1
|
Huber C, Elsaeed O, Lahmer P, Moertl S. Ionizing radiation effects on blood-derived extracellular vesicles: insights into miR-34a-5p-mediated cellular responses and biomarker potential. Cell Commun Signal 2024; 22:471. [PMID: 39358789 PMCID: PMC11446100 DOI: 10.1186/s12964-024-01845-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Adverse effects of ionizing radiation on normal tissues limit the radiation dose in cancer treatment, thereby compromising treatment efficiency. Among the consistently affected non-cancer cells, peripheral blood mononuclear cells (PBMCs) exhibit high radiosensitivity and have the potential to induce systemic effects. PBMC-released extracellular vesicles (EVs), contribute to the communication of such systemic effects. This study aimed to investigate the effects of ionizing radiation on EVs as part of the systemic response of PBMCs in terms of microRNA cargo and biological functions.Therefore, whole blood samples from healthy donors were irradiated ex-vivo (0 Gy, 1 Gy, 2 Gy, 4 Gy) and EVs from PBMCs were isolated after 96 h by PEG precipitation or ultracentrifugation. Candidate microRNAs were examined in PBMC-derived EVs from individual donors. The uptake of membrane-stained fluorescent EVs by different recipient cells was quantified by fluorescence-activated cell sorting analysis. The biological effects of increased miR-34a-5p and of total EVs on recipient cells were assessed.Irradiation of PBMCs induced a dose-dependent upregulation of miR-34a-5p within EVs and PBMCs. However, interindividual differences between donors were noticed in the extent of upregulation, and small EVs displayed more pronounced changes in microRNA levels in comparison to large EVs. Irradiation in presence of the small molecule inhibitor KU-60019 demonstrated that this upregulation is dependent on ATM (Ataxia telangiectasia mutated) activation. Moreover, fibroblasts and keratinocytes were identified as preferred EV recipients. Increased miR-34a-5p levels led to a significant reduction in viability and induction of senescence in keratinocytes but not in fibroblasts, indicating a cell type-specific response.In conclusion, this study further elucidated the complex cellular response of normal tissue after radiation exposure. It confirmed radiation-induced modifications of microRNA expression levels in EVs from PBMCs and identified a robust upregulation of miR-34a-5p in the small EV subfraction, suggesting this microRNA as a potential novel candidate for the development of biomarkers for radiation exposure. Moreover, the different uptake efficiencies observed among specific cell types suggested that EVs induce cell type-specific responses in the intercellular communication of systemic radiation effects.
Collapse
Affiliation(s)
- Chiara Huber
- Department of Effects and Risks of Ionizing & Non-Ionizing Radiation, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| | - Omar Elsaeed
- Department of Effects and Risks of Ionizing & Non-Ionizing Radiation, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| | - Pia Lahmer
- Department of Effects and Risks of Ionizing & Non-Ionizing Radiation, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| | - Simone Moertl
- Department of Effects and Risks of Ionizing & Non-Ionizing Radiation, Federal Office for Radiation Protection (BfS), Neuherberg, Germany.
| |
Collapse
|
2
|
Carreca AP, Tinnirello R, Miceli V, Galvano A, Gristina V, Incorvaia L, Pampalone M, Taverna S, Iannolo G. Extracellular Vesicles in Lung Cancer: Implementation in Diagnosis and Therapeutic Perspectives. Cancers (Basel) 2024; 16:1967. [PMID: 38893088 PMCID: PMC11171234 DOI: 10.3390/cancers16111967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Lung cancer represents the leading cause of cancer-related mortality worldwide, with around 1.8 million deaths in 2020. For this reason, there is an enormous interest in finding early diagnostic tools and novel therapeutic approaches, one of which is extracellular vesicles (EVs). EVs are nanoscale membranous particles that can carry proteins, lipids, and nucleic acids (DNA and RNA), mediating various biological processes, especially in cell-cell communication. As such, they represent an interesting biomarker for diagnostic analysis that can be performed easily by liquid biopsy. Moreover, their growing dataset shows promising results as drug delivery cargo. The aim of our work is to summarize the recent advances in and possible implications of EVs for early diagnosis and innovative therapies for lung cancer.
Collapse
Affiliation(s)
| | - Rosaria Tinnirello
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (V.M.)
| | - Vitale Miceli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (V.M.)
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, 90133 Palermo, Italy; (A.G.); (V.G.); (L.I.)
| | - Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, 90133 Palermo, Italy; (A.G.); (V.G.); (L.I.)
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, 90133 Palermo, Italy; (A.G.); (V.G.); (L.I.)
| | | | - Simona Taverna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 90146 Palermo, Italy;
| | - Gioacchin Iannolo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (V.M.)
| |
Collapse
|
3
|
Menjivar NG, Oropallo J, Gebremedhn S, Souza LA, Gad A, Puttlitz CM, Tesfaye D. MicroRNA Nano-Shuttles: Engineering Extracellular Vesicles as a Cutting-Edge Biotechnology Platform for Clinical Use in Therapeutics. Biol Proced Online 2024; 26:14. [PMID: 38773366 PMCID: PMC11106895 DOI: 10.1186/s12575-024-00241-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous transporters of various active biomolecules with inflicting phenotypic capabilities, that are naturally secreted by almost all cells with a promising vantage point as a potential leading drug delivery platform. The intrinsic characteristics of their low toxicity, superior structural stability, and cargo loading capacity continue to fuel a multitude of research avenues dedicated to loading EVs with therapeutic and diagnostic cargos (pharmaceutical compounds, nucleic acids, proteins, and nanomaterials) in attempts to generate superior natural nanoscale delivery systems for clinical application in therapeutics. In addition to their well-known role in intercellular communication, EVs harbor microRNAs (miRNAs), which can alter the translational potential of receiving cells and thus act as important mediators in numerous biological and pathological processes. To leverage this potential, EVs can be structurally engineered to shuttle therapeutic miRNAs to diseased recipient cells as a potential targeted 'treatment' or 'therapy'. Herein, this review focuses on the therapeutic potential of EV-coupled miRNAs; summarizing the biogenesis, contents, and function of EVs, as well as providing both a comprehensive discussion of current EV loading techniques and an update on miRNA-engineered EVs as a next-generation platform piloting benchtop studies to propel potential clinical translation on the forefront of nanomedicine.
Collapse
Affiliation(s)
- Nico G Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jaiden Oropallo
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
- Orthopaedic Research Center (ORC), Translational Medicine Institute (TMI), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- J.R. Simplot Company, 1099 W. Front St, Boise, ID, 83702, USA
| | - Luca A Souza
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, 225 Av. Duque de Caxias Norte, Pirassununga, SP, 13635-900, Brazil
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Christian M Puttlitz
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
4
|
Logan CJ, Staton CC, Oliver JT, Bouffard J, Kazmirchuk TDD, Magi M, Brett CL. Thermotolerance in S. cerevisiae as a model to study extracellular vesicle biology. J Extracell Vesicles 2024; 13:e12431. [PMID: 38711329 DOI: 10.1002/jev2.12431] [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: 08/03/2023] [Revised: 02/26/2024] [Accepted: 03/22/2024] [Indexed: 05/08/2024] Open
Abstract
The budding yeast Saccharomyces cerevisiae is a proven model organism for elucidating conserved eukaryotic biology, but to date its extracellular vesicle (EV) biology is understudied. Here, we show yeast transmit information through the extracellular medium that increases survival when confronted with heat stress and demonstrate the EV-enriched samples mediate this thermotolerance transfer. These samples contain vesicle-like particles that are exosome-sized and disrupting exosome biogenesis by targeting endosomal sorting complexes required for transport (ESCRT) machinery inhibits thermotolerance transfer. We find that Bro1, the yeast ortholog of the human exosome biomarker ALIX, is present in EV samples, and use Bro1 tagged with green fluorescent protein (GFP) to track EV release and uptake by endocytosis. Proteomics analysis reveals that heat shock protein 70 (HSP70) family proteins are enriched in EV samples that provide thermotolerance. We confirm the presence of the HSP70 ortholog stress-seventy subunit A2 (Ssa2) in EV samples and find that mutant yeast cells lacking SSA2 produce EVs but they fail to transfer thermotolerance. We conclude that Ssa2 within exosomes shared between yeast cells contributes to thermotolerance. Through this work, we advance Saccharomyces cerevisiae as an emerging model organism for elucidating molecular details of eukaryotic EV biology and establish a role for exosomes in heat stress and proteostasis that seems to be evolutionarily conserved.
Collapse
Affiliation(s)
- Curtis John Logan
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Claire C Staton
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | | - Jeff Bouffard
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | | - Melissa Magi
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | |
Collapse
|
5
|
Cappe B, Vandenabeele P, Riquet FB. A guide to the expanding field of extracellular vesicles and their release in regulated cell death programs. FEBS J 2024; 291:2068-2090. [PMID: 37872002 DOI: 10.1111/febs.16981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Homeostasis disruption is visible at the molecular and cellular levels and may often lead to cell death. This vital process allows us to maintain the more extensive system's integrity by keeping the different features (genetic, metabolic, physiologic, and individual) intact. Interestingly, while cells can die in different manners, dying cells still communicate with their environment. This communication was, for a long time, perceived as only driven by the release of soluble factors. However, it has now been reconsidered with the increasing interest in extracellular vesicles (EVs), which are discovered to be released during different regulated cell death programs, with the observation of specific effects. EVs are game changers in the paradigm of cell-cell communication with tremendous implications in fundamental research with regard to noncell autonomous functions, as well as in biomarkers research, all of which are geared toward diagnostic and therapeutic purposes. This review is composed of two main parts. The first is a comprehensive presentation of the state of the art of the EV field at large. In the second part, we focus on EVs discovered to be released during different regulated cell death programs, also known as cell death EVs (cdEVs), and EV-associated specific effects on recipient cells in the context of cell death and inflammation/inflammatory responses.
Collapse
Affiliation(s)
- Benjamin Cappe
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Franck B Riquet
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
- University of Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, France
| |
Collapse
|
6
|
Shea AG, Idrissou MB, Torres AI, Chen T, Hernandez R, Morris ZS, Sodji QH. Immunological effects of radiopharmaceutical therapy. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1331364. [PMID: 39355211 PMCID: PMC11440989 DOI: 10.3389/fnume.2024.1331364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 10/03/2024]
Abstract
Radiation therapy (RT) is a pillar of cancer therapy used by more than half of all cancer patients. Clinically, RT is mostly delivered as external beam radiation therapy (EBRT). However, the scope of EBRT is limited in the metastatic setting, where all sites of disease need to be irradiated. Such a limitation is attributed to radiation-induced toxicities, for example on bone marrow and hematologic toxicities, resulting from a large EBRT field. Radiopharmaceutical therapy (RPT) has emerged as an alternative to EBRT for the irradiation of all sites of metastatic disease. While RPT can reduce tumor burden, it can also impact the immune system and anti-tumor immunity. Understanding these effects is crucial for predicting and managing treatment-related hematological toxicities and optimizing their integration with other therapeutic modalities, such as immunotherapies. Here, we review the immunomodulatory effects of α- and β-particle emitter-based RPT on various immune cell lines, such as CD8+ and CD4+ T cells, natural killer (NK) cells, and regulatory T (Treg) cells. We briefly discuss Auger electron-emitter (AEE)-based RPT, and finally, we highlight the combination of RPT with immune checkpoint inhibitors, which may offer potential therapeutic synergies for patients with metastatic cancers.
Collapse
Affiliation(s)
- Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Malick Bio Idrissou
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Ana Isabel Torres
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tessa Chen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Reiner Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Quaovi H. Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
7
|
Dellar ER, Hill C, Carter DRF, Baena‐Lopez LA. Oxidative stress-induced changes in the transcriptomic profile of extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e150. [PMID: 38938847 PMCID: PMC11080704 DOI: 10.1002/jex2.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/04/2024] [Accepted: 04/04/2024] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) have been proposed to play dual roles in cellular homeostasis, functioning both to remove unwanted intracellular molecules, and to enable communication between cells as a means of modulating cellular responses in different physiological and pathological scenarios. EVs contain a broad range of cargoes, including multiple biotypes of RNA, which can vary depending on the cell status, and may function as signalling molecules. In this study, we carried out comparative transcriptomic analysis of Drosophila EVs and cells, demonstrating that the RNA profile of EVs is distinct from cells and shows dose-dependent changes in response to oxidative stress. We identified a high abundance of snoRNAs in EVs, alongside an enrichment of intronic and untranslated regions (UTRs) of mRNAs under stress. We also observed an increase in the relative abundance of either aberrant or modified mRNAs under stress. These findings suggest that EVs may function both for the elimination of specific cellular RNAs, and for the incorporation of RNAs that may hold signalling potential.
Collapse
Affiliation(s)
- Elizabeth R. Dellar
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Department of Biological and Medical SciencesOxford Brookes UniversityOxfordUK
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Claire Hill
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Centre for Public HealthQueen's University BelfastBelfastUK
| | - David R. F. Carter
- Department of Biological and Medical SciencesOxford Brookes UniversityOxfordUK
- Evox Therapeutics LimitedOxford Science ParkOxfordUK
| | | |
Collapse
|
8
|
Liu J, You Q, Liang F, Ma L, Zhu L, Wang C, Yang Y. Ultrasound-nanovesicles interplay for theranostics. Adv Drug Deliv Rev 2024; 205:115176. [PMID: 38199256 DOI: 10.1016/j.addr.2023.115176] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/04/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Nanovesicles (NVs) are widely used in the treatment and diagnosis of diseases due to their excellent vascular permeability, good biocompatibility, high loading capacity, and easy functionalization. However, their yield and in vivo penetration depth limitations and their complex preparation processes still constrain their application and development. Ultrasound, as a fundamental external stimulus with deep tissue penetration, concentrated energy sources, and good safety, has been proven to be a patient-friendly and highly efficient strategy to overcome the restrictions of traditional clinical medicine. Recent research has shown that ultrasound can drive the generation of NVs, increase their yield, simplify their preparation process, and provide direct therapeutic effects and intelligent control to enhance the therapeutic effect of NVs. In addition, NVs, as excellent drug carriers, can enhance the targeting efficiency of ultrasound-based sonodynamic therapy or sonogenetic regulation and improve the accuracy of ultrasound imaging. This review provides a detailed introduction to the classification, generation, and modification strategies of NVs, emphasizing the impact of ultrasound on the formation of NVs and summarizing the enhanced treatment and diagnostic effects of NVs combined with ultrasound for various diseases.
Collapse
Affiliation(s)
- Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lilusi Ma
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
9
|
Ahmadi M, Abbasi R, Rezaie J. Tumor immune escape: extracellular vesicles roles and therapeutics application. Cell Commun Signal 2024; 22:9. [PMID: 38167133 PMCID: PMC10763406 DOI: 10.1186/s12964-023-01370-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/28/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Immune escape, a process by which tumor cells evade immune surveillance, remains a challenge for cancer therapy. Tumor cells produce extracellular vesicles (EVs) that participate in immune escape by transferring bioactive molecules between cells. EVs refer to heterogeneous vesicles that participate in intercellular communication. EVs from tumor cells usually carry tumor antigens and have been considered a source of tumor antigens to induce anti-tumor immunity. However, evidence also suggests that these EVs can accelerate immune escape by carrying heat shock proteins (HSPs), programmed death-ligand 1 (PD-L1), etc. to immune cells, suppressing function and exhausting the immune cells pool. EVs are progressively being evaluated for therapeutic implementation in cancer therapies. EVs-based immunotherapies involve inhibiting EVs generation, using natural EVs, and harnessing engineering EVs. All approaches are associated with advantages and disadvantages. The EVs heterogeneity and diverse physicochemical properties are the main challenges to their clinical applications. SHORT CONCLUSION Although EVs are criminal; they can be useful for overcoming immune escape. This review discusses the latest knowledge on EVs population and sheds light on the function of tumor-derived EVs in immune escape. It also describes EVs-based immunotherapies with a focus on engineered EVs, followed by challenges that hinder the clinical translation of EVs that are essential to be addressed in future investigations. Video Abstract.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Abbasi
- Department of Biology, Urmia University, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
10
|
Lisi V, Senesi G, Balbi C. Converging protective pathways: Exploring the linkage between physical exercise, extracellular vesicles and oxidative stress. Free Radic Biol Med 2023; 208:718-727. [PMID: 37739138 DOI: 10.1016/j.freeradbiomed.2023.09.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Physical Exercise (EXR) has been shown to have numerous beneficial effects on various systems in the human body. It leads to a decrease in the risk of mortality from chronic diseases, such as cardiovascular disease, cancer, metabolic and central nervous system disorders. EXR results in improving cardiovascular fitness, cognitive function, immune activity, endocrine action, and musculoskeletal health. These positive effects make EXR a valuable intervention for promoting overall health and well-being in individuals of all ages. These beneficial effects are partially mediated by the role of the regular EXR in the adaptation to redox homeostasis counteracting the sudden increase of ROS, the hallmark of many chronic diseases. EXR can trigger the release of numerous humoral factors, e.g. protein, microRNA (miRs), and DNA, that can be shuttled as cargo of Extracellular vesicles (EVs). EVs show different cargo modification after oxidative stress stimuli as well as after EXR. In this review, we aim to highlight the main studies on the role of EVs released during EXR and oxidative stress conditions in enhancing the antioxidant enzymes pathway and in the decrease of oxidative stress environment mediated by their cargo.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy.
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| |
Collapse
|
11
|
Menjivar NG, Gad A, Thompson RE, Meyers MA, Hollinshead FK, Tesfaye D. Bovine oviductal organoids: a multi-omics approach to capture the cellular and extracellular molecular response of the oviduct to heat stress. BMC Genomics 2023; 24:646. [PMID: 37891479 PMCID: PMC10605953 DOI: 10.1186/s12864-023-09746-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The mammalian oviduct is a complex, fibromuscular organ known for its role in orchestrating a series of timely and dynamic changes to suitably support early embryogenesis. Climate change-induced heat stress (HS) is one of the largest single stressors compromising reproductive function in humans and farm animals via systemic changes in the redox status of the maternal environment, adversely affecting fertilization and early embryonic development. Oviductal organoids represent a unique 3-dimensional, biomimetic model to study the physiology of the oviduct and its subsequent impact on embryo development under various environmental conditions. RESULTS Our study is the first to demonstrate an innovative approach to understanding the cascade of molecular changes sustained by bovine oviductal organoids under HS and the subsequent maternal signals harnessed within their secreted extracellular vesicles (EVs). Transcriptomic analysis of oviductal organoids exposed to HS revealed 2,570 differentially expressed genes (1,222 up- and 1,348 downregulated), while EV-coupled miRNome analysis disclosed 18 miRNAs with significant differential expression (12 up- and 6 downregulated) in EVs from thermally stressed organoids compared to EVs released from organoids cultured under thermoneutral conditions. Genes activated in oviductal organoids in response to thermal stress, include: COX1, ACTB, CST6, TPT1, and HSPB1, while miR-1246, miR-148a, miR21-5p, miR-451, and miR-92a represent the top highly abundant EV-coupled miRNAs released in response to HS. Pathway analysis of genes enriched in organoids exposed to thermal stress showed the enrichment of endocrine resistance, cellular senescence, and notch signaling pathways. Similarly, EV-coupled miRNAs released from thermally stressed organoids showed their potential regulation of genes involved in cellular senescence, p53 signaling, and TGF-beta signaling pathways. CONCLUSIONS In conclusion, the cellular and extracellular response of bovine oviductal organoids to in vitro HS conditions reveal the prospective impact of environmental HS on the physiology of the oviduct and the probable subsequent impacts on oocyte fertilization and early embryo development. Future studies elucidating the potential impact of HS-associated EVs from oviductal organoids on oocyte fertilization and preimplantation embryo development, would justify the use of an organoid model to optimally understand the oviduct-embryo communication under suboptimal environments.
Collapse
Affiliation(s)
- Nico G Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 3107 Rampart Rd, Fort Collins, CO, 80521, USA
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 3107 Rampart Rd, Fort Collins, CO, 80521, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Riley E Thompson
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Mindy A Meyers
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Fiona K Hollinshead
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 3107 Rampart Rd, Fort Collins, CO, 80521, USA.
| |
Collapse
|
12
|
Crewe C. Energetic Stress-Induced Metabolic Regulation by Extracellular Vesicles. Compr Physiol 2023; 13:5051-5068. [PMID: 37358503 PMCID: PMC10414774 DOI: 10.1002/cphy.c230001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Recent studies have demonstrated that extracellular vesicles (EVs) serve powerful and complex functions in metabolic regulation and metabolic-associated disease, although this field of research is still in its infancy. EVs are released into the extracellular space from all cells and carry a wide range of cargo including miRNAs, mRNA, DNA, proteins, and metabolites that have robust signaling effects in receiving cells. EV production is stimulated by all major stress pathways and, as such, has a role in both restoring homeostasis during stress and perpetuating disease. In metabolic regulation, the dominant stress signal is a lack of energy due to either nutrient deficits or damaged mitochondria from nutrient excess. This stress signal is termed "energetic stress," which triggers a robust and evolutionarily conserved response that engages major cellular stress pathways, the ER unfolded protein response, the hypoxia response, the antioxidant response, and autophagy. This article proposes the model that energetic stress is the dominant stimulator of EV release with a focus on metabolically important cells such as hepatocytes, adipocytes, myocytes, and pancreatic β-cells. Furthermore, this article will discuss how the cargo in stress-stimulated EVs regulates metabolism in receiving cells in both beneficial and detrimental ways. © 2023 American Physiological Society. Compr Physiol 13:5051-5068, 2023.
Collapse
Affiliation(s)
- Clair Crewe
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Menjivar NG, Gad A, Gebremedhn S, Ghosh S, Tesfaye D. Granulosa cell-derived extracellular vesicles mitigate the detrimental impact of thermal stress on bovine oocytes and embryos. Front Cell Dev Biol 2023; 11:1142629. [PMID: 37091982 PMCID: PMC10116072 DOI: 10.3389/fcell.2023.1142629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Climate change-induced global warming results in rises in body temperatures above normal physiological levels (hyperthermia) with negative impacts on reproductive function in dairy and beef animals. Extracellular vesicles (EVs), commonly described as nano-sized, lipid-enclosed complexes, harnessed with a plethora of bioactive cargoes (RNAs, proteins, and lipids), are crucial to regulating processes like folliculogenesis and the initiation of different signaling pathways. The beneficial role of follicular fluid-derived EVs in inducing thermotolerance to oocytes during in vitro maturation (IVM) has been evidenced. Here we aimed to determine the capacity of in vitro cultured granulosa cell-derived EVs (GC-EVs) to modulate bovine oocytes’ thermotolerance to heat stress (HS) during IVM. Moreover, this study tested the hypothesis that EVs released from thermally stressed GCs (S-EVs) shuttle protective messages to provide protection against subsequent HS in bovine oocytes. For this, sub-populations of GC-EVs were generated from GCs subjected to 38.5°C (N-EVs) or 42°C (S-EVs) and supplemented to cumulus-oocyte complexes (COCs) matured in vitro at the normal physiological body temperature of the cow (38.5°C) or HS (41°C) conditions. Results indicate that S-EVs improve the survival of oocytes by reducing ROS accumulation, improving mitochondrial function, and suppressing the expression of stress-associated genes thereby reducing the severity of HS on oocytes. Moreover, our findings indicate a carryover impact from the addition of GC-EVs during oocyte maturation in the development to the blastocyst stage with enhanced viability.
Collapse
Affiliation(s)
- Nico G. Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Genus Plc, Deforest, WI, United States
| | - Soham Ghosh
- Cellular Engineering and Mechanobiology Laboratory (CEML), Department of Mechanical Engineering, Translational Medicine Institute (TMI), Colorado State University, Fort Collins, CO, United States
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- *Correspondence: Dawit Tesfaye,
| |
Collapse
|
14
|
Hill C, Dellar ER, Baena‐Lopez LA. Caspases help to spread the message via extracellular vesicles. FEBS J 2023; 290:1954-1972. [PMID: 35246932 PMCID: PMC10952732 DOI: 10.1111/febs.16418] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022]
Abstract
Cell-cell communication is an essential aspect of multicellular life, key for coordinating cell proliferation, growth, and death in response to environmental changes. Whilst caspases are well-known for facilitating apoptotic and pyroptotic cell death, several recent investigations are uncovering new roles for these enzymes in biological scenarios requiring long-range intercellular signalling mediated by extracellular vesicles (EVs). EVs are small membrane-bound nanoparticles released from cells that may carry and deliver cargo between distant cells, thus helping to coordinate their behaviour. Intriguingly, there is emerging evidence indicating a key contribution of caspases in the biogenesis of EVs, the selection of their cargo content, and EV uptake/function in recipient cells. Here, we discuss the latest findings supporting the interplay between caspases and EVs, and the biological relevance of this molecular convergence for cellular signalling, principally in non-apoptotic scenarios.
Collapse
Affiliation(s)
- Claire Hill
- Sir William Dunn School of PathologyUniversity of OxfordUK
| | - Elizabeth R. Dellar
- Sir William Dunn School of PathologyUniversity of OxfordUK
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordUK
| | | |
Collapse
|
15
|
Robado de Lope L, Sánchez‐Herrero E, Serna‐Blasco R, Provencio M, Romero A. Cancer as an infective disease: the role of EVs in tumorigenesis. Mol Oncol 2023; 17:390-406. [PMID: 36168102 PMCID: PMC9980310 DOI: 10.1002/1878-0261.13316] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/19/2022] [Accepted: 09/26/2022] [Indexed: 11/09/2022] Open
Abstract
Cancer is conventionally considered an evolutionary disease where tumor cells adapt to the environment and evolve eventually leading to the formation of metastasis through the seeding and growth of metastasis-initiating cells in distant organs. Tumor cell and tumor-stroma communication via soluble factors and extracellular vesicles (EVs) are essential for the success of the metastatic process. As the field of EVs advances, growing data support the role of tumor-derived EVs not only in modifying the microenvironment to facilitate tumor progression but also in inducing changes in cells outside the primary tumor that may lead to a malignant transformation. Thus, an alternative hypothesis has emerged suggesting the conceptualization of cancer as an 'infective' disease. Still, tackling EVs as a possible cancer treatment has not been widely explored. A major understanding is needed to unveil possible additional contributions of EVs in progression and metastasis, which may be essential for the development of novel approaches to treat cancer patients. Here, we review the contribution of EVs to cancer progression and the possible implication of these factors in the oncogenic transformation of indolent cells.
Collapse
Affiliation(s)
- Lucia Robado de Lope
- Liquid Biopsy LaboratoryBiomedical Sciences Research Institute Puerta de Hierro‐MajadahondaSpain
| | - Estela Sánchez‐Herrero
- Liquid Biopsy LaboratoryBiomedical Sciences Research Institute Puerta de Hierro‐MajadahondaSpain
- Atrys HealthBarcelonaSpain
| | - Roberto Serna‐Blasco
- Liquid Biopsy LaboratoryBiomedical Sciences Research Institute Puerta de Hierro‐MajadahondaSpain
| | - Mariano Provencio
- Liquid Biopsy LaboratoryBiomedical Sciences Research Institute Puerta de Hierro‐MajadahondaSpain
- Medical Oncology DepartmentHospital Universitario Puerta de Hierro‐MajadahondaSpain
| | - Atocha Romero
- Liquid Biopsy LaboratoryBiomedical Sciences Research Institute Puerta de Hierro‐MajadahondaSpain
- Medical Oncology DepartmentHospital Universitario Puerta de Hierro‐MajadahondaSpain
| |
Collapse
|
16
|
Worsley CM, Veale RB, Mayne ES. The effect of acute acid exposure on immunomodulatory protein secretion, cell survival, and cell cycle progression in tumour cell lines. Cytokine 2023; 162:156118. [PMID: 36584453 DOI: 10.1016/j.cyto.2022.156118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/05/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Cancer develops when multiple systems fail to suppress uncontrolled cell proliferation. Breast cancers and oesophageal squamous cell carcinoma (OSCC) are common cancers prone to genetic instability. They typically occur in acidic microenvironments which impacts on cell proliferation, apoptosis, and their influence on surrounding cells to support tumour growth and immune evasion. This study aimed to evaluate the impact of the acidic tumour microenvironment on the production of pro-tumorigenic and immunomodulatory factors in cancer cell lines. Multiple factors that may mediate immune evasion were secreted including IL-6, IL-8, G-CSF, IP-10, GDF-15, Lipocalin-2, sICAM-1, and myoglobin. Others, such as VEGF, FGF, and EGF that are essential for tumour cell survival were also detected. Treatment with moderate acidity did not significantly affect secretion of most proteins, whereas very low pH did. Distinct differences in apoptosis were noted between the cell lines, with WHCO6 being better adapted to survive at moderate acid levels. Conditioned medium from acid-treated cells stimulated increased cell viability and proliferation in WHCO6, but increased cell death in MCF-7. This study highlights the importance of acidic tumour microenvironment in controlling apoptosis, cell proliferation, and immune evasion which may be different at different anatomical sites. Immunomodulatory molecules and growth factors provide therapeutic targets to improve the prognosis of individuals with cancer.
Collapse
Affiliation(s)
- Catherine M Worsley
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa; Department of Haematology and Molecular Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa; National Health Laboratory Service, South Africa.
| | - Rob B Veale
- School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, South Africa
| | - Elizabeth S Mayne
- National Health Laboratory Service, South Africa; Department of Immunology Faculty of Health Sciences, University of the Witwatersrand, South Africa; Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa
| |
Collapse
|
17
|
Rayamajhi S, Sulthana S, Ferrel C, Shrestha TB, Aryal S. Extracellular vesicles production and proteomic cargo varies with incubation time and temperature. Exp Cell Res 2023; 422:113454. [PMID: 36584743 PMCID: PMC9878443 DOI: 10.1016/j.yexcr.2022.113454] [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: 09/05/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022]
Abstract
Extracellular vesicles (EVs) are heterogenous populations of proteolipid bi-layered vesicles secreted by cells as an important biological process. EVs cargo can reflect the cellular environmental conditions in which cells grow. The use of serum-free conditioned media to harvest EVs leads to stress-mediated cellular changes with longer incubation time and impacts EV production and functionality. This study aims to explore the role of incubation time and temperature on EV production and proteomic cargo. For this purpose, an optimized ultrafiltration-size exclusion chromatography-based technique is developed, which isolates small EVs ranging from 130 to 220 nm. The result shows higher EVs production in cancerous cells (K7M2) compared to noncancerous cells (NIH/3T3), which increases with longer incubation time and elevated temperature. Mass spectrometry-based proteomic characterization of EVs showed incubation time and temperature-dependent proteomic profile. A set of enriched EV proteins were identified in EVs isolated at nutrient-stress (72 h incubation time) and heat-stress (40 °C incubation temperature) environment. Enrichment of Serpinb1a in EVs isolated in heat stress was further validated via immunoblot. Gene enrichment analysis revealed that enriched EV proteins following nutrient stress were involved in negative regulation of transcription, response to oxidative stress, and protein folding. Likewise, enriched EV proteins following heat stress were involved in oxaloacetate and aspartate metabolism, and glutamate catabolic process. EVs isolated under nutrient stress showed pro-proliferative activity whereas EVs isolated under heat stress showed anti-proliferative activity. Our results show that incubation time and temperature can alter EV production, its proteomic cargo, and functionality, which can be used to design need-based standard isolation parameters for reproducible EV research.
Collapse
Affiliation(s)
- Sagar Rayamajhi
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA; Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS, 66506, USA
| | - Shoukath Sulthana
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler. W.T. Brookshire Hall 370, Tyler, TX, 75799, USA
| | - Colin Ferrel
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS, 66506, USA
| | - Tej B Shrestha
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS, 66506, USA; Department of Anatomy & Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler. W.T. Brookshire Hall 370, Tyler, TX, 75799, USA.
| |
Collapse
|
18
|
Alberti G, Russo E, Corrao S, Anzalone R, Kruzliak P, Miceli V, Conaldi PG, Di Gaudio F, La Rocca G. Current Perspectives on Adult Mesenchymal Stromal Cell-Derived Extracellular Vesicles: Biological Features and Clinical Indications. Biomedicines 2022; 10:2822. [PMID: 36359342 PMCID: PMC9687875 DOI: 10.3390/biomedicines10112822] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 08/10/2023] Open
Abstract
Extracellular vesicles (EVs) constitute one of the main mechanisms by which cells communicate with the surrounding tissue or at distance. Vesicle secretion is featured by most cell types, and adult mesenchymal stromal cells (MSCs) of different tissue origins have shown the ability to produce them. In recent years, several reports disclosed the molecular composition and suggested clinical indications for EVs derived from adult MSCs. The parental cells were already known for their roles in different disease settings in regulating inflammation, immune modulation, or transdifferentiation to promote cell repopulation. Interestingly, most reports also suggested that part of the properties of parental cells were maintained by isolated EV populations. This review analyzes the recent development in the field of cell-free therapies, focusing on several adult tissues as a source of MSC-derived EVs and the available clinical data from in vivo models.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Eleonora Russo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Simona Corrao
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Rita Anzalone
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Peter Kruzliak
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | | | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
19
|
Huda MN, Nurunnabi M. Potential Application of Exosomes in Vaccine Development and Delivery. Pharm Res 2022; 39:2635-2671. [PMID: 35028802 PMCID: PMC8757927 DOI: 10.1007/s11095-021-03143-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023]
Abstract
Exosomes are cell-derived components composed of proteins, lipid, genetic information, cytokines, and growth factors. They play a vital role in immune modulation, cell-cell communication, and response to inflammation. Immune modulation has downstream effects on the regeneration of damaged tissue, promoting survival and repair of damaged resident cells, and promoting the tumor microenvironment via growth factors, antigens, and signaling molecules. On top of carrying biological messengers like mRNAs, miRNAs, fragmented DNA, disease antigens, and proteins, exosomes modulate internal cell environments that promote downstream cell signaling pathways to facilitate different disease progression and induce anti-tumoral effects. In this review, we have summarized how vaccines modulate our immune response in the context of cancer and infectious diseases and the potential of exosomes as vaccine delivery vehicles. Both pre-clinical and clinical studies show that exosomes play a decisive role in processes like angiogenesis, prognosis, tumor growth metastasis, stromal cell activation, intercellular communication, maintaining cellular and systematic homeostasis, and antigen-specific T- and B cell responses. This critical review summarizes the advancement of exosome based vaccine development and delivery, and this comprehensive review can be used as a valuable reference for the broader delivery science community.
Collapse
Affiliation(s)
- Md Nurul Huda
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
20
|
Indirect mediators of systemic health outcomes following nanoparticle inhalation exposure. Pharmacol Ther 2022; 235:108120. [PMID: 35085604 PMCID: PMC9189040 DOI: 10.1016/j.pharmthera.2022.108120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
The growing field of nanoscience has shed light on the wide diversity of natural and anthropogenic sources of nano-scale particulates, raising concern as to their impacts on human health. Inhalation is the most robust route of entry, with nanoparticles (NPs) evading mucociliary clearance and depositing deep into the alveolar region. Yet, impacts from inhaled NPs are evident far outside the lung, particularly on the cardiovascular system and highly vascularized organs like the brain. Peripheral effects are partly explained by the translocation of some NPs from the lung into the circulation; however, other NPs largely confined to the lung are still accompanied by systemic outcomes. Omic research has only just begun to inform on the complex myriad of molecules released from the lung to the blood as byproducts of pulmonary pathology. These indirect mediators are diverse in their molecular make-up and activity in the periphery. The present review examines systemic outcomes attributed to pulmonary NP exposure and what is known about indirect pathological mediators released from the lung into the circulation. Further focus was directed to outcomes in the brain, a highly vascularized region susceptible to acute and longer-term outcomes. Findings here support the need for big-data toxicological studies to understand what drives these health outcomes and better predict, circumvent, and treat the potential health impacts arising from NP exposure scenarios.
Collapse
|
21
|
Stinson LF, Trevenen ML, Geddes DT. Effect of Cold Storage on the Viable and Total Bacterial Populations in Human Milk. Nutrients 2022; 14:1875. [PMID: 35565846 PMCID: PMC9099816 DOI: 10.3390/nu14091875] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Expression and cold storage of human milk is a common practice. Current guidelines for cold storage of expressed milk do not take into account the impact on the milk microbiome. Here, we investigated the impact of cold storage on viable bacterial populations in human milk. Freshly expressed milk samples (n = 10) were collected and analysed immediately, stored at 4 °C for four days, −20 °C for 2.25 months and 6 months, and −80 °C for 6 months. Samples were analysed using propidium monoazide (PMA; a cell viability dye) coupled with full-length 16S rRNA gene. An aliquot of each sample was additionally analysed without PMA to assess the impact of cold storage on the total DNA profile of human milk. Cold storage significantly altered the composition of both the viable microbiome and total bacterial DNA profile, with differences in the relative abundance of several OTUs observed across each storage condition. However, cold storage did not affect the richness nor diversity of the samples (PERMANOVA all p > 0.2). Storage of human milk under typical and recommended conditions results in alterations to the profile of viable bacteria, with potential implications for infant gut colonisation and infant health.
Collapse
Affiliation(s)
- Lisa F. Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6000, Australia;
| | - Michelle L. Trevenen
- Centre for Applied Statistics, The University of Western Australia, Perth, WA 6000, Australia;
| | - Donna T. Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6000, Australia;
| |
Collapse
|
22
|
Clarke-Bland CE, Bill RM, Devitt A. Emerging roles for AQP in mammalian extracellular vesicles. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183826. [PMID: 34843700 PMCID: PMC8755917 DOI: 10.1016/j.bbamem.2021.183826] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Recent research in the aquaporin (AQP) field has identified a role for diverse AQPs in extracellular vesicles (EV). Though still in its infancy, there is a growing body of knowledge in the area; AQPs in EV have been suggested as biomarkers for disease, as drug targets and show potential as therapeutics. To advance further in this field, AQPs in EV must be better understood. Here we summarize current knowledge of the presence and function of AQPs in EV and hypothesise their roles in health and disease.
Collapse
Affiliation(s)
| | - Roslyn M Bill
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Andrew Devitt
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK.
| |
Collapse
|
23
|
Tesfaye D, Menjivar N, Gebremedhn S. Current knowledge and the future potential of extracellular vesicles in mammalian reproduction. Reprod Fertil Dev 2021; 34:174-189. [PMID: 35231266 DOI: 10.1071/rd21277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs), which contain various functional classes of vesicles, namely exosomes, microvesicles, and apoptotic bodies, represent the major nano-shuttle to transfer bioactive molecules from donor to recipient cells to facilitate cell-to-cell communication in the follicular, oviduct, and uterine microenvironments. In addition to transferring various molecular cargos in the form of miRNAs, mRNAs, proteins, lipids, and DNA molecules, the relative proportion of those molecular cargos in the reproductive fluids can be associated with the physiological and pathological condition of the host animal. Inside the follicle, EV-mediated circulation of miRNAs has been reported to be associated with the growth status of the enclosed oocytes, the metabolic status, and the advanced maternal aging of the animal. Importantly, EVs have the potential to protect their cargo molecules from extracellular degradation or modification while travelling to the recipient cells. This fact together with the enormous availability in almost all biological fluids and spent culture media make them attractive in the search for biomarkers of oocyte/embryo developmental competence, receptive maternal environment and a multitude of reproductive pathophysiological conditions. One of the key factors that have contributed to the lower efficiency of assisted reproductive technologies (ART) is the absence of several maternal in vivo factors in the ART procedures. For this, several studies have been conducted to supplement various components present in the follicular and oviductal fluids into the existing ART procedures and significant positive impacts have been observed in terms of embryo cleavage rate, blastocyst rate, resistance to stress, and survival after cryopreservation. The potential of EVs in shuttling protective messages against environmental and physiological stressors has been evidenced. The effective use of the EV-coupled molecular signals against stress-associated conditions has the potential to pave the path for the application of these protective signals against oxidative stress-associated pathological conditions including PCOS, ageing, and endometritis. In this review, we provide current knowledge and potential future use of EVs as remedies in reproductive pathophysiological conditions, mainly in follicular and oviductal microenvironments.
Collapse
Affiliation(s)
- Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Road, Fort Collins, CO 80521, USA
| | - Nico Menjivar
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Road, Fort Collins, CO 80521, USA
| | | |
Collapse
|
24
|
Pascual-Antón L, Cardeñes B, Sainz de la Cuesta R, González-Cortijo L, López-Cabrera M, Cabañas C, Sandoval P. Mesothelial-to-Mesenchymal Transition and Exosomes in Peritoneal Metastasis of Ovarian Cancer. Int J Mol Sci 2021; 22:ijms222111496. [PMID: 34768926 PMCID: PMC8584135 DOI: 10.3390/ijms222111496] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Most patients with ovarian cancer (OvCA) present peritoneal disseminated disease at the time of diagnosis. During peritoneal metastasis, cancer cells detach from the primary tumor and disseminate through the intraperitoneal fluid. The peritoneal mesothelial cell (PMC) monolayer that lines the abdominal cavity is the first barrier encountered by OvCA cells. Subsequent progression of tumors through the peritoneum leads to the accumulation into the peritoneal stroma of a sizeable population of carcinoma-associated fibroblasts (CAFs), which is mainly originated from a mesothelial-to-mesenchymal transition (MMT) process. A common characteristic of OvCA patients is the intraperitoneal accumulation of ascitic fluid, which is composed of cytokines, chemokines, growth factors, miRNAs, and proteins contained in exosomes, as well as tumor and mesothelial suspended cells, among other components that vary in proportion between patients. Exosomes are small extracellular vesicles that have been shown to mediate peritoneal metastasis by educating a pre-metastatic niche, promoting the accumulation of CAFs via MMT, and inducing tumor growth and chemoresistance. This review summarizes and discusses the pivotal role of exosomes and MMT as mediators of OvCA peritoneal colonization and as emerging diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Lucía Pascual-Antón
- Tissue and Organ Homeostasis Program, Cell-Cell Communication and Inflammation Unit, Centro de Biología Molecular “Severo Ochoa” (UAM-CSIC), Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; (L.P.-A.); (B.C.); (M.L.-C.)
| | - Beatriz Cardeñes
- Tissue and Organ Homeostasis Program, Cell-Cell Communication and Inflammation Unit, Centro de Biología Molecular “Severo Ochoa” (UAM-CSIC), Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; (L.P.-A.); (B.C.); (M.L.-C.)
| | | | | | - Manuel López-Cabrera
- Tissue and Organ Homeostasis Program, Cell-Cell Communication and Inflammation Unit, Centro de Biología Molecular “Severo Ochoa” (UAM-CSIC), Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; (L.P.-A.); (B.C.); (M.L.-C.)
| | - Carlos Cabañas
- Tissue and Organ Homeostasis Program, Cell-Cell Communication and Inflammation Unit, Centro de Biología Molecular “Severo Ochoa” (UAM-CSIC), Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; (L.P.-A.); (B.C.); (M.L.-C.)
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Lymphocyte Immunobiology Group, Inflammatory and Immune Disorders Area, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain
- Correspondence: (C.C.); (P.S.); Tel.: +34-91-196-4513 (C.C.); +34-91-196-4707 (P.S.)
| | - Pilar Sandoval
- Tissue and Organ Homeostasis Program, Cell-Cell Communication and Inflammation Unit, Centro de Biología Molecular “Severo Ochoa” (UAM-CSIC), Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; (L.P.-A.); (B.C.); (M.L.-C.)
- Correspondence: (C.C.); (P.S.); Tel.: +34-91-196-4513 (C.C.); +34-91-196-4707 (P.S.)
| |
Collapse
|
25
|
Hariharan H, Kesavan Y, Raja NS. Impact of native and external factors on exosome release: understanding reactive exosome secretion and its biogenesis. Mol Biol Rep 2021; 48:7559-7573. [PMID: 34626311 DOI: 10.1007/s11033-021-06733-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/29/2021] [Indexed: 02/04/2023]
Abstract
Exosomes are minuscule vesicles secreted in the endolytic region of most mammalian cells. The release of exosomes from the cell engenders cell-to-cell signaling between cellular-compartments. The trading of exosomes between tumor and yonder cells plays a hypercritical role in tumor growth and progression. The exosome released from each tumor cell sequestrates a unique biogenetic pathway reflecting its cellular origin depending on the tumor type. However, treatment of tumor cells with certain physiological factors like drugs, chemotherapy, radiation, etc., enhance the release of exosomes and alters its biogenetic pathway compared with untreated tumor cells. In this review, we will discuss how the non-native physiological factors influence the release of exosomes and how these reactive exosomes orchestrate a unique patterning of a cargo sorting mechanism. We will also discuss the role of reactively secreted exosomes in mediating tumor metastasis, angiogenesis, and tumor progression.
Collapse
Affiliation(s)
- Harini Hariharan
- MPI Lab, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu, India
| | - Yasodha Kesavan
- MPI Lab, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu, India
| | - Natesan Sella Raja
- MPI Lab, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu, India.
| |
Collapse
|
26
|
Shaw A, Gullerova M. Home and Away: The Role of Non-Coding RNA in Intracellular and Intercellular DNA Damage Response. Genes (Basel) 2021; 12:1475. [PMID: 34680868 PMCID: PMC8535248 DOI: 10.3390/genes12101475] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
Non-coding RNA (ncRNA) has recently emerged as a vital component of the DNA damage response (DDR), which was previously believed to be solely regulated by proteins. Many species of ncRNA can directly or indirectly influence DDR and enhance DNA repair, particularly in response to double-strand DNA breaks, which may hold therapeutic potential in the context of cancer. These include long non-coding RNA (lncRNA), microRNA, damage-induced lncRNA, DNA damage response small RNA, and DNA:RNA hybrid structures, which can be categorised as cis or trans based on the location of their synthesis relative to DNA damage sites. Mechanisms of RNA-dependent DDR include the recruitment or scaffolding of repair factors at DNA break sites, the regulation of repair factor expression, and the stabilisation of repair intermediates. DDR can also be communicated intercellularly via exosomes, leading to bystander responses in healthy neighbour cells to generate a population-wide response to damage. Many microRNA species have been directly implicated in the propagation of bystander DNA damage, autophagy, and radioresistance, which may prove significant for enhancing cancer treatment via radiotherapy. Here, we review recent developments centred around ncRNA and their contributions to intracellular and intercellular DDR mechanisms.
Collapse
Affiliation(s)
| | - Monika Gullerova
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK;
| |
Collapse
|
27
|
The Role and Clinical Interest of Extracellular Vesicles in Pregnancy and Ovarian Cancer. Biomedicines 2021; 9:biomedicines9091257. [PMID: 34572444 PMCID: PMC8464910 DOI: 10.3390/biomedicines9091257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer and pregnancy are two states in which the host immune system is exposed to novel antigens. Indeed, both the tumor and placenta must invade tissues, remodel vasculature to establish a robust blood supply, and evade detection by the immune system. Interestingly, tumor and placenta tissue use similar mechanisms to induce these necessary changes. One mediator is emerging as a key player in invasion, vascular remodeling, and immune evasion: extracellular vesicles (EVs). Many studies have identified EVs as a key mediator of cell-to-cell communication. Specifically, the cargo carried by EVs, which includes proteins, nucleic acids, and lipids, can interact with cells to induce changes in the target cell ranging from gene expression to migration and metabolism. EVs can promote cell division and tissue invasion, immunosuppression, and angiogenesis which are essential for both cancer and pregnancy. In this review, we examine the role of EVs in ovarian cancer metastasis, chemoresistance, and immune modulation. We then focus on the role of EVs in pregnancy with special attention on the vascular remodeling and regulation of the maternal immune system. Lastly, we discuss the clinical utility of EVs as markers and therapeutics for ovarian cancer and pre-eclampsia.
Collapse
|
28
|
Role of Extracellular Vesicles in Compromising Cellular Resilience to Environmental Stressors. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9912281. [PMID: 34337063 PMCID: PMC8321721 DOI: 10.1155/2021/9912281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs), like exosomes, are nanosized membrane-enveloped vesicles containing different bioactive cargo, such as proteins, lipids, mRNA, miRNA, and other small regulatory RNAs. Cell-derived EVs, including EVs originating from stem cells, may capture components from damaged cells or cells impacted by therapeutic treatments. Interestingly, EVs derived from stem cells can be preconditioned to produce and secrete EVs with different therapeutic properties, particularly with respect to heat-shock proteins and other molecular cargo contents. This behavior is consistent with stem cells that also respond differently to various microenvironments. Heat-shock proteins play roles in cellular protection and mediate cellular resistance to radiotherapy, chemotherapy, and heat shock. This review highlights the possible roles EVs play in mediating cellular plasticity and survival when exposed to different physical and chemical stressors, with a special focus on the respiratory distress due to the air pollution.
Collapse
|
29
|
Hannon G, Tansi FL, Hilger I, Prina‐Mello A. The Effects of Localized Heat on the Hallmarks of Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
30
|
Akbar N, Paget D, Choudhury RP. Extracellular Vesicles in Innate Immune Cell Programming. Biomedicines 2021; 9:biomedicines9070713. [PMID: 34201592 PMCID: PMC8301301 DOI: 10.3390/biomedicines9070713] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EV) are a heterogeneous group of bilipid-enclosed envelopes that carry proteins, metabolites, RNA, DNA and lipids from their parent cell of origin. They mediate cellular communication to other cells in local tissue microenvironments and across organ systems. EV size, number and their biologically active cargo are often altered in response to pathological processes, including infection, cancer, cardiovascular diseases and in response to metabolic perturbations such as obesity and diabetes, which also have a strong inflammatory component. Here, we discuss the broad repertoire of EV produced by neutrophils, monocytes, macrophages, their precursor hematopoietic stem cells and discuss their effects on the innate immune system. We seek to understand the immunomodulatory properties of EV in cellular programming, which impacts innate immune cell differentiation and function. We further explore the possibilities of using EV as immune targeting vectors, for the modulation of the innate immune response, e.g., for tissue preservation during sterile injury such as myocardial infarction or to promote tissue resolution of inflammation and potentially tissue regeneration and repair.
Collapse
Affiliation(s)
- Naveed Akbar
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.P.); (R.P.C.)
- Correspondence:
| | - Daan Paget
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.P.); (R.P.C.)
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Robin P. Choudhury
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.P.); (R.P.C.)
| |
Collapse
|
31
|
Gurunathan S, Kang MH, Qasim M, Khan K, Kim JH. Biogenesis, Membrane Trafficking, Functions, and Next Generation Nanotherapeutics Medicine of Extracellular Vesicles. Int J Nanomedicine 2021; 16:3357-3383. [PMID: 34040369 PMCID: PMC8140893 DOI: 10.2147/ijn.s310357] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/25/2021] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membrane-limited vesicles and multi-signal messengers loaded with biomolecules. Exosomes and ectosomes are two different types of EVs generated by all cell types. Their formation depends on local microdomains assembled in endocytic membranes for exosomes and in the plasma membrane for ectosomes. Further, EV release is a fundamental process required for intercellular communication in both normal physiology and pathological conditions to transmit/exchange bioactive molecules to recipient cells and the extracellular environment. The unique structure and composition of EVs enable them to serve as natural nanocarriers, and their physicochemical properties and biological functions can be used to develop next-generation nano and precision medicine. Knowledge of the cellular processes that govern EVs biology and membrane trafficking is essential for their clinical applications. However, in this rapidly expanding field, much remains unknown regarding EV origin, biogenesis, cargo sorting, and secretion, as well as EV-based theranostic platform generation. Hence, we present a comprehensive overview of the recent advances in biogenesis, membrane trafficking, and functions of EVs, highlighting the impact of nanoparticles and oxidative stress on EVs biogenesis and release and finally emphasizing the role of EVs as nanotherapeutic agents.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Muhammad Qasim
- Center of Bioengineering and Nanomedicine, Department of Food Science, University of Otago, Dunedin, 9054, New Zealand
| | - Khalid Khan
- Science and Technology KPK, Peshawar, Pakistan
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| |
Collapse
|
32
|
Zhang J, Zhang Y, Mo F, Patel G, Butterworth K, Shao C, Prise KM. The Roles of HIF-1α in Radiosensitivity and Radiation-Induced Bystander Effects Under Hypoxia. Front Cell Dev Biol 2021; 9:637454. [PMID: 33869184 PMCID: PMC8044822 DOI: 10.3389/fcell.2021.637454] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022] Open
Abstract
Radiation-induced bystander effects (RIBE) may have potential implications for radiotherapy, yet the radiobiological impact and underlying mechanisms in hypoxic tumor cells remain to be determined. Using two human tumor cell lines, hepatoma HepG2 cells and glioblastoma T98G cells, the present study found that under both normoxic and hypoxic conditions, increased micronucleus formation and decreased cell survival were observed in non-irradiated bystander cells which had been co-cultured with X-irradiated cells or treated with conditioned-medium harvested from X-irradiated cells. Although the radiosensitivity of hypoxic tumor cells was lower than that of aerobic cells, the yield of micronucleus induced in bystander cells under hypoxia was similar to that measured under normoxia indicating that RIBE is a more significant factor in overall radiation damage of hypoxic cells. When hypoxic cells were treated with dimethyl sulfoxide (DMSO), a scavenger of reactive oxygen species (ROS), or aminoguanidine (AG), an inhibitor of nitric oxide synthase (NOS), before and during irradiation, the bystander response was partly diminished. Furthermore, when only hypoxic bystander cells were pretreated with siRNA hypoxia-inducible factor-1α (HIF-1α), RIBE were decreased slightly but if irradiated cells were treated with siRNA HIF-1α, hypoxic RIBE decreased significantly. In addition, the expression of HIF-1α could be increased in association with other downstream effector molecules such as glucose transporter 1 (GLUT-1), vascular endothelial growth factor (VEGF), and carbonic anhydrase (CA9) in irradiated hypoxic cells. However, the expression of HIF-1α expression in bystander cells was decreased by a conditioned medium from isogenic irradiated cells. The current results showed that under hypoxic conditions, irradiated HepG2 and T98G cells showed reduced radiosensitivity by increasing the expression of HIF-1α and induced a syngeneic bystander effect by decreasing the expression of HIF-1α and regulating its downstream target genes in both the irradiated or bystander cells.
Collapse
Affiliation(s)
- Jianghong Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Yuhong Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Fang Mo
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Gaurang Patel
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Karl Butterworth
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Kevin M Prise
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
33
|
Oxidative Stress and Gene Expression Modifications Mediated by Extracellular Vesicles: An In Vivo Study of the Radiation-Induced Bystander Effect. Antioxidants (Basel) 2021; 10:antiox10020156. [PMID: 33494540 PMCID: PMC7911176 DOI: 10.3390/antiox10020156] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/27/2022] Open
Abstract
Radiation-induced bystander effect is a biological response in nonirradiated cells receiving signals from cells exposed to ionising radiation. The aim of this in vivo study was to analyse whether extracellular vesicles (EVs) originating from irradiated mice could induce modifications in the redox status and expression of radiation-response genes in bystander mice. C57BL/6 mice were whole-body irradiated with 0.1-Gy and 2-Gy X-rays, and EVs originating from mice irradiated with the same doses were injected into naïve, bystander mice. Lipid peroxidation in the spleen and plasma reactive oxygen metabolite (ROM) levels increased 24 h after irradiation with 2 Gy. The expression of antioxidant enzyme genes and inducible nitric oxide synthase 2 (iNOS2) decreased, while cell cycle arrest-, senescence- and apoptosis-related genes were upregulated after irradiation with 2 Gy. In bystander mice, no significant alterations were observed in lipid peroxidation or in the expression of genes connected to cell cycle arrest, senescence and apoptosis. However, there was a systemic increase in the circulating ROM level after an intravenous EV injection, and EVs originating from 2-Gy-irradiated mice caused a reduced expression of antioxidant enzyme genes and iNOS2 in bystander mice. In conclusion, we showed that ionising radiation-induced alterations in the cellular antioxidant system can be transmitted in vivo in a bystander manner through EVs originating from directly irradiated animals.
Collapse
|
34
|
Gebremedhn S, Ali A, Gad A, Prochazka R, Tesfaye D. Extracellular Vesicles as Mediators of Environmental and Metabolic Stress Coping Mechanisms During Mammalian Follicular Development. Front Vet Sci 2020; 7:602043. [PMID: 33330723 PMCID: PMC7710682 DOI: 10.3389/fvets.2020.602043] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles are evolutionarily conserved nano-sized phospholipid membraned structures and released from virtually all types of cells into the extracellular space. Their ability to carry various molecular cargos (mRNA, miRNA, proteins, and lipids) from one cell to the other to exert functional impact on the target cells enables them to play a significant role in cell to cell communication during follicular development. As the molecular signals carried by extracellular vesicles reflect the physiological status of the cells of origin, they are expected to mediate any effect of environmental or metabolic stress on the follicualr cells and the growing oocyte. Recent studies have evidenced that reproductive cells exposed to various environmental stressors (heat and oxidative stress) released extracellular vesicles enriched with mRNA and miRNA associated with stress response mechanisms. Moreover, the metabolic status of post-calving cows could be well-reflected in the follicular extracellular vesicle's miRNA profile, which signified the potential role of extracellular cellular vesicle molecular signals in mediating the effect of metabolic stress on follicular and oocyte development. In the present review, the potential role of extracellular vesicles in mediating the effect of environmental and metabolic stress in various reproductive cells and oocytes are thoroughly discussed Moreover, considering the importance of extracellular vesicles in shuttling protective or rescuing molecular signals during stress, their potential usage as means of targeted delivery of molecules to mitigate the effect of stress on oocytes are addressed as the focus of future research.
Collapse
Affiliation(s)
- Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States.,Department of Animal, Rangeland and Wildlife Sciences, Mekelle University, Mekelle, Ethiopia
| | - Asghar Ali
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Ahmed Gad
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia.,Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Radek Prochazka
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States.,Department of Animal, Rangeland and Wildlife Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
35
|
Li Z, Deng J, Sun J, Ma Y. Hyperthermia Targeting the Tumor Microenvironment Facilitates Immune Checkpoint Inhibitors. Front Immunol 2020; 11:595207. [PMID: 33240283 PMCID: PMC7680736 DOI: 10.3389/fimmu.2020.595207] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have ushered in a new era of cancer therapy; however, ICIs are only effective in selective patients. The efficacy of ICIs is closely related to the tumor microenvironment. Fever for a long time was thought to directly regulate the immune response, and artificial “fever” from hyperthermia modulates the tumor immune microenvironment by providing danger signals with heat shock proteins (HSPs) as well as subsequent activation of immune systems. Encouraging results have been achieved in preclinical studies focused on potential synergetic effects by combining hyperthermia with ICIs. In this review, we summarized a cluster of immune-related factors that not only make hyperthermia a treatment capable of defending against cancer but also make hyperthermia a reliable treatment that creates a type I-like tumor microenvironment (overexpression of PD-L1 and enrichment of tumor infiltrating lymphocytes) in complementary for the enhancement of the ICIs. Then we reviewed recent preclinical data of the combination regimens involving hyperthermia and ICIs that demonstrated the combined efficacy and illustrated possible approaches to further boost the effectiveness of this combination.
Collapse
Affiliation(s)
- Zihui Li
- Oncology Department, The Third People's Hospital of Hubei Province, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Jie Deng
- Oncology Department, The Third People's Hospital of Hubei Province, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Jianhai Sun
- Oncology Department, The Third People's Hospital of Hubei Province, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Yanling Ma
- Oncology Department, The Third People's Hospital of Hubei Province, Affiliated Hospital of Jianghan University, Wuhan, China
| |
Collapse
|
36
|
Zhang YM, Zhang LY, Zhou H, Li YY, Wei KX, Li CH, Zhou T, Wang JF, Wei WJ, Hua JR, He Y, Hong T, Liu YQ. Astragalus polysaccharide inhibits radiation-induced bystander effects by regulating apoptosis in Bone Mesenchymal Stem Cells (BMSCs). Cell Cycle 2020; 19:3195-3207. [PMID: 33121344 DOI: 10.1080/15384101.2020.1838793] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The purpose of this study was to investigate the effects of astragalus polysaccharides (APS) on the proliferation and apoptosis of bone marrow mesenchymal stem cells (BMSCs) induced by X-ray radiation-induced A549 cells bystander effect (RIBE), and to explore their mechanisms. In this study, APS increased the reduced cell proliferation rate induced by RIBE and inhibiting the apoptosis of bystander cells. In terms of mechanism, APS up-regulates the proteins Bcl-2, Bcl-xl, and down-regulates the proteins Bax and Bak, which induces a decrease in mitochondrial membrane potential, which induces the release of Cyt-c and AIF, which leads to caspase-dependent and caspase-independent pathway to cause apoptosis. In addition, we believe that ROS may be the main cause of these protein changes. APS can inhibit the generation of ROS in bystander cells and thus inhibit the activation of the mitochondrial pathway, further preventing cellular damage caused by RIBE.
Collapse
Affiliation(s)
- Yi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Heng Zhou
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Yang-Yang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Kong-Xi Wei
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Cheng-Hao Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Ju-Fang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Wen-Jun Wei
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Jun-Rui Hua
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences , Lanzhou, China
| | - Yun He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Tao Hong
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine , Lanzhou, China
| |
Collapse
|
37
|
Baxter AA. Stoking the Fire: How Dying Cells Propagate Inflammatory Signalling through Extracellular Vesicle Trafficking. Int J Mol Sci 2020; 21:ijms21197256. [PMID: 33019535 PMCID: PMC7583891 DOI: 10.3390/ijms21197256] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Communication between dying cells and their environment is a critical process that promotes tissue homeostasis during normal cellular turnover, whilst during disease settings, it can contribute to inflammation through the release of intracellular factors. Extracellular vesicles (EVs) are a heterogeneous class of membrane-bound cell-derived structures that can engage in intercellular communication via the trafficking of bioactive molecules between cells and tissues. In addition to the well-described functions of EVs derived from living cells, the ability of dying cells to release EVs capable of mediating functions on target cells or tissues is also of significant interest. In particular, during inflammatory settings such as acute tissue injury, infection and autoimmunity, the EV-mediated transfer of proinflammatory cargo from dying cells is an important process that can elicit profound proinflammatory effects in recipient cells and tissues. Furthermore, the biogenesis of EVs via unique cell-death-associated pathways has also been recently described, highlighting an emerging niche in EV biology. This review outlines the mechanisms and functions of dying-cell-derived EVs and their ability to drive inflammation during various modes of cell death, whilst reflecting on the challenges and knowledge gaps in investigating this subgenre of extracellular vesicles research.
Collapse
Affiliation(s)
- Amy A Baxter
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
38
|
Gebremedhn S, Gad A, Aglan HS, Laurincik J, Prochazka R, Salilew-Wondim D, Hoelker M, Schellander K, Tesfaye D. Extracellular vesicles shuttle protective messages against heat stress in bovine granulosa cells. Sci Rep 2020; 10:15824. [PMID: 32978452 PMCID: PMC7519046 DOI: 10.1038/s41598-020-72706-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/31/2020] [Indexed: 01/15/2023] Open
Abstract
Elevated summer temperature is reported to be the leading cause of stress in dairy and beef cows, which negatively affects various reproductive functions. Follicular cells respond to heat stress (HS) by activating the expression of heat shock family proteins (HSPs) and other antioxidants. HS is reported to negatively affect the bi-directional communication between the follicular cells and the oocyte, which is partly mediated by follicular fluid extracellular vesicles (EVs) released from surrounding cells. As carriers of bioactive molecules (DNA, RNA, protein, and lipids), the involvement of EVs in mediating the stress response in follicular cells is not fully understood. Here we used an in vitro model to decipher the cellular and EV-coupled miRNAs of bovine granulosa cells in response to HS. Moreover, the protective role of stress-related EVs against subsequent HS was assessed. For this, bovine granulosa cells from smaller follicles were cultured in vitro and after sub-confluency, cells were either kept at 37 °C or subjected to HS (42 °C). Results showed that granulosa cells exposed to HS increased the accumulation of ROS, total oxidized protein, apoptosis, and the expression of HSPs and antioxidants, while the viability of cells was reduced. Moreover, 14 and 6 miRNAs were differentially expressed in heat-stressed granulosa cells and the corresponding EVs, respectively. Supplementation of stress-related EVs in cultured granulosa cells has induced adaptive response to subsequent HS. However, this potential was not pronounced when the cells were kept under 37 °C. Taking together, EVs generated from granulosa cells exposed to HS has the potential to shuttle bioactive molecules to recipient cells and make them robust to subsequent HS.
Collapse
Affiliation(s)
- Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 1351 Rampart Rd, Fort Collins, CO, 80525, USA.,Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany.,Department of Animal, Rangeland and Wildlife Sciences, Mekelle University, Mekelle, Ethiopia
| | - Ahmed Gad
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic.,Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Hoda Samir Aglan
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Jozef Laurincik
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic.,Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Radek Prochazka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic
| | - Dessie Salilew-Wondim
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Michael Hoelker
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Karl Schellander
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 1351 Rampart Rd, Fort Collins, CO, 80525, USA. .,Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany. .,Department of Animal, Rangeland and Wildlife Sciences, Mekelle University, Mekelle, Ethiopia.
| |
Collapse
|
39
|
Yuan P, Hu X, Zhou Q. The nanomaterial-induced bystander effects reprogrammed macrophage immune function and metabolic profile. Nanotoxicology 2020; 14:1137-1155. [PMID: 32916084 DOI: 10.1080/17435390.2020.1817598] [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: 01/08/2023]
Abstract
Bystander effects in biological systems are the responses shown by nontargeted neighboring cells, and critical to the bio-nano interface interactions. In addition to direct effects, bystander effects also determine the design, applications and safety of nanomaterials, although the related information of nanomaterial-induced bystander effects remain largely unknown. A coculture system of A549 and THP-1 was established to mimic the lung microenvironment to study the bystander effects of WS2 nanosheets (representative transition-metal dichalcogenide nanosheets) on microenvironment macrophages during the inhalation exposure or the nanomaterial biomedical application in the lung. Lung cells exposed to WS2 nanosheet resulted in an increase in reactive oxygen species and the depolarization of mitochondrial membrane potential in neighboring macrophages. Bystander exposure also induced macrophage polarization toward the anti-inflammatory M2 phenotype, which is adverse to disease therapy. Metabolomics showed that WS2 nanosheets disturbed the energy metabolism and amino acid metabolism of macrophages, consistent with the metabolic characteristics of M2 macrophages. Nitric oxide-transforming growth factor-β1 played an important mediator in the bystander effects. Importantly, WS2 nanosheet bystander exposure affected macrophage phagocytosis and migration and altered the macrophage immune response to endotoxin. This study improves the current understanding of bio-nano interactions and highlights the importance of neighboring cell responses, allowing us to use the maximum benefits of nanomaterials while limiting their adverse bystander effects.
Collapse
Affiliation(s)
- Peng Yuan
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, China.,School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Qixing Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| |
Collapse
|
40
|
The influence of secreted factors and extracellular vesicles in ovarian cancer metastasis. EJC Suppl 2020; 15:38-48. [PMID: 33240441 PMCID: PMC7573474 DOI: 10.1016/j.ejcsup.2019.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/02/2019] [Accepted: 09/15/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer cells mainly metastasise within the peritoneal cavity, the lethal consequence of tumour progression in this cancer type. Classically, changes in tumour cells, such as epithelial to mesenchymal transition, involve the down-regulatinon of E-cadherin, activation of extracellular proteases and integrin-mediated adhesion. However, our current understanding of ovarian tumour progression suggests the implication of both intrinsic and extrinsic factors. It has been proposed that ovarian cancer metastases are a consequence of the crosstalk between cancer cells and the tumour microenvironment by soluble factors and extracellular vesicles. Characterisation of the alterations in both the tumour cells and the surrounding microenvironment has emerged as a new research field to understand ovarian cancer metastasis. In this mini review, we will summarise the most recent findings, focusing our attention on the role of secreted factors and extracellular vesicles in ovarian cancer metastasis. During ovarian cancer metastasis, tumour cells metastasise in the mesothelium as primarily ‘soil’ for ovarian cancer ‘seeds’. Soluble factors and extracellular vesicles secreted by tumor cells are involved in the generation of the pre-metastatic niche. Cancer-associated fibroblasts (CAFs) represent the majority of stromal cells in various types of human carcinoma, including ovarian cancer. Analysis of early metastasis to the omentum indicates that ovarian cancer cells rely on the interaction with immune cells such as macrophages. Liquid biopsy analyses in ovarian cancer may help to define novel biomarkers improving patient survival and reduce lethality.
Collapse
|
41
|
Sheybani ND, Batts AJ, Mathew AS, Thim EA, Price RJ. Focused Ultrasound Hyperthermia Augments Release of Glioma-derived Extracellular Vesicles with Differential Immunomodulatory Capacity. Theranostics 2020; 10:7436-7447. [PMID: 32642004 PMCID: PMC7330848 DOI: 10.7150/thno.46534] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Increasing evidence points to the critical role of extracellular vesicles (EVs) as molecular parcels that carry a diverse array of bioactive payloads for coordination of complex intracellular signaling. Focused ultrasound (FUS) hyperthermia is a technique for non-invasive, non-ionizing sublethal heating of cells in a near-instantaneous manner; while it has been shown to improve drug delivery and immunological recognition of tumors, its impact on EVs has not been explored to date. The goal of this study was to determine whether FUS impacts the release, proteomic profile, and immune-activating properties of tumor-derived EVs. Methods: Monolayered murine glioma cells were seeded within acoustically transparent cell culture chambers, and FUS hyperthermia was applied to achieve complete coverage of the chamber. Glioma-derived EVs (GEVs) were isolated for characterization by Nanoparticle Tracking Analysis, cryo-electron microscopy and mass spectrometry. An in vitro experimental setup was designed to further dissect the impact of GEVs on innate inflammation; immortalized murine dendritic cells (DCs) were pulsed with GEVs (either naïve or FUS hyperthermia-exposed) and assayed for production of IL-12p70, an important regulator of DC maturation and T helper cell polarization toward the interferon-γ-producing type 1 phenotype. Results: We confirmed that FUS hyperthermia significantly augments GEV release (by ~46%) as well as shifts the proteomic profile of these GEVs. Such shifts included enrichment of common EV-associated markers, downregulation of markers associated with cancer progression and resistance and modulation of inflammation-associated markers. When DCs were pulsed with GEVs, we noted that naïve GEVs suppressed IL-12p70 production by DCs in a GEV dose-dependent manner. In contrast, GEVs from cells exposed to FUS hyperthermia promoted a significant upregulation in IL-12p70 production by DCs, consistent with a pro-inflammatory stimulus. Conclusion: FUS hyperthermia triggers release of proteomically distinct GEVs that are capable of facilitating an important component of innate immune activation, lending both to a potential mechanism by which FUS interfaces with the tumor-immune landscape and to a role for GEV-associated biomarkers in monitoring response to FUS.
Collapse
Affiliation(s)
- Natasha D. Sheybani
- Department of Biomedical Engineering; University of Virginia, Charlottesville, VA 22908
| | - Alec J. Batts
- Department of Biomedical Engineering; University of Virginia, Charlottesville, VA 22908
| | - Alexander S. Mathew
- Department of Biomedical Engineering; University of Virginia, Charlottesville, VA 22908
| | - E. Andrew Thim
- Department of Biomedical Engineering; University of Virginia, Charlottesville, VA 22908
| | - Richard J. Price
- Department of Biomedical Engineering; University of Virginia, Charlottesville, VA 22908
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
42
|
Wiest EF, Zubair AC. Challenges of manufacturing mesenchymal stromal cell-derived extracellular vesicles in regenerative medicine. Cytotherapy 2020; 22:606-612. [PMID: 32532592 DOI: 10.1016/j.jcyt.2020.04.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022]
Abstract
The field of regenerative medicine has expanded greatly in the past decade, with more than 1000 current clinical trials involving mesenchymal stromal cell (MSC) treatment. Multiple recent publications have demonstrated that the beneficial effects from MSCs are not simply due to engraftment into the target organ as classically thought but rather are largely attributable to the release of paracrine factors including cytokines, growth factors and extracellular vesicles (EVs). These EVs contain miRNAs, free fatty acids and proteins that promote regeneration, proliferation and cell function and improve inflammation. Although EVs have shown promising results in animal studies, there are many obstacles to the manufacturing of EVs for clinical applications. This review discusses challenges associated with the manufacturing of clinical-grade EVs in regard to identity, purity, reproducibility, sterility, storage, potency and safety. We discuss currently employed methods and approaches for developing clinical Good Manufacturing Practices (GMP)-grade EVs and the limitations for each. We further discuss the best approaches to overcome the current hurdles in developing clinical GMP-grade EVs.
Collapse
Affiliation(s)
- Elani F Wiest
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Abba C Zubair
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA.
| |
Collapse
|
43
|
Moertl S, Buschmann D, Azimzadeh O, Schneider M, Kell R, Winkler K, Tapio S, Hornhardt S, Merl-Pham J, Pfaffl MW, Atkinson MJ. Radiation Exposure of Peripheral Mononuclear Blood Cells Alters the Composition and Function of Secreted Extracellular Vesicles. Int J Mol Sci 2020; 21:ijms21072336. [PMID: 32230970 PMCID: PMC7178185 DOI: 10.3390/ijms21072336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/24/2022] Open
Abstract
Normal tissue toxicity is a dose-limiting factor in radiation therapy. Therefore, a detailed understanding of the normal tissue response to radiation is necessary to predict the risk of normal tissue toxicity and to development strategies for tissue protection. One component of normal tissue that is continuously exposed during therapeutic irradiation is the circulating population of peripheral blood mononuclear cells (PBMC). PBMCs are highly sensitive to ionizing radiation (IR); however, little is known about how IR affects the PBMC response on a systemic level. It was the aim of this study to investigate whether IR was capable to induce changes in the composition and function of extracellular vesicles (EVs) secreted from PBMCs after radiation exposure to different doses. Therefore, whole blood samples from healthy donors were exposed to X-ray radiation in the clinically relevant doses of 0, 0.1, 2 or 6 Gy and PBMC-secreted EVs were isolated 72 h later. Proteome and miRNome analysis of EVs as well as functional studies were performed. Secreted EVs showed a dose-dependent increase in the number of significantly deregulated proteins and microRNAs. For both, proteome and microRNA data, principal component analysis showed a dose-dependent separation of control and exposed groups. Integrated pathway analysis of the radiation-regulated EV proteins and microRNAs consistently predicted an association of deregulated molecules with apoptosis, cell death and survival. Functional studies identified endothelial cells as an efficient EV recipient system, in which irradiation of recipient cells further increased the uptake. Furthermore an apoptosis suppressive effect of EVs from irradiated PBMCs in endothelial recipient cells was detected. In summary, this study demonstrates that IR modifies the communication between PBMCs and endothelial cells. EVs from irradiated PBMC donors were identified as transmitters of protective signals to irradiated endothelial cells. Thus, these data may lead to the discovery of biomarker candidates for radiation dosimetry and even more importantly, they suggest EVs as a novel systemic communication pathway between irradiated normal, non-cancer tissues.
Collapse
Affiliation(s)
- Simone Moertl
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
- Federal Office for Radiation Protection, 85764 Oberschleißheim, Germany; (S.H.); (M.W.P.)
- Correspondence:
| | - Dominik Buschmann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany;
| | - Omid Azimzadeh
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Michael Schneider
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Rosemarie Kell
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Klaudia Winkler
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Soile Tapio
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
| | - Sabine Hornhardt
- Federal Office for Radiation Protection, 85764 Oberschleißheim, Germany; (S.H.); (M.W.P.)
| | - Juliane Merl-Pham
- Helmholtz Zentrum München, German Research Center for Environmental Health, Research Unit Protein Science, 80939 München, Germany;
| | - Michael W. Pfaffl
- Federal Office for Radiation Protection, 85764 Oberschleißheim, Germany; (S.H.); (M.W.P.)
| | - Michael J. Atkinson
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany; (O.A.); (M.S.); (R.K.); (K.W.); (S.T.); (M.J.A.)
- Chair of Radiation Biology, Technical University of Munich, 80333 Munich, Germany
| |
Collapse
|
44
|
Tesfaye D, Hailay T, Salilew-Wondim D, Hoelker M, Bitseha S, Gebremedhn S. Extracellular vesicle mediated molecular signaling in ovarian follicle: Implication for oocyte developmental competence. Theriogenology 2020; 150:70-74. [PMID: 32088041 DOI: 10.1016/j.theriogenology.2020.01.075] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022]
Abstract
The bidirectional communication between the oocyte and the companion somatic cells in the follicular environment is known to be mediated by either a direct communication via gap junction or transzonal projections or indirectly through endocrine, paracrine and autocrine signaling factors. Extracellular vesicles (EVs), which are found in various biological fluids, including follicular fluid (FF) are known to play important roles in mediating the communication between the oocyte and the surrounding somatic cells through shuttling bioactive molecules to facilitate follicular growth and oocyte maturation. As vesicles in the extracellular space are known to reflect the physiological status of the donor or the releasing cells, molecules carried by the EVs in the follicular environment could be markers of the internal and external stressors. EVs exhibit greater degree of heterogeneity in their size, biogenesis and the bioactive molecule they carry. The process of biogenesis of EVs is known to be regulated by several proteins associated with the endosomal sorting complex required for transport (ESCRT) proteins. The type of EVs and surface proteins markers vary according to the type of protein involved in their biogenesis. EVs are recently reported to play indispensable role in promoting cell-to-cell communication during follicular growth. Recent advancements in EV research opened the possibilities to load EVs with specific molecules like miRNA, siRNA, CRISPR-cas9 complex and protein, which showed a new horizon for their application in therapeutics. The present review explores the biogenesis, the role and the future prospects of EVs with a special emphasis given to follicular growth and oocyte maturation.
Collapse
Affiliation(s)
- Dawit Tesfaye
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory (ARBL), Colorado State University, Fort Collins, CO, USA.
| | - Tsige Hailay
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, Bonn, Germany
| | - Dessie Salilew-Wondim
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, Bonn, Germany
| | - Michael Hoelker
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, Bonn, Germany
| | - Simret Bitseha
- Hawassa University, College of Agriculture, Department of Animal Sciences, Hawassa, Ethiopia
| | - Samuel Gebremedhn
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory (ARBL), Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
45
|
Purity Determines the Effect of Extracellular Vesicles Derived from Mesenchymal Stromal Cells. Cells 2020; 9:cells9020422. [PMID: 32059497 PMCID: PMC7072280 DOI: 10.3390/cells9020422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) have been recently identified as vital components of cell-based therapies based on the observation that conditioned media from cultured stromal cells reproduce some of the beneficial effects of intact cells. In order to obtain clinically active EVs derived from Mesenchymal Stromal Cells (MSCs) different procedures have been reported in the literature. Usually, non-confluent cells are incubated with culture medium for 48 h either with EV-depleted Fetal Bovine Serum (FBS) or without FBS. Our aim was to compare the effects of EVs isolated by ultracentrifugation from human umbilical cord MSC conditioned media obtained using these two conditions: with EV-depleted FBS (UC) or without FBS (UCw/o) on the mRNA expression levels of extracellular matrix related genes using the mouse chondrogenic cell line ATDC-5. We observed a deleterious effect on chondrogenic cells treated with UCw/o, showing higher mRNA expression levels of different metalloproteinases and decorin (Dcn) and lower collagen (Col1a1 and Col2a1) and aggrecan (Acan) mRNA levels. To elucidate whether this deleterious effect was induced by the EVs or by any proteins co-purified in the EV pellet, we used size exclusion chromatography (SEC) to further purify the EV pellet, obtaining an EV enriched fraction (EV or EVw/o) and a protein enriched fraction (Prot or Protw/o). Our results pointed that the negative effect on the chondrogenic cell line was due to the contaminant proteins coisolated with the EVs by ultracentrifugation and not from the EVs themselves. Thus, these results highlight the importance of working with well purified EV preparations to specifically achieve their therapeutic effect.
Collapse
|
46
|
Melling GE, Carollo E, Conlon R, Simpson JC, Carter DRF. The Challenges and Possibilities of Extracellular Vesicles as Therapeutic Vehicles. Eur J Pharm Biopharm 2019; 144:50-56. [DOI: 10.1016/j.ejpb.2019.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/31/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022]
|
47
|
Taqi MO, Saeed‐Zidane M, Gebremedhn S, Salilew‐Wondim D, Khdrawy O, Rings F, Neuhoff C, Hoelker M, Schellander K, Tesfaye D. Sexual dimorphic expression and release of transcription factors in bovine embryos exposed to oxidative stress. Mol Reprod Dev 2019; 86:2005-2019. [DOI: 10.1002/mrd.23272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Mohamed O. Taqi
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Mohammed Saeed‐Zidane
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Samuel Gebremedhn
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Dessie Salilew‐Wondim
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Omar Khdrawy
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Franca Rings
- Teaching and Research Station Frankenforst, Faculty of AgricultureUniversity of Bonn Königswinter Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| | - Christiane Neuhoff
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Michael Hoelker
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
- Teaching and Research Station Frankenforst, Faculty of AgricultureUniversity of Bonn Königswinter Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| | - Karl Schellander
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| | - Dawit Tesfaye
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| |
Collapse
|
48
|
Schuldner M, Dörsam B, Shatnyeva O, Reiners KS, Kubarenko A, Hansen HP, Finkernagel F, Roth K, Theurich S, Nist A, Stiewe T, Paschen A, Knittel G, Reinhardt HC, Müller R, Hallek M, von Strandmann EP. Exosome-dependent immune surveillance at the metastatic niche requires BAG6 and CBP/p300-dependent acetylation of p53. Theranostics 2019; 9:6047-6062. [PMID: 31534536 PMCID: PMC6735508 DOI: 10.7150/thno.36378] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/15/2019] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles released by tumor cells contribute to the reprogramming of the tumor microenvironment and interfere with hallmarks of cancer including metastasis. Notably, melanoma cell-derived EVs are able to establish a pre-metastatic niche in distant organs, or on the contrary, exert anti-tumor activity. However, molecular insights into how vesicles are selectively packaged with cargo defining their specific functions remain elusive. Methods: Here, we investigated the role of the chaperone Bcl2-associated anthogene 6 (BAG6, synonym Bat3) for the formation of pro- and anti-tumor EVs. EVs collected from wildtype cells and BAG6-deficient cells were characterized by mass spectrometry and RNAseq. Their tumorigenic potential was analyzed using the B-16V transplantation mouse melanoma model. Results: We demonstrate that EVs from B-16V cells inhibit lung metastasis associated with the mobilization of Ly6Clow patrolling monocytes. The formation of these anti-tumor-EVs was dependent on acetylation of p53 by the BAG6/CBP/p300-acetylase complex, followed by recruitment of components of the endosomal sorting complexes required for transport (ESCRT) via a P(S/T)AP double motif of BAG6. Genetic ablation of BAG6 and disruption of this pathway led to the release of a distinct EV subtype, which failed to suppress metastasis but recruited tumor-promoting neutrophils to the pre-metastatic niche. Conclusion: We conclude that the BAG6/CBP/p300-p53 axis is a key pathway directing EV cargo loading and thus a potential novel microenvironmental therapeutic target.
Collapse
|
49
|
Ionizing Radiation Increases the Activity of Exosomal Secretory Pathway in MCF-7 Human Breast Cancer Cells: A Possible Way to Communicate Resistance against Radiotherapy. Int J Mol Sci 2019; 20:ijms20153649. [PMID: 31349735 PMCID: PMC6696324 DOI: 10.3390/ijms20153649] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022] Open
Abstract
Radiation therapy, which applies high-energy rays, to eradicate tumor cells, is considered an essential therapy for the patients with breast cancer. Most tumor cells secrete exosomes, which are involved in cell-to-cell communication in tumor tissue and contribute therapeutic resistance and promote tumor aggressiveness. Here, we investigated the effect of clinically applicable doses of X-ray irradiation (2, 4, 6, 8, 10 Gy) on the dynamics of the exosomes' activity in MCF-7 breast cancer cells. Survival and apoptosis rate of cells against X-ray doses was examined using MTT and flow cytometry assays, respectively. Whereas, the levels of reactive oxygen species (ROS) in the X-ray-treated cells were detected by fluorometric method. The mRNA levels of vital genes involved in exosome biogenesis and secretion including Alix, Rab11, Rab27a, Rab27b, TSPA8, and CD63 were measured by real-time PCR. The protein level of CD63 was examined by Western blotting. Additionally, exosomes were characterized by monitoring acetylcholinesterase activity, transmission electron microscopy, size determination, and zeta potential. The result showed that in comparison with control group cell survival and the percentage of apoptotic cells as well as amount of ROS dose-dependently decreased and increased in irradiated cells respectively (p < 0.05). The expression level of genes including Alix, Rab27a, Rab27b, TSPA8, and CD63 as well as the protein level of CD63 upraised according to an increase in X-ray dose (p < 0.05). We found that concurrent with an increasing dose of X-ray, the acetylcholinesterase activity, size, and zeta-potential values of exosomes from irradiated cells increased (p < 0.05). Data suggest X-ray could activate exosome biogenesis and secretion in MCF-7 cells in a dose-dependent way, suggesting the therapeutic response of cells via ROS and exosome activity.
Collapse
|
50
|
Different Types of Cellular Stress Affect the Proteome Composition of Small Extracellular Vesicles: A Mini Review. Proteomes 2019; 7:proteomes7020023. [PMID: 31126168 PMCID: PMC6631412 DOI: 10.3390/proteomes7020023] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/14/2019] [Accepted: 05/19/2019] [Indexed: 12/27/2022] Open
Abstract
Extracellular vesicles (EVs) are well-known mediators of the cellular response to different stress factors, yet the exact mechanism of their action remains unclear. Hence, the characterization of their cargo, consisting of proteins, nucleic acids, and different classes of metabolites, helps to elucidate an understanding of their function in stress-related communication. The unexpected diversity and complexity of these vesicles requires the incorporation of multiple technologically advanced approaches in EV-oriented studies. This mini review focuses on the invaluable role of proteomics, especially mass spectrometry-based tools, in the investigation of the role of small EVs in their response to stress. Though relatively few experimental works address this issue to date, the available data indicate that stress conditions would affect the composition of protein cargo of vesicles released by stressed cells, as evidenced by the functional importance of such changes in the context of the response of recipient cells.
Collapse
|