that have the potential to specifically deliver molecules to the central nervous system (CNS). This unique feature also makes exosomes attractive as biomarkers in diagnostics, prognostics, and therapeutics in the context of multiple significant public health conditions, including acquired neurological disorders. The purpose of this review is to summarize the state of the science surrounding the relevance of extracellular vesicles (EVs), particularly exosomes, to acquire neurological disorders, specifically traumatic brain injury (TBI), spinal cord injury (SCI), and ischemic stroke. In total, ten research articles were identified that examined exosomes in the context of TBI, SCI, or stroke; these manuscripts were reviewed and synthesized to further understand the current role of exosomes in the context of acquired neurological disorders. Of the ten published studies, four focused exclusively on TBI, one on both TBI and SCI, and five on ischemic stroke; notably, eight of the ten studies were limited to pre-clinical samples. The present review is the first to discuss the current body of knowledge surrounding the role of exosomes in the pathophysiology, diagnosis, and prognosis, as well as promising therapeutic strategies in TBI, SCI, and stroke research.
Collapse
Affiliation(s)
- Nicole Osier
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA. .,University of Texas at Austin, Austin, TX, USA.
| | - Vida Motamedi
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA
| | - Katie Edwards
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA.,Healthcare Genetics Doctoral Program, Clemson University School of Nursing, 508 Edwards, Clemson, SC, 29631, USA
| | - Ava Puccio
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite B-400, Pittsburgh, PA, 15213, USA
| | - Ramon Diaz-Arrastia
- University of Pennsylvania School of Medicine, Suite 205 Medical Office Building, 51 N 39TH ST, Philadelphia, PA, 19104, USA
| | - Kimbra Kenney
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Building 51, Room 2306, 4860 South Palmer Road, Bethesda, MD, 20889-5649, USA
| | - Jessica Gill
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA
| |
Collapse
|
23
|
Xin X, Zhang P, Fu X, Mao X, Meng F, Tian M, Zhu X, Sun H, Meng L, Zhou J. Saline is a more appropriate solution for microvesicles for flow cytometric analyses. Oncotarget 2018; 8:34576-34585. [PMID: 28423667 PMCID: PMC5470992 DOI: 10.18632/oncotarget.15987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
Microvesicles (MVs) are carriers of molecular and oncogenic signatures present in subsets of tumor cells and tumor-associated stroma, and a focus of cancer research. Although methods to detect MVs are mature, we were concerned that the buffer used could lead to false results when quantitating MVs by flow cytometry. In this work,we detected MVs by flow cytometry withthree different solutions: water, saline, and phosphate-buffered saline (PBS). The results demonstrated that PBS, when reacted with annexin V binding buffer, produced nano-sized vesicles even when there were no MVs in the sample. No similar events occurred in the saline and water groups (P < 0.01). Annexin V positive rate increased significantly when PBS was used as the buffer, compared to saline and water. These false negative results were also observed when we quantified some markers of MVs such as CD3 and CD19. A probable explanation for these findings is the production of insoluble Ca(H2PO4)2 or Ca3PO4 from calcium in the binding buffer and phosphate in PBS. Thus, considering the osmotic pressure of water, we suggest that saline is a more suitable buffer when counting MVs by flow cytometry.
Collapse
Affiliation(s)
- Xing Xin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Peiling Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Xing Fu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Fankai Meng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Ming Tian
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Hanying Sun
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Li Meng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| |
Collapse
|
24
|
Exosomes: Outlook for Future Cell-Free Cardiovascular Disease Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 998:285-307. [PMID: 28936747 DOI: 10.1007/978-981-10-4397-0_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the number one cause of death globally with an estimated 7.4 million people dying from coronary heart disease. Studies have been conducted to identify the therapeutic utility of exosomes in many diseases, including cardiovascular diseases. It has been demonstrated that exosomes are immune modulators, can be used to treat cardiac ischemic injury, pulmonary hypertension and many other diseases, including cancers. Exosomes can be used as a biomarker for disease and cell-free drug delivery system for targeting the cells. Many studies suggest that exosomes can be used as a cell-free vaccine for many diseases. In this chapter, we explore the possibility of future therapeutic potential of exosomes in various cardiovascular diseases.
Collapse
|
25
|
Zhou J, Benito-Martin A, Mighty J, Chang L, Ghoroghi S, Wu H, Wong M, Guariglia S, Baranov P, Young M, Gharbaran R, Emerson M, Mark MT, Molina H, Canto-Soler MV, Selgas HP, Redenti S. Retinal progenitor cells release extracellular vesicles containing developmental transcription factors, microRNA and membrane proteins. Sci Rep 2018; 8:2823. [PMID: 29434302 PMCID: PMC5809580 DOI: 10.1038/s41598-018-20421-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 01/15/2018] [Indexed: 12/27/2022] Open
Abstract
A range of cell types, including embryonic stem cells, neurons and astrocytes have been shown to release extracellular vesicles (EVs) containing molecular cargo. Across cell types, EVs facilitate transfer of mRNA, microRNA and proteins between cells. Here we describe the release kinetics and content of EVs from mouse retinal progenitor cells (mRPCs). Interestingly, mRPC derived EVs contain mRNA, miRNA and proteins associated with multipotency and retinal development. Transcripts enclosed in mRPC EVs, include the transcription factors Pax6, Hes1, and Sox2, a mitotic chromosome stabilizer Ki67, and the neural intermediate filaments Nestin and GFAP. Proteomic analysis of EV content revealed retinogenic growth factors and morphogen proteins. mRPC EVs were shown to transfer GFP mRNA between cell populations. Finally, analysis of EV mediated functional cargo delivery, using the Cre-loxP recombination system, revealed transfer and uptake of Cre+ EVs, which were then internalized by target mRPCs activating responder loxP GFP expression. In summary, the data supports a paradigm of EV genetic material encapsulation and transfer within RPC populations. RPC EV transfer may influence recipient RPC transcriptional and post-transcriptional regulation, representing a novel mechanism of differentiation and fate determination during retinal development.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA.,Biology Doctoral Program, The Graduate School and University Center, City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Alberto Benito-Martin
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, New York, 10021, USA
| | - Jason Mighty
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA.,Biology Doctoral Program, The Graduate School and University Center, City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Lynne Chang
- Nikon Instruments Inc, 1300 Walt Whitman Road, Melville, NY, 11747, USA
| | - Shima Ghoroghi
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA
| | - Hao Wu
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA.,Biology Doctoral Program, The Graduate School and University Center, City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Madeline Wong
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA
| | - Sara Guariglia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th St, New York, NY, 10032, USA
| | - Petr Baranov
- The Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Michael Young
- The Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Rajendra Gharbaran
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA
| | - Mark Emerson
- Biology Doctoral Program, The Graduate School and University Center, City University of New York, 365 5th Avenue, New York, NY, 10016, USA.,Department of Biology, The City College of New York, City University of New York, New York, NY, 10031, USA
| | - Milica Tesic Mark
- Proteomics Resource Center, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - M Valeria Canto-Soler
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Hector Peinado Selgas
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, New York, 10021, USA.,Microenvironment and Metastasis Laboratory, Department of Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro, 3, Madrid, E28029, Spain
| | - Stephen Redenti
- Department of Biological Sciences, Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY, 10468, USA. .,Biology Doctoral Program, The Graduate School and University Center, City University of New York, 365 5th Avenue, New York, NY, 10016, USA. .,Biochemistry Doctoral Program, The Graduate School and University Center, City University of New York, 365 5th Avenue, New York, NY, 10016, USA.
| |
Collapse
|
26
|
Zamora JLR, Aguilar HC. Flow virometry as a tool to study viruses. Methods 2017; 134-135:87-97. [PMID: 29258922 DOI: 10.1016/j.ymeth.2017.12.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 12/10/2017] [Accepted: 12/14/2017] [Indexed: 01/01/2023] Open
Abstract
In the last few decades, flow cytometry has redefined the field of biology, exponentially enhancing our understanding of cells, immunology, and microbiology. Flow cytometry recently gave birth to flow virometry, a new way to detect, analyze, and characterize single viral particles. Detection of viruses by flow cytometry is possible due to improvements in current flow cytometers, calibration, and tuning methods. We summarize the recent birth and novel uses of flow virometry and the progressive evolution of this tool to advance the field of virology. We also discuss the various flow virometry methods used to identify and analyze viruses. We briefly summarize other applications of flow virometry, including: virus detection, quantification, population discrimination, and viral particles' antigenic properties. Finally, we summarize how viral sorting will allow further progress of flow virometry to relate viral surface characteristics to infectivity properties.
Collapse
Affiliation(s)
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
27
|
Adamiak M, Cheng G, Bobis-Wozowicz S, Zhao L, Kedracka-Krok S, Samanta A, Karnas E, Xuan YT, Skupien-Rabian B, Chen X, Jankowska U, Girgis M, Sekula M, Davani A, Lasota S, Vincent RJ, Sarna M, Newell KL, Wang OL, Dudley N, Madeja Z, Dawn B, Zuba-Surma EK. Induced Pluripotent Stem Cell (iPSC)-Derived Extracellular Vesicles Are Safer and More Effective for Cardiac Repair Than iPSCs. Circ Res 2017; 122:296-309. [PMID: 29118058 DOI: 10.1161/circresaha.117.311769] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 12/21/2022]
Abstract
RATIONALE Extracellular vesicles (EVs) are tiny membrane-enclosed droplets released by cells through membrane budding or exocytosis. The myocardial reparative abilities of EVs derived from induced pluripotent stem cells (iPSCs) have not been directly compared with the source iPSCs. OBJECTIVE To examine whether iPSC-derived EVs can influence the biological functions of cardiac cells in vitro and to compare the safety and efficacy of iPSC-derived EVs (iPSC-EVs) and iPSCs for cardiac repair in vivo. METHODS AND RESULTS Murine iPSCs were generated, and EVs isolated from culture supernatants by sequential centrifugation. Atomic force microscopy, high-resolution flow cytometry, real-time quantitative RT-PCR, and mass spectrometry were used to characterize EV morphology and contents. iPSC-EVs were enriched in miRNAs and proteins with proangiogenic and cytoprotective properties. iPSC-EVs enhanced angiogenic, migratory, and antiapoptotic properties of murine cardiac endothelial cells in vitro. To compare the cardiac reparative capacities in vivo, vehicle, iPSCs, and iPSC-EVs were injected intramyocardially at 48 hours after a reperfused myocardial infarction in mice. Compared with vehicle-injected mice, both iPSC- and iPSC-EV-treated mice exhibited improved left ventricular function at 35 d after myocardial infarction, albeit iPSC-EVs rendered greater improvement. iPSC-EV injection also resulted in reduction in left ventricular mass and superior perfusion in the infarct zone. Both iPSCs and iPSC-EVs preserved viable myocardium in the infarct zone, whereas reduction in apoptosis was significant with iPSC-EVs. iPSC injection resulted in teratoma formation, whereas iPSC-EV injection was safe. CONCLUSIONS iPSC-derived EVs impart cytoprotective properties to cardiac cells in vitro and induce superior cardiac repair in vivo with regard to left ventricular function, vascularization, and amelioration of apoptosis and hypertrophy. Because of their acellular nature, iPSC-EVs represent a safer alternative for potential therapeutic applications in patients with ischemic myocardial damage.
Collapse
Affiliation(s)
- Marta Adamiak
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Guangming Cheng
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Sylwia Bobis-Wozowicz
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Lin Zhao
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Sylwia Kedracka-Krok
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Anweshan Samanta
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Elzbieta Karnas
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Yu-Ting Xuan
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Bozena Skupien-Rabian
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Xing Chen
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Urszula Jankowska
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Magdy Girgis
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Malgorzata Sekula
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Arash Davani
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Slawomir Lasota
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Robert J Vincent
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Michal Sarna
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Kathy L Newell
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Ou-Li Wang
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Nathaniel Dudley
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Zbigniew Madeja
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Buddhadeb Dawn
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna).
| | - Ewa K Zuba-Surma
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna).
| |
Collapse
|
28
|
Kondratov KA, Petrova TA, Mikhailovskii VY, Ivanova AN, Kostareva AA, Fedorov AV. A study of extracellular vesicles isolated from blood plasma conducted by low-voltage scanning electron microscopy. ACTA ACUST UNITED AC 2017. [DOI: 10.1134/s1990519x17030051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Jung JH, Fu X, Yang PC. Exosomes Generated From iPSC-Derivatives: New Direction for Stem Cell Therapy in Human Heart Diseases. Circ Res 2017; 120:407-417. [PMID: 28104773 PMCID: PMC5260934 DOI: 10.1161/circresaha.116.309307] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in modern society. The adult heart innately lacks the capacity to repair and regenerate the damaged myocardium from ischemic injury. Limited understanding of cardiac tissue repair process hampers the development of effective therapeutic solutions to treat CVD such as ischemic cardiomyopathy. In recent years, rapid emergence of induced pluripotent stem cells (iPSC) and iPSC-derived cardiomyocytes presents a valuable opportunity to replenish the functional cells to the heart. The therapeutic effects of iPSC-derived cells have been investigated in many preclinical studies. However, the underlying mechanisms of iPSC-derived cell therapy are still unclear, and limited engraftment of iPSC-derived cardiomyocytes is well known. One facet of their mechanism is the paracrine effect of the transplanted cells. Microvesicles such as exosomes secreted from the iPSC-derived cardiomyocytes exert protective effects by transferring the endogenous molecules to salvage the injured neighboring cells by regulating apoptosis, inflammation, fibrosis, and angiogenesis. In this review, we will focus on the current advances in the exosomes from iPSC derivatives and discuss their therapeutic potential in the treatment of CVD.
Collapse
Affiliation(s)
- Ji-Hye Jung
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA
| | - Xuebin Fu
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA
| | - Phillip C Yang
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA.
| |
Collapse
|
30
|
Herpes Simplex Virus Capsid Localization to ESCRT-VPS4 Complexes in the Presence and Absence of the Large Tegument Protein UL36p. J Virol 2016; 90:7257-7267. [PMID: 27252536 PMCID: PMC4984650 DOI: 10.1128/jvi.00857-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/24/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED UL36p (VP1/2) is the largest protein encoded by herpes simplex virus 1 (HSV-1) and resides in the innermost layer of tegument, the complex protein layer between the capsid and envelope. UL36p performs multiple functions in the HSV life cycle, including a critical but unknown role in capsid cytoplasmic envelopment. We tested whether UL36p is essential for envelopment because it is required to engage capsids with the cellular ESCRT/Vps4 apparatus. A green fluorescent protein (GFP)-fused form of the dominant negative ATPase Vps4-EQ was used to irreversibly tag ESCRT envelopment sites during infection by UL36p-expressing and UL36-null HSV strains. Using fluorescence microscopy and scanning electron microscopy, we quantitated capsid/Vps4-EQ colocalization and examined the ultrastructure of the corresponding viral assembly intermediates. We found that loss of UL36p resulted in a two-thirds reduction in the efficiency of capsid/Vps4-EQ association but that the remaining UL36p-null capsids were still able to engage the ESCRT envelopment apparatus. It appears that although UL36p helps to couple HSV capsids to the ESCRT pathway, this is likely not the sole reason for its absolute requirement for envelopment. IMPORTANCE Envelopment of the HSV capsid is essential for the assembly of an infectious virion and requires the complex interplay of a large number of viral and cellular proteins. Critical to envelope assembly is the virally encoded protein UL36p, whose function is unknown. Here we test the hypothesis that UL36p is essential for the recruitment of cellular ESCRT complexes, which are also known to be required for envelopment.
Collapse
|
31
|
Potential Therapies by Stem Cell-Derived Exosomes in CNS Diseases: Focusing on the Neurogenic Niche. Stem Cells Int 2016; 2016:5736059. [PMID: 27195011 PMCID: PMC4853949 DOI: 10.1155/2016/5736059] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/27/2016] [Indexed: 12/31/2022] Open
Abstract
Neurodegenerative disorders are one of the leading causes of death and disability and one of the biggest burdens on health care systems. Novel approaches using various types of stem cells have been proposed to treat common neurodegenerative disorders such as Alzheimer's Disease, Parkinson's Disease, or stroke. Moreover, as the secretome of these cells appears to be of greater benefit compared to the cells themselves, the extracellular components responsible for its therapeutic benefit have been explored. Stem cells, as well as most cells, release extracellular vesicles such as exosomes, which are nanovesicles able to target specific cell types and thus to modify their function by delivering proteins, lipids, and nucleic acids. Exosomes have recently been tested in vivo and in vitro as therapeutic conveyors for the treatment of diseases. As such, they could be engineered to target specific populations of cells within the CNS. Considering the fact that many degenerative brain diseases have an impact on adult neurogenesis, we discuss how the modulation of the adult neurogenic niches may be a therapeutic target of stem cell-derived exosomes. These novel approaches should be examined in cellular and animal models to provide better, more effective, and specific therapeutic tools in the future.
Collapse
|