101
|
Adamo A, Dal Collo G, Bazzoni R, Krampera M. Role of mesenchymal stromal cell-derived extracellular vesicles in tumour microenvironment. Biochim Biophys Acta Rev Cancer 2018; 1871:192-198. [PMID: 30599188 DOI: 10.1016/j.bbcan.2018.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/22/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Stromal cells, deriving from mesenchymal stromal cells (MSCs), are crucial component of tumour microenvironment and represent key regulators of tumour processes. MSCs can be recruited to the tumour environment and interact with many cellular elements, thus influencing tumour biology. Cell-to-cell communication is in part mediated by the release of extracellular vesicle (EVs). EVs can induce significant molecular changes in recipient cells, delivering bioactive molecules. In this review, we describe the MSC-derived EVs content and discuss their role in different processes related to cancer biology. Furthermore, we summarize chemical or biological EVs modifications aiming to develop more efficient antitumor therapies.
Collapse
Affiliation(s)
- A Adamo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - G Dal Collo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - R Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - M Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
102
|
Kornicka K, Houston J, Marycz K. Dysfunction of Mesenchymal Stem Cells Isolated from Metabolic Syndrome and Type 2 Diabetic Patients as Result of Oxidative Stress and Autophagy may Limit Their Potential Therapeutic Use. Stem Cell Rev Rep 2018; 14:337-345. [PMID: 29611042 PMCID: PMC5960487 DOI: 10.1007/s12015-018-9809-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSC) have become a promising tool for therapeutic intervention. Their unique features, including self-renewal, multipotency and immunomodulatory properties draw the worldwide attention of researchers and physicians with respect to their application in disease treatment. However, the environment (so-called niche) from which MSCs are isolated may determine their usefulness. Many studies indicated the involvement of MSCs in ageing and disease. In this review, we have focused on how type 2 diabetes (T2D) and metabolic syndrome (MS) affect MSC properties, and thus limit their therapeutic potential. Herein, we mainly focus on apoptosis, autophagy and mitochondria deterioration processes that indirectly affect MSC fate. Based on the data presented, special attention should be paid when considering autologous MSC therapy in T2D or MS treatments, as their therapeutic potential may be restricted.
Collapse
Affiliation(s)
- Katarzyna Kornicka
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland.
| | - Jenny Houston
- PferdePraxis Dr. Med. Vet. Daniel Weiss, Postmatte 14, CH-8807, Freienbach, Switzerland
| | - Krzysztof Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland.,Wroclaw Research Centre EIT+, 54-066, Wroclaw, Poland
| |
Collapse
|
103
|
Lopatina T, Grange C, Fonsato V, Tapparo M, Brossa A, Fallo S, Pitino A, Herrera-Sanchez MB, Kholia S, Camussi G, Bussolati B. Extracellular vesicles from human liver stem cells inhibit tumor angiogenesis. Int J Cancer 2018; 144:322-333. [PMID: 30110127 DOI: 10.1002/ijc.31796] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 12/30/2022]
Abstract
Human liver stem-like cells (HLSC) and derived extracellular vesicles (EVs) were previously shown to exhibit anti-tumor activity. In our study, we investigated whether HLSC-derived EVs (HLSC-EVs) were able to inhibit tumor angiogenesis in vitro and in vivo, in comparison with EVs derived from mesenchymal stem cells (MSC-EVs). The results obtained indicated that HLSC-EVs, but not MSC-EVs, inhibited the angiogenic properties of tumor-derived endothelial cells (TEC) both in vitro and in vivo in a model of subcutaneous implantation in Matrigel. Treatment of TEC with HLSC-EVs led to the down-regulation of pro-angiogenic genes. Since HLSC-EVs carry a specific set of microRNAs (miRNAs) that could target these genes, we investigated their potential role by transfecting TEC with HLSC-EV specific miRNAs. We observed that four miRNAs, namely miR-15a, miR-181b, miR-320c and miR-874, significantly inhibited the angiogenic properties of TEC in vitro, and decreased the expression of some predicted target genes (ITGB3, FGF1, EPHB4 and PLAU). In parallel, TEC treated with HLSC-EVs significantly enhanced expression of miR-15a, miR-181b, miR-320c and miR-874 associated with the down-regulation of FGF1 and PLAU. In summary, HLSC-EVs possess an anti-tumorigenic effect, based on their ability to inhibit tumor angiogenesis.
Collapse
Affiliation(s)
- Tatiana Lopatina
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Valentina Fonsato
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl, University of Turin, Turin, Italy
| | - Marta Tapparo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessia Brossa
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Sofia Fallo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Adriana Pitino
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl, University of Turin, Turin, Italy
| | - Maria Beatriz Herrera-Sanchez
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl, University of Turin, Turin, Italy
| | - Sharad Kholia
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
104
|
Toh WS, Zhang B, Lai RC, Lim SK. Immune regulatory targets of mesenchymal stromal cell exosomes/small extracellular vesicles in tissue regeneration. Cytotherapy 2018; 20:1419-1426. [PMID: 30352735 DOI: 10.1016/j.jcyt.2018.09.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cell (MSC) therapies have demonstrated therapeutic efficacy in a wide-ranging array of tissue injury and disease indications. An important aspect of MSC-mediated therapeutic activities is immune modulation. Consistent with the concentration of MSC therapeutic potency in its secretion, a significant proportion of MSC immune potency resides in the small extracellular vesicles (sEVs) secreted by MSCs. These sEVs, which also include exosomes, carry a large cargo enriched in proteins with potent immunomodulatory activities. They have been reported to exert potent effects on humoral and cellular components of the immune system in vitro and in vivo, and may have the potential to support the diametrically opposite pro- and anti-inflammatory functions necessary for tissue repair and regeneration following injury. Following injury, pro-inflammatory activities are necessary to neutralize injury and remove dead or injured tissue, while anti-inflammatory activities to facilitate migration and proliferation of reparative cell types and to increase vascularization and nutrient supply are necessary to repair and regenerate new tissue. Therefore, a critical immunomodulatory requisite of MSC sEVs in tissue regeneration is the capacity to support the appropriate immune activities at the appropriate time. Here, we review how some of the immune regulatory targets of MSC sEVs could support the dynamic immunomodulatory activities during tissue repair and regeneration.
Collapse
Affiliation(s)
- Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore; Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - Bin Zhang
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Ruenn Chai Lai
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
105
|
WANG WENTING, LI ZIJIAN, FENG JUAN. The potential role of exosomes in the diagnosis and therapy of ischemic diseases. Cytotherapy 2018; 20:1204-1219. [DOI: 10.1016/j.jcyt.2018.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/14/2018] [Accepted: 06/22/2018] [Indexed: 12/13/2022]
|
106
|
Reza-Zaldivar EE, Hernández-Sapiéns MA, Minjarez B, Gutiérrez-Mercado YK, Márquez-Aguirre AL, Canales-Aguirre AA. Potential Effects of MSC-Derived Exosomes in Neuroplasticity in Alzheimer's Disease. Front Cell Neurosci 2018; 12:317. [PMID: 30319358 PMCID: PMC6165870 DOI: 10.3389/fncel.2018.00317] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia affecting regions of the central nervous system that exhibit synaptic plasticity and are involved in higher brain functions such as learning and memory. AD is characterized by progressive cognitive dysfunction, memory loss and behavioral disturbances of synaptic plasticity and energy metabolism. Cell therapy has emerged as an alternative treatment of AD. The use of adult stem cells, such as neural stem cells and Mesenchymal Stem Cells (MSCs) from bone marrow and adipose tissue, have the potential to decrease cognitive deficits, possibly by reducing neuronal loss through blocking apoptosis, increasing neurogenesis, synaptogenesis and angiogenesis. These processes are mediated primarily by the secretion of many growth factors, anti-inflammatory proteins, membrane receptors, microRNAs (miRNA) and exosomes. Exosomes encapsulate and transfer several functional molecules like proteins, lipids and regulatory RNA which can modify cell metabolism. In the proteomic characterization of the content of MSC-derived exosomes, more than 730 proteins have been identified, some of which are specific cell type markers and others are involved in the regulation of binding and fusion of exosomes with adjacent cells. Furthermore, some factors were found that promote the recruitment, proliferation and differentiation of other cells like neural stem cells. Moreover, within exosomal cargo, a wide range of miRNAs were found, which can control functions related to neural remodeling as well as angiogenic and neurogenic processes. Taking this into consideration, the use of exosomes could be part of a strategy to promote neuroplasticity, improve cognitive impairment and neural replacement in AD. In this review, we describe how exosomes are involved in AD pathology and discuss the therapeutic potential of MSC-derived exosomes mediated by miRNA and protein cargo.
Collapse
Affiliation(s)
- Edwin E Reza-Zaldivar
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Mercedes A Hernández-Sapiéns
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Benito Minjarez
- Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| | - Yanet K Gutiérrez-Mercado
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Ana L Márquez-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Alejandro A Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico.,Profesor del programa de Maestría en Ciencias de la Salud Ambiental, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
107
|
Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine - a new paradigm for tissue repair. Biomater Sci 2018; 6:60-78. [PMID: 29184934 DOI: 10.1039/c7bm00479f] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue regeneration by stem cells is driven by the paracrine activity of shedding vesicles and exosomes, which deliver specific cargoes to the recipient cells. Proteins, RNA, cytokines and subsequent gene expression, orchestrate the regeneration process by improving the microenvironment to promote cell survival, controlling inflammation, repairing injury and enhancing the healing process. The action of microRNA is widely accepted as an essential driver of the regenerative process through its impact on multiple downstream biological pathways, and its ability to regulate the host immune response. Here, we present an overview of the recent potential uses of exosomes for regenerative medicine and tissue engineering. We also highlight the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal microRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.
Collapse
Affiliation(s)
- I M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | | | | | | | | |
Collapse
|
108
|
Abbasian N, Herbert KE, Pawluczyk I, Burton JO, Bevington A. Vesicles bearing gifts: the functional importance of micro-RNA transfer in extracellular vesicles in chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1430-F1443. [PMID: 30110570 DOI: 10.1152/ajprenal.00318.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including microparticles (MPs) and exosomes (EXOs), are derived from a wide range of mammalian cells including blood platelets, endothelial cells, and kidney cells and can be detected in body fluids including blood and urine. While EVs are well established as diagnostic markers under pathophysiological and stress conditions, there is also mounting evidence of their functional significance as vehicles for communication between cells mediated by the presence of nucleic acids, especially microRNAs (miRs), encapsulated in the EVs. miRs regulate gene expression, are transported both in MPs and EXOs, and exert profound effects in the kidney. Here we review current understanding of the links between EVs and miRs, discuss the importance of miRs in kidney disease, and shed light on the role of EVs in transferring miRs through the circulation among the renal, vascular, and inflammatory cell populations that are functionally important in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, and Leicester National Institute of Health Research Cardiovascular Biomedical Research Unit , Leicester , United Kingdom
| | - Izabella Pawluczyk
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - James O Burton
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom.,John Walls Renal Unit, University Hospitals of Leicester , Leicester , United Kingdom
| | - Alan Bevington
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
109
|
Perez-Hernandez J, Olivares D, Forner MJ, Ortega A, Solaz E, Martinez F, Chaves FJ, Redon J, Cortes R. Urinary exosome miR-146a is a potential marker of albuminuria in essential hypertension. J Transl Med 2018; 16:228. [PMID: 30107841 PMCID: PMC6092786 DOI: 10.1186/s12967-018-1604-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/10/2018] [Indexed: 01/21/2023] Open
Abstract
Background There is increasing interest in using extracellular vesicle-derived microRNAs (miRNAs) as biomarkers in renal dysfunction and injury. Preliminary evidence indicates that miRNAs regulate the progression of glomerular disease. Indeed, exosomes from the renal system have provided novel evidence in the clinical setting of albuminuria. Thus, the aim of this study was to quantify the urinary miRNAs present in exosome and microvesicles (MVs), and to assess their association with the presence of increased urinary albumin excretion in essential hypertension. Methods Exosomes were collected from urine specimens from a cohort of hypertensive patients with (n = 24) or without albuminuria (n = 28), and from 20 healthy volunteers as a control group. Urinary exosomes were phenotyped by Western blot, tunable resistive pulse sensing, and electronic microscopy. Expression of miR-146a and miR-335* was analysed by qRT-PCR and any associations between albuminuria and exosomal miRNAs were analysed. Results Urinary miRNAs are highly enriched in exosome subpopulations compared to MVs, both in patients with or without increased albuminuria (p < 0.001), but not in the control group. High albuminuria was associated with 2.5-fold less miR-146a in exosomes (p = 0.017), whereas miR-146a levels in MV did not change. In addition, exosome miR-146a levels were inversely associated with albuminuria (r = 0.65, p < 0.0001), and discriminated the presence of urinary albumin excretion presence [area under the curve = 0.80, 95% confidence interval: 0.66–0.95; p = 0.0013]. Conclusions Our results indicate that miRNAs were enriched in the urinary exosome subpopulation in hypertensive patients and that low miR-146a expression in exosomes was associated with the presence of albuminuria. Thus, urinary exosome miR-146a may be a potentially useful tool for studying early renal injury in hypertension.
Collapse
Affiliation(s)
- Javier Perez-Hernandez
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain.,Genomic and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Dolores Olivares
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain.,Genomic and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Maria J Forner
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain.,Internal Medicine Unit, Hospital Clínico Universitario, Valencia, Spain
| | - Ana Ortega
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain
| | - Elena Solaz
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain.,Internal Medicine Unit, Hospital Clínico Universitario, Valencia, Spain
| | - Fernando Martinez
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain.,Internal Medicine Unit, Hospital Clínico Universitario, Valencia, Spain
| | - Felipe J Chaves
- Genomic and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Valencia, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Institute of Health Carlos III, Minister of Health, Barcelona, Spain
| | - Josep Redon
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain.,Internal Medicine Unit, Hospital Clínico Universitario, Valencia, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Minister of Health, Madrid, Spain
| | - Raquel Cortes
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, Avd. Menendez Pelayo, accesorio 4, 46010, Valencia, Spain. .,Genomic and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Valencia, Spain.
| |
Collapse
|
110
|
MSC exosome works through a protein-based mechanism of action. Biochem Soc Trans 2018; 46:843-853. [PMID: 29986939 PMCID: PMC6103455 DOI: 10.1042/bst20180079] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cell (MSC) exosome specifically defines the 50–200 nm vesicles that are secreted into the extracellular space when multivesicular bodies in the MSC fuse with the plasma membrane. However, the exosome is just one of several 50–200 nm extracellular vesicles (EVs) known to be secreted by cells. Nevertheless, the term ‘MSC exosome’ is often used to describe populations of 50–200 nm EVs that are prepared from culture medium conditioned by MSCs on the basis that these populations collectively exhibited typical exosome-associated proteins such as endosomal proteins, TSG101 and Alix, and tetraspanin proteins, CD9, CD63 and CD81. They also carry a rich diverse RNA cargo. MSC exosomes are increasingly implicated as the mediator of many of the MSC-associated therapeutic potencies. They elicit therapeutic activity by delivering their cargo of potentially therapeutic proteins and RNAs to the recipient cells. The therapeutic potency of MSC exosomes is usually rationalized on the presence of a biologically relevant protein or RNA in the MSC exosome. In the present paper, we expanded this rationale beyond a physical presence to include biologically relevant concentration, biochemical functionality and the potential to elicit an appropriate timely biochemical response. Based on these, we propose that MSC exosomes most probably work through the protein rather than the RNA.
Collapse
|
111
|
Jing H, He X, Zheng J. Exosomes and regenerative medicine: state of the art and perspectives. Transl Res 2018; 196:1-16. [PMID: 29432720 DOI: 10.1016/j.trsl.2018.01.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/19/2022]
Abstract
Exosomes have attracted the attention of the scientific community in recent years due to their widespread distribution, their possible functions as biomarkers of disease, and their great potential to be applied as therapeutic agents. Exosomes carry proteins and nucleic acids that can facilitate their uptake by distant target cells through endocytosis, such that exosomes could be targeted to a specific cell or cells to enhance or interfere with specific biological processes. This review will mainly focus on their roles in tissue repair and regenerative processes. Exosomal engineering and their potential applications in tissue regeneration are also reviewed here as an outlook for future research.
Collapse
Affiliation(s)
- Hui Jing
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jinghao Zheng
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
112
|
Suppression of Elp2 prevents renal fibrosis and inflammation induced by unilateral ureter obstruction (UUO) via inactivating Stat3-regulated TGF-β1 and NF-κB pathways. Biochem Biophys Res Commun 2018; 501:400-407. [DOI: 10.1016/j.bbrc.2018.04.227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/29/2018] [Indexed: 01/02/2023]
|
113
|
Exosomes in Myocardial Repair: Advances and Challenges in the Development of Next-Generation Therapeutics. Mol Ther 2018; 26:1635-1643. [PMID: 29807783 DOI: 10.1016/j.ymthe.2018.04.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 01/08/2023] Open
Abstract
Myocardial disease is a leading cause of morbidity and mortality worldwide. Given the limited regenerative capacity of the human heart following myocardial injury, stem cell-based therapies have emerged as a promising approach for improving cardiac repair and function. The discovery of extracellular vesicles including exosomes as a key component of the beneficial function of stem cells has generated hope for their use to advance cell-based regenerative therapies for cardiac repair. Exosomes secreted from stem cells are membranous bionanovesicles, naturally loaded with various proteins, lipids, and nucleic acids. They have been found to have anti-apoptotic, anti-fibrotic, as well as pro-angiogenic effects, all of which are crucial to restore function of the damaged myocardium. In this brief review, we will focus on the latest research and debates on cardiac repair and regenerative potential of exosomes from a variety of sources such as cardiac and non-cardiac stem and progenitor cells, somatic cells, and body fluids. We will also highlight important barriers involved in translating these findings into developing clinically suitable exosome-based therapies.
Collapse
|
114
|
Andrade L, Rodrigues CE, Gomes SA, Noronha IL. Acute Kidney Injury as a Condition of Renal Senescence. Cell Transplant 2018; 27:739-753. [PMID: 29701108 PMCID: PMC6047270 DOI: 10.1177/0963689717743512] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Acute kidney injury (AKI), characterized by a sharp drop in glomerular filtration, continues to be a significant health burden because it is associated with high initial mortality, morbidity, and substantial health-care costs. There is a strong connection between AKI and mechanisms of senescence activation. After ischemic or nephrotoxic insults, a wide range of pathophysiological events occur. Renal tubular cell injury is characterized by cell membrane damage, cytoskeleton disruption, and DNA degradation, leading to tubular cell death by necrosis and apoptosis. The senescence mechanism involves interstitial fibrosis, tubular atrophy, and capillary rarefaction, all of which impede the morphological and functional recovery of the kidneys, suggesting a strong link between AKI and the progression of chronic kidney disease. During abnormal kidney repair, tubular epithelial cells can assume a senescence-like phenotype. Cellular senescence can occur as a result of cell cycle arrest due to increased expression of cyclin kinase inhibitors (mainly p21), downregulation of Klotho expression, and telomere shortening. In AKI, cellular senescence is aggravated by other factors including oxidative stress and autophagy. Given this scenario, the main question is whether AKI can be repaired and how to avoid the senescence process. Stem cells might constitute a new therapeutic approach. Mesenchymal stem cells (MSCs) can ameliorate kidney injury through angiogenesis, immunomodulation, and fibrosis pathway blockade, as well as through antiapoptotic and promitotic processes. Young umbilical cord–derived MSCs are better at increasing Klotho levels, and thus protecting tissues from senescence, than are adipose-derived MSCs. Umbilical cord–derived MSCs improve glomerular filtration and tubular function to a greater degree than do those obtained from adult tissue. Although senescence-related proteins and microRNA are upregulated in AKI, they can be downregulated by treatment with umbilical cord–derived MSCs. In summary, stem cells derived from young tissues, such as umbilical cord–derived MSCs, could slow the post-AKI senescence process.
Collapse
Affiliation(s)
- Lucia Andrade
- 1 Laboratory of Basic Science LIM-12, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Camila E Rodrigues
- 1 Laboratory of Basic Science LIM-12, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Samirah A Gomes
- 2 Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Irene L Noronha
- 2 Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| |
Collapse
|
115
|
Cappariello A, Loftus A, Muraca M, Maurizi A, Rucci N, Teti A. Osteoblast-Derived Extracellular Vesicles Are Biological Tools for the Delivery of Active Molecules to Bone. J Bone Miner Res 2018; 33:517-533. [PMID: 29091316 DOI: 10.1002/jbmr.3332] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/21/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
Abstract
Extracellular vesicles (EVs) are newly appreciated regulators of tissue homeostasis and a means of intercellular communication. Reports have investigated the role of EVs and their cargoes in cellular regulation and have tried to fine-tune their biotechnological use, but to date very little is known on their function in bone biology. To investigate the relevance of EV-mediated communication between bone cells, we isolated EVs from primary mouse osteoblasts and assessed membrane integrity, size, and structure by transmission electron microscopy (TEM) and fluorescence-activated cell sorting (FACS). EVs actively shuttled loaded fluorochromes to osteoblasts, monocytes, and endothelial cells. Moreover, osteoblast EVs contained mRNAs shared with donor cells. Osteoblasts are known to regulate osteoclastogenesis, osteoclast survival, and osteoclast function by the pro-osteoclastic cytokine, receptor activator of nuclear factor κ-B ligand (Rankl). Osteoblast EVs were enriched in Rankl, which increased after PTH treatment. These EVs were biologically active, supporting osteoclast survival. EVs isolated from rankl-/- osteoblasts lost this pro-osteoclastic function, indicating its Rankl-dependence. They integrated ex vivo into murine calvariae, and EV-shuttled fluorochromes were quickly taken up by the bone upon in vivo EV systemic administration. Rankl-/- mice lack the osteoclast lineage and are negative for its specific marker tartrate-resistant acid phosphatase (TRAcP). Treatment of rankl-/- mice with wild-type osteoblast EVs induced the appearance of TRAcP-positive cells in an EV density-dependent manner. Finally, osteoblast EVs internalized and shuttled anti-osteoclast drugs (zoledronate and dasatinib), inhibiting osteoclast activity in vitro and in vivo. We conclude that osteoblast EVs are involved in intercellular communication between bone cells, contribute to the Rankl pro-osteoclastic effect, and shuttle anti-osteoclast drugs, representing a potential means of targeted therapeutic delivery. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alexander Loftus
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
116
|
Manfredi AA, Ramirez GA, Rovere-Querini P, Maugeri N. The Neutrophil's Choice: Phagocytose vs Make Neutrophil Extracellular Traps. Front Immunol 2018. [PMID: 29515586 PMCID: PMC5826238 DOI: 10.3389/fimmu.2018.00288] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophils recognize particulate substrates of microbial or endogenous origin and react by sequestering the cargo via phagocytosis or by releasing neutrophil extracellular traps (NETs) outside the cell, thus modifying and alerting the environment and bystander leukocytes. The signals that determine the choice between phagocytosis and the generation of NETs are still poorly characterized. Neutrophils that had phagocytosed bulky particulate substrates, such as apoptotic cells and activated platelets, appear to be “poised” in an unresponsive state. Environmental conditions, the metabolic, adhesive and activation state of the phagocyte, and the size of and signals associated with the tethered phagocytic cargo influence the choice of the neutrophils, prompting either phagocytic clearance or the generation of NETs. The choice is dichotomic and apparently irreversible. Defects in phagocytosis may foster the intravascular generation of NETs, thus promoting vascular inflammation and morbidities associated with diseases characterized by defective phagocytic clearance, such as systemic lupus erythematosus. There is a strong potential for novel treatments based on new knowledge of the events determining the inflammatory and pro-thrombotic function of inflammatory leukocytes.
Collapse
Affiliation(s)
- Angelo A Manfredi
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Giuseppe A Ramirez
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Norma Maugeri
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
117
|
Lässer C, Jang SC, Lötvall J. Subpopulations of extracellular vesicles and their therapeutic potential. Mol Aspects Med 2018; 60:1-14. [PMID: 29432782 DOI: 10.1016/j.mam.2018.02.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/17/2018] [Accepted: 02/06/2018] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs), such as exosomes and microvesicles, have over the last 10-15 years been recognized to convey key messages in the molecular communication between cells. Indeed, EVs have the capacity to shuttle proteins, lipids, and nucleotides such as RNA between cells, leading to an array of functional changes in the recipient cells. Importantly, the EV secretome changes significantly in diseased cells and under conditions of cellular stress. More recently, it has become evident that the EV secretome is exceptionally diverse, with many different types of EVs being released by a single cell type, and these EVs can be described in terms of differences in density, molecular cargos, and morphology. This review will discuss the diversity of EVs, will introduce some suggestions for how to categorize them, and will propose how EVs and their subpopulations might be used for very different therapeutic purposes.
Collapse
Affiliation(s)
- Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Su Chul Jang
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden; Codiak BioSciences, Cambridge, MA 02139, USA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
118
|
IL-3R-alpha blockade inhibits tumor endothelial cell-derived extracellular vesicle (EV)-mediated vessel formation by targeting the β-catenin pathway. Oncogene 2017; 37:1175-1191. [PMID: 29238040 PMCID: PMC5861089 DOI: 10.1038/s41388-017-0034-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/18/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022]
Abstract
The proangiogenic cytokine Interleukin-3 (IL-3) is released by inflammatory cells in breast and ovarian cancer tissue microenvironments and also acts as an autocrine factor for human breast and kidney tumor-derived endothelial cells (TECs). We have previously shown that IL-3-treated endothelial cells (ECs) release extracellular vesicles (EVs), which serve as a paracrine mechanism for neighboring ECs, by transferring active molecules. The impact of an anti-IL-3R-alpha blocking antibody on the proangiogenic effect of EVs released from TECs (anti-IL-3R-EVs) has therefore been investigated in this study. We have found that anti-IL-3R-EV treatment prevented neovessel formation and, more importantly, also induced the regression of in vivo TEC-derived neovessels. Two miRs that target the canonical wingless (Wnt)/β-catenin pathway, at different levels, were found to be differentially regulated when comparing the miR-cargo of naive TEC-derived EVs (EVs) and anti-IL-3R-EVs. miR-214-3p, which directly targets β-catenin, was found to be upregulated, whereas miR-24-3p, which targets adenomatous polyposis coli (APC) and glycogen synthase kinase-3β (GSK3β), was found to be downregulated. In fact, upon their transfer into the cell, low β-catenin content and high levels of the two members of the “β-catenin destruction complex” were detected. Moreover, c-myc downregulation was found in TECs treated with anti-IL-3R-EVs, pre-miR-214-3p-EVs and antago-miR-24-3p-EVs, which is consistent with network analyses of miR-214-3p and miR-24-3p gene targeting. Finally, in vivo studies have demonstrated the impaired growth of vessels in pre-miR-214-3p-EV- and antago-miR-24-3p-EV-treated animals. These effects became much more evident when combo treatment was applied. The results of the present study identify the canonical Wnt/β-catenin pathway as a relevant mechanism of TEC-derived EV proangiogenic action. Furthermore, we herein provide evidence that IL-3R blockade may yield some significant advantages, than miR targeting, in inhibiting the proangiogenic effects of naive TEC-derived EVs by changing TEC-EV-miR cargo.
Collapse
|
119
|
Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis 2017; 8:3200. [PMID: 29233979 PMCID: PMC5870585 DOI: 10.1038/s41419-017-0041-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Renal ischemia-reperfusion is a main cause of acute kidney injury (AKI), which is associated with high mortality. Here we show that extracellular vesicles (EVs) secreted from hiPSC-MSCs play a critical role in protection against renal I/R injury. hiPSC-MSCs-EVs can fuse with renal cells and deliver SP1 into target cells, subsequently active SK1 expression and increase S1P formation. Chromatin immunoprecipitation (ChIP) analyses and luciferase assay were used to confirm SP1 binds directly to the SK1 promoter region and promote promoter activity. Moreover, SP1 inhibition (MIT) or SK1 inhibition (SKI-II) completely abolished the renal protective effect of hiPSC-MSCs-EVs in rat I/R injury mode. However, pre-treatment of necroptosis inhibitor Nec-1 showed no difference with the administration of hiPSC-MSCs-EVs only. We then generated an SP1 knockout hiPSC-MSC cell line by CRISPR/Cas9 system and found that SP1 knockout failed to show the protective effect of hiPSC-MSCs-EVs unless restoring the level of SP1 by Ad-SP1 in vitro and in vivo. In conclusion, this study describes an anti-necroptosis effect of hiPSC-MSCs-EVs against renal I/R injury via delivering SP1 into target renal cells and intracellular activating the expression of SK1 and the generation of S1P. These findings suggest a novel mechanism for renal protection against I/R injury, and indicate a potential therapeutic approach for a variety of renal diseases and renal transplantation.
Collapse
|
120
|
Cellular and Molecular Mechanisms of Mesenchymal Stem Cell Actions. Stem Cells Int 2017; 2017:2489041. [PMID: 29362567 PMCID: PMC5738548 DOI: 10.1155/2017/2489041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 01/02/2023] Open
|
121
|
Ratajczak MZ. Looking Back at the Past Year of Stem Cell Reviews and Reports. Stem Cell Rev Rep 2017; 13:703-704. [PMID: 29064017 DOI: 10.1007/s12015-017-9779-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute, University of Louisville, 500 South Floyd Street, Louisville, 40202, USA.
| |
Collapse
|
122
|
Grange C, Iampietro C, Bussolati B. Stem cell extracellular vesicles and kidney injury. Stem Cell Investig 2017; 4:90. [PMID: 29270416 DOI: 10.21037/sci.2017.11.02] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) appear as a new promising cell-free therapy for acute and chronic renal diseases. EVs retain characteristics of the cell of origin and those derived from stem cells may mimic their regenerative properties per se. In fact, EVs contain many active molecules such as proteins and RNA species that act on target cells through different mechanisms, stimulating proliferation and angiogenesis and reducing apoptosis and inflammation. There are several reports that demonstrate a general regenerative potential of EVs derived from mesenchymal stromal cells (MSCs) of different sources in kidney injury models. In addition, a promising new approach is the use of EVs in the graft perfusion solution for kidney conditioning before transplant. Here we summarize the application of EVs released by stem cells in preclinical models of acute and chronic renal damage, comparing animal models, use of EVs of different cell origin and of their sub-fractions, doses, route of administration and efficacy of treatment.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Corinne Iampietro
- Department of Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
123
|
Ratajczak MZ, Ratajczak J. Extracellular Microvesicles as Game Changers in Better Understanding the Complexity of Cellular Interactions-From Bench to Clinical Applications. Am J Med Sci 2017; 354:449-452. [PMID: 29173353 PMCID: PMC5726870 DOI: 10.1016/j.amjms.2017.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 01/13/2023]
Abstract
Recent research has led to wide acceptance and better understanding of a novel mechanism for cell-cell communication that employs a network of extracellular microvesicles (ExMVs). Derived from the plasma membrane or the endosomal membrane compartment, these small, spherical membrane fragments are secreted from the cell surface or in the process of exocytosis from endosomal membrane compartment and (1) with ligands expressed on their surface directly stimulate target cells in a paracrine manner, (2) transfer cell membrane receptors to target cells or (3) deliver encapsulated messenger RNA, microRNA, proteins and bioactive lipids to target cells. This represents an evolutionarily ancient mechanism by which cells signal their presence in the microenvironment, communicate with each other and affect the biology of neighboring cells. Evidence suggests the pivotal role of ExMVs in almost all biological processes within the body as well as their involvement in certain pathologies. Moreover, liquid biopsies based on deciphering the molecular signature of ExMVs promise to revolutionize laboratory diagnostics. At the same time, there are ongoing attempts to employ them as delivery vehicles for drugs as well as therapeutics in regenerative medicine, oncology and immunotherapy.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Department of Medicine, Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky.
| | - Janina Ratajczak
- Department of Medicine, Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
124
|
Lv LL, Wu WJ, Feng Y, Li ZL, Tang TT, Liu BC. Therapeutic application of extracellular vesicles in kidney disease: promises and challenges. J Cell Mol Med 2017; 22:728-737. [PMID: 29083099 PMCID: PMC5783839 DOI: 10.1111/jcmm.13407] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized, membrane‐bound vesicles released from different cells. Recent studies have revealed that EVs may participate in renal tissue damage and regeneration through mediating inter‐nephron communication. Thus, the potential use of EVs as therapeutic vector has gained considerable interest. In this review, we will discuss the basic characteristics of EVs and its role in nephron cellular communication. Then, the application of EVs as therapeutic vector based on its natural content or as carriers of drug, in acute and chronic kidney injury, was discussed. Finally, perspectives and challenges of EVs in therapy of kidney disease were described.
Collapse
Affiliation(s)
- Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Wei-Jun Wu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Ye Feng
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
125
|
Lindoso RS, Collino F, Vieyra A. Extracellular vesicles as regulators of tumor fate: crosstalk among cancer stem cells, tumor cells and mesenchymal stem cells. Stem Cell Investig 2017; 4:75. [PMID: 29057247 DOI: 10.21037/sci.2017.08.08] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/27/2017] [Indexed: 12/16/2022]
Abstract
The tumor microenvironment comprises a heterogeneous population of tumorigenic and non-tumorigenic cells. Cancer stem cells (CSCs) and mesenchymal stem cells (MSCs) are components of this microenvironment and have been described as key regulators of different aspects of tumor physiology. They act differently on the tumor: CSCs are described as tumor initiators and are associated with tumor growth, drug resistance and metastasis; MSCs can integrate the tumor microenvironment after recruitment and interact with cancer cells to promote tumor modifications. Extracellular vesicles (EVs) have emerged as an important mechanism of cell communication under the physiological and pathological conditions. In cancer, secretion of EVs seems to be one of the main mechanisms by which stem cells interact with other tumor and non-tumor cells. The transfer of bioactive molecules (lipids, proteins and RNAs) compartmentalized into EVs triggers different responses in the target cells, regulating several processes in the tumor as angiogenesis, tumor invasiveness and immune escape. This review focuses on the role of CSCs and MSCs in modulating the tumor microenvironment through secretion of EVs, addressing different aspects of the multidirectional interactions among stem cells, tumor and tumor-associated cells.
Collapse
Affiliation(s)
- Rafael Soares Lindoso
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging-CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Federica Collino
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging-CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging-CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,Graduate Program of Translational Biomedicine, Grande Rio University, 25071-202 Duque de Caxias, Brazil
| |
Collapse
|
126
|
Grange C, Tapparo M, Kholia S, Bussolati B, Camussi G. The Distinct Role of Extracellular Vesicles Derived from Normal and Cancer Stem Cells. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0092-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
127
|
Feigerlová E, Battaglia-Hsu SF, Hauet T, Guéant JL. Extracellular vesicles as immune mediators in response to kidney injury. Am J Physiol Renal Physiol 2017; 314:F9-F21. [PMID: 28855190 DOI: 10.1152/ajprenal.00336.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Important progress has been made on cytokine signaling in response to kidney injury in the past decade, especially cytokine signaling mediated by extracellular vesicles (EVs). For example, EVs released by injured renal tubular epithelial cells (TECs) can regulate intercellular communications and influence tissue recovery via both regulating the expression and transferring cytokines, growth factors, as well as other bioactive molecules at the site of injury. The effects of EVs on kidney tissue seem to vary depending on the sources of EVs; however, the literature data are often inconsistent. For example, in rodents EVs derived from mesenchymal stem cells (MSC-EVs) and endothelial progenitor cells (EPC-EVs) can have both beneficial and harmful effects on injured renal tissue. Caution is thus needed in the interpretation of these data as contradictory findings on EVs may not only be related to the origin of EVs, they can also be caused by the different methods used for EV isolation and the physiological and pathological states of the tissues/cells under which they were obtained. Here, we review and discuss our current understanding related to the immunomodulatory function of EVs in renal tubular repair in the hope of encouraging further investigations on mechanisms related to their antiinflammatory and reparative role to better define the therapeutic potential of EVs in renal diseases.
Collapse
Affiliation(s)
- Eva Feigerlová
- INSERM U954, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine, Lorraine, France.,Medical Faculty, University of Lorraine, Lorraine, France.,INSERM, UMR 1082, Poitiers , France.,Medical and Pharmaceutical Faculty, University of Poitiers , Poitiers , France
| | - Shyue-Fang Battaglia-Hsu
- INSERM U954, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine, Lorraine, France.,Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Thierry Hauet
- INSERM, UMR 1082, Poitiers , France.,Medical and Pharmaceutical Faculty, University of Poitiers , Poitiers , France.,Service de Biochimie, Pôle BIOSPHARM, CHU de Poitiers, Poitiers , France
| | - Jean-Louis Guéant
- INSERM U954, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine, Lorraine, France.,Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| |
Collapse
|
128
|
Serum-derived extracellular vesicles (EVs) impact on vascular remodeling and prevent muscle damage in acute hind limb ischemia. Sci Rep 2017; 7:8180. [PMID: 28811546 PMCID: PMC5557987 DOI: 10.1038/s41598-017-08250-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/10/2017] [Indexed: 12/21/2022] Open
Abstract
Serum is an abundant and accessible source of circulating extracellular vesicles (EVs). Serum-EV (sEV) pro-angiogenic capability and mechanisms are herein analyzed using an in vitro assay which predicts sEV angiogenic potential in vivo. Effective sEVs (e-sEVs) also improved vascular remodeling and prevented muscle damage in a mouse model of acute hind limb ischemia. e-sEV angiogenic proteomic and transcriptomic analyses show a positive correlation with matrix-metalloproteinase activation and extracellular matrix organization, cytokine and chemokine signaling pathways, Insulin-like Growth Factor and platelet pathways, and Vascular Endothelial Growth Factor signaling. A discrete gene signature, which highlights differences in e-sEV and ineffective-EV biological activity, was identified using gene ontology (GO) functional analysis. An enrichment of genes associated with the Transforming Growth Factor beta 1 (TGFβ1) signaling cascade is associated with e-sEV administration but not with ineffective-EVs. Chromatin immunoprecipitation analysis on the inhibitor of DNA binding I (ID1) promoter region, and the knock-down of small mother against decapentaplegic (SMAD)1–5 proteins confirmed GO functional analyses. This study demonstrates sEV pro-angiogenic activity, validates a simple, sEV pro-angiogenic assay which predicts their biological activity in vivo, and identifies the TGFβ1 cascade as a relevant mediator. We propose serum as a readily available source of EVs for therapeutic purposes.
Collapse
|
129
|
Bruno S, Tapparo M, Collino F, Chiabotto G, Deregibus MC, Soares Lindoso R, Neri F, Kholia S, Giunti S, Wen S, Quesenberry P, Camussi G. Renal Regenerative Potential of Different Extracellular Vesicle Populations Derived from Bone Marrow Mesenchymal Stromal Cells. Tissue Eng Part A 2017; 23:1262-1273. [PMID: 28471327 PMCID: PMC5689130 DOI: 10.1089/ten.tea.2017.0069] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) derived from human bone marrow mesenchymal stromal cells (MSCs) promote the regeneration of kidneys in different animal models of acute kidney injury (AKI) in a manner comparable with the cells of origin. However, due to the heterogeneity observed in the EVs isolated from MSCs, it is unclear which population is responsible for the proregenerative effects. We therefore evaluated the effect of various EV populations separated by differential ultracentrifugation (10K population enriched with microvesicles and 100K population enriched with exosomes) on AKI recovery. Only the exosomal-enriched population induced an improvement of renal function and morphology comparable with that of the total EV population. Interestingly, the 100K EVs exerted a proproliferative effect on murine tubular epithelial cells, both in vitro and in vivo. Analysis of the molecular content from the different EV populations revealed a distinct profile. The 100K population, for instance, was enriched in specific mRNAs (CCNB1, CDK8, CDC6) reported to influence cell cycle entry and progression; miRNAs involved in regulating proliferative/antiapoptotic pathways and growth factors (hepatocyte growth factor and insulin-like growth factor-1) that could explain the effect of renal tubular cell proliferation. On the other hand, the EV population enriched in microvesicles (10K) was unable to induce renal regeneration and had a molecular profile with lower expression of proproliferative molecules. In conclusion, the different molecular composition of exosome- and microvesicle-enriched populations may explain the regenerative effect of EVs observed in AKI.
Collapse
Affiliation(s)
- Stefania Bruno
- 1 Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Marta Tapparo
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Federica Collino
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy .,3 Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| | - Giulia Chiabotto
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Maria Chiara Deregibus
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Rafael Soares Lindoso
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy .,3 Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| | - Francesco Neri
- 4 Leibniz Institute on Aging-Fritz Lipmann Institute (FLI) Beutenbergstrasse , Jena, Germany .,5 Human Genetics Foundation, University of Torino , Torino, Italy
| | - Sharad Kholia
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Sara Giunti
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Sicheng Wen
- 6 Division of Hematology/Oncology, Brown University , Rhode Island Hospital, Providence, Rhode Island
| | - Peter Quesenberry
- 6 Division of Hematology/Oncology, Brown University , Rhode Island Hospital, Providence, Rhode Island
| | - Giovanni Camussi
- 2 Department of Medical Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| |
Collapse
|