51
|
Wang N, Pei B, Yuan X, Yi C, Wiredu Ocansey DK, Qian H, Mao F. Emerging roles of mesenchymal stem cell-derived exosomes in gastrointestinal cancers. Front Bioeng Biotechnol 2022; 10:1019459. [PMID: 36338118 PMCID: PMC9631450 DOI: 10.3389/fbioe.2022.1019459] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal tumours are the most common solid tumours, with a poor prognosis and remain a major challenge in cancer treatment. Mesenchymal stem cells (MSC) are multipotent stromal cells with the potential to differentiate into multiple cell types. Several studies have shown that MSC-derived exosomes have become essential regulators of intercellular communication in a variety of physiological and pathological processes. Notably, MSC-derived exosomes support or inhibit tumour progression in different cancers through the delivery of proteins, RNA, DNA, and bioactive lipids. Herein, we summarise current advances in MSC-derived exosomes in cancer research, with particular reference to their role in gastrointestinal tumour development. MSC-derived exosomes are expected to be a novel potential strategy for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Naijian Wang
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu, China
| | - Xinyi Yuan
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Hua Qian
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Hua Qian,
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
52
|
Guillaume VGJ, Ruhl T, Boos AM, Beier JP. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:394-406. [PMID: 35274703 PMCID: PMC9052412 DOI: 10.1093/stcltm/szac002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/22/2021] [Indexed: 11/14/2022] Open
Abstract
Adipose-derived stem or stromal cells (ASCs) possess promising potential in the fields of tissue engineering and regenerative medicine due to their secretory activity, their multilineage differentiation potential, their easy harvest, and their rich yield compared to other stem cell sources. After the first identification of ASCs in humans in 2001, the knowledge of their cell biology and cell characteristics have advanced, and respective therapeutic options were determined. Nowadays, ASC-based therapies are on the verge of translation into clinical practice. However, conflicting evidence emerged in recent years about the safety profile of ASC applications as they may induce tumor progression and invasion. Numerous in-vitro and in-vivo studies demonstrate a potential pro-oncogenic effect of ASCs on various cancer entities. This raises questions about the safety profile of ASCs and their broad handling and administration. However, these findings spark controversy as in clinical studies ASC application did not elevate tumor incidence rates, and other experimental studies reported an inhibitory effect of ASCs on different cancer cell types. This comprehensive review aims at providing up-to-date information about ASCs and cancer cell interactions, and their potential carcinogenesis and tumor tropism. The extracellular signaling activity of ASCs, the interaction of ASCs with the tumor microenvironment, and 3 major organ systems (the breast, the skin, and genitourinary system) will be presented with regard to cancer formation and progression.
Collapse
Affiliation(s)
- Vincent G J Guillaume
- Corresponding author: Vincent G. J. Guillaume, Resident Physician and Research Assistant, Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany. Tel: 0049-241-80-89700; Fax: 0241-80-82448;
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Anja M Boos
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
53
|
Yassine S, Alaaeddine N. Mesenchymal Stem Cell Exosomes and Cancer: Controversies and Prospects. Adv Biol (Weinh) 2021; 6:e2101050. [PMID: 34939371 DOI: 10.1002/adbi.202101050] [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] [Received: 06/17/2021] [Revised: 11/11/2021] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have displayed a novel therapeutic strategy for a wide range of diseases and conditions. Their secretome and exosome-based paracrine activity are considered as the main processes harboring their diverse therapeutic properties. Several investigations have examined the effects of MSC-derived exosomes on cancer growth, yet, controversial results have always emerged. Although MSC-derived exosomes are able to rigorously enforce the repression of cancer proliferation and progression, it is shown that MSCs exosomal activity displays numerous protumorigenic effects. This discrepancy over the dual effects of MSCs on cancer growth may be mediated by many factors including experimental design, stem cells origins, culture conditions, in addition to cancer-MSCs cross-talks. Despite the controversial effects of MSCs on carcinogenesis, scientists are able to overcome a number of obstacles by modifying MSCs to deliver antioncogenic miRNAs, anticancer drugs, and oncolytic viruses into tumor sites. This review discusses the controversial effects of MSC-derived exosomes on tumorigenesis, investigates the main causes that underlie this discrepancy, summarizes the pattern of engineered-MSCs, and finally highlights how future studies should advance the research in the field of MSCs-based cancer therapies in order to accelerate the transition from preclinical studies to clinical practice.
Collapse
Affiliation(s)
- Sirine Yassine
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, 1100, Lebanon
| | - Nada Alaaeddine
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, 1100, Lebanon
| |
Collapse
|
54
|
Abstract
Exosomes are nano-sized extracellular vesicles (30–160 nm diameter) with lipid bilayer membrane secrete by various cells that mediate the communication between cells and tissue, which contain a variety of non-coding RNAs, mRNAs, proteins, lipids and other functional substances. Adipose tissue is important energy storage and endocrine organ in the organism. Recent studies have revealed that adipose tissue-derived exosomes (AT-Exosomes) play a critical role in many physiologically and pathologically functions. Physiologically, AT-Exosomes could regulate the metabolic homoeostasis of various organs or cells including liver and skeletal muscle. Pathologically, they could be used in the treatment of disease and or that they may be involved in the progression of the disease. In this review, we describe the basic principles and methods of exosomes isolation and identification, as well as further summary the specific methods. Moreover, we categorize the relevant studies of AT-Exosomes and summarize the different components and biological functions of mammalian exosomes. Most importantly, we elaborate AT-Exosomes crosstalk within adipose tissue and their functions on other tissues or organs from the physiological and pathological perspective. Based on the above analysis, we discuss what remains to be discovered problems in AT-Exosomes studies and prospect their directions needed to be further explored in the future.
Collapse
Affiliation(s)
- Rui Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Tiantian Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Zhaozhao He
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Rui Cai
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| |
Collapse
|
55
|
Choi DW, Cho KA, Kim J, Lee HJ, Kim YH, Park JW, Woo SY. Extracellular vesicles from tonsil‑derived mesenchymal stromal cells show anti‑tumor effect via miR‑199a‑3p. Int J Mol Med 2021; 48:221. [PMID: 34676871 PMCID: PMC8559701 DOI: 10.3892/ijmm.2021.5054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are mesoderm‑originated adult SCs that possess multidirectional differentiation potential. MSCs migrate to injured tissue and secrete a range of paracrine factors that induce regeneration in damaged tissue and exert immune modulation. Because tumor progression is dependent on cross‑talk between the tumor and its microenvironment, MSCs also produce extracellular vesicles (EVs) that mediate information transfer in the tumor microenvironment. However, the effect of MSC‑derived EVs on tumor development and progression is still controversial. To date, tonsil‑derived MSCs (T‑MSCs) have been shown to possess all the defined characteristics of MSCs and show distinctive features of differential potential and immune modulation. To observe the effect of soluble mediators from T‑MSCs on tumor growth, human liver cancer cell line (HepG2) cells were injected into nude mice and HepG2 cell scratch migration assay was performed using conditioned medium (CM) of T‑MSCs. T‑MSC CM inhibited tumor growth and progression and it was hypothesized that EVs from T‑MSCs could inhibit tumor progression. microRNA (miRNA or miR) sequencing using five different origins of T‑MSC‑derived EVs was performed and highly expressed miRNAs, such as miR‑199a‑3p, miR‑214‑3p, miR‑199a‑5p and miR‑199b‑5p, were selected. T‑MSCs inhibited tumor growth and HepG2 cell migration, potentially via miR‑199a‑3p targeting CD151, integrin α3 and 6 in HepG2 cells.
Collapse
Affiliation(s)
- Da-Won Choi
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Jungwoo Kim
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Hyun-Ji Lee
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Yu-Hee Kim
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Jang-Won Park
- Department of Orthopaedic Surgery, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
56
|
Weng Z, Zhang B, Wu C, Yu F, Han B, Li B, Li L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J Hematol Oncol 2021; 14:136. [PMID: 34479611 PMCID: PMC8414028 DOI: 10.1186/s13045-021-01141-y] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membrane structures enclosing proteins, lipids, RNAs, metabolites, growth factors, and cytokines. EVs have emerged as essential intercellular communication regulators in multiple physiological and pathological processes. Previous studies revealed that mesenchymal stem cells (MSCs) could either support or suppress tumor progression in different cancers by paracrine signaling via MSC-derived EVs. Evidence suggested that MSC-derived EVs could mimic their parental cells, possessing pro-tumor and anti-tumor effects, and inherent tumor tropism. Therefore, MSC-derived EVs can be a cell-free cancer treatment alternative. This review discusses different insights regarding MSC-derived EVs' roles in cancer treatment and summarizes bioengineered MSC-derived EVs’ applications as safe and versatile anti-tumor agent delivery platforms. Meanwhile, current hurdles of moving MSC-derived EVs from bench to bedside are also discussed.
Collapse
Affiliation(s)
- Zhijie Weng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bowen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Comfort Care Dental Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Comfort Care Dental Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
57
|
Panda B, Sharma Y, Gupta S, Mohanty S. Mesenchymal Stem Cell-Derived Exosomes as an Emerging Paradigm for Regenerative Therapy and Nano-Medicine: A Comprehensive Review. Life (Basel) 2021; 11:life11080784. [PMID: 34440528 PMCID: PMC8399916 DOI: 10.3390/life11080784] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal Stem Cells are potent therapeutic candidates in the field of regenerative medicine, owing to their immunomodulatory and differentiation potential. However, several complications come with their translational application like viability, duration, and degree of expansion, long-term storage, and high maintenance cost. Therefore, drawbacks of cell-based therapy can be overcome by a novel therapeutic modality emerging in translational research and application, i.e., exosomes. These small vesicles derived from mesenchymal stem cells are emerging as new avenues in the field of nano-medicine. These nano-vesicles have caught the attention of researchers with their potency as regenerative medicine both in nanotherapeutics and drug delivery systems. In this review, we discuss the current knowledge in the biology and handling of exosomes, with their limitations and future applications. Additionally, we highlight current perspectives that primarily focus on their effect on various diseases and their potential as a drug delivery vehicle.
Collapse
|
58
|
Liu M, Yang Y, Zhao B, Yang Y, Wang J, Shen K, Yang X, Hu D, Zheng G, Han J. Exosomes Derived From Adipose-Derived Mesenchymal Stem Cells Ameliorate Radiation-Induced Brain Injury by Activating the SIRT1 Pathway. Front Cell Dev Biol 2021; 9:693782. [PMID: 34395427 PMCID: PMC8358610 DOI: 10.3389/fcell.2021.693782] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Objective Studies have shown that the therapeutic effects of mesenchymal stem cells (MSCs) are mediated in a paracrine manner, mainly through extracellular vesicles such as exosomes. Here, we designed a study to investigate whether exosomes derived from adipose-derived mesenchymal stem cells (ADMSC-Exos) had protective effects in a rat model of radiation-induced brain injury and in microglia. Methods Male adult Sprague-Dawley (SD) rats were randomly divided into three groups: the control group, the radiation group (30 Gy), and the radiation + exosomes group (30 Gy + 100 ug exosomes). Meanwhile, microglia were divided into four groups: the control group, the radiation group (10 Gy), the radiation + exosomes group (10 Gy + 4 ug exosomes), and radiation + exosomes + EX527 group (10 Gy + 4 ug exosomes + 100 nM EX527). Tissue samples and the levels of oxidative stress and inflammatory factors in each group were compared. Results Statistical analysis showed that after irradiation, ADMSC-Exos intervention in vivo significantly reduced the levels of caspase-3, malondialdehyde (MDA), 8-hydroxydeoxyguanosine (8-OHdG), tumor necrosis factor-α (TNF-α), interleukin-4 (IL-4), and promoted the recovery of superoxide dismutase (SOD), catalase (CAT), IL-4, and IL-10. Moreover, ADMSC-Exos intervention inhibited microglial infiltration and promoted the expression of SIRT1. Furthermore, the results in vitro showed that the above effects of ADMSC-Exos could be reversed by SIRT-1 inhibitor EX527. Conclusion This study demonstrated that ADMSC-Exos exerted protective effects against radiation-induced brain injury by reducing oxidative stress, inflammation and microglial infiltration via activating the SIRT1 pathway. ADMSC-Exos may serve as a promising therapeutic tool for radiation-induced brain injury.
Collapse
Affiliation(s)
- Mengdong Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Yunshu Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Bin Zhao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Yuefan Yang
- Department of Biomedical Engineering, Air Force Military Medical University, Xi'an, China.,Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| | - Guoxu Zheng
- State key laboratory of Cancer Biology, Department of Immunology, Air Force Military Medical University, Xi'an, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
59
|
Hamilton G, Teufelsbauer M. Adipose-derived stromal/stem cells and extracellular vesicles for cancer therapy. Expert Opin Biol Ther 2021; 22:67-78. [PMID: 34236014 DOI: 10.1080/14712598.2021.1954156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Mesenchymal stromal/stem cells (MSCs) hold great perspective for the therapy of a host of diseases due to regenerative and anti-inflammatory properties by differentiation into diverse cell populations, homing to damaged tissue regions, paracrine effects, and release of extracellular vesicles. AREAS COVERED This review describes the isolation, characterization, and potential use of MSCs and ADSCs for benign and malignant diseases. The MSCs may be administered as whole cells or in form of their secretome that is held responsible for most of their beneficial effects. A special constituent of the paracrine components are the extracellular vesicles (EVs) that carry a biologically potent cargo of proteins, cytokines, and RNA. EXPERT OPINION The applications of MSCs and ADSCs are amply documented and have been investigated in preclinical models and many unregulated and a few controlled trials. Larger numbers of MSCs and ADSCs can be obtained for allogeneic transfer but imply difficulties including perseverance of the cells in vivo and possible differentiation into harmful cell types. MSC-derived cell-free preparations are easier to handle and manufacture for various applications. Especially, with the help of bioreactors, EVs can be obtained in excessive numbers and preloaded or charged with proteins, cytokines, and regulatory RNA specimen to treat inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria.,Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Maryana Teufelsbauer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria.,Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
60
|
Eiro N, Fraile M, Fernández-Francos S, Sánchez R, Costa LA, Vizoso FJ. Importance of the origin of mesenchymal (stem) stromal cells in cancer biology: "alliance" or "war" in intercellular signals. Cell Biosci 2021; 11:109. [PMID: 34112253 PMCID: PMC8194017 DOI: 10.1186/s13578-021-00620-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play a central role in the intercellular signaling within the tumor microenvironment (TME), exchanging signals with cancer cells and tumor stromal cells, such as cancer-associated fibroblasts and inflammatory mononuclear cells. Research attributes both pro-tumor and anti-tumor actions to MSCs; however, evidence indicates that MSCs specific effect on the tumor depends on the source of the MSCs and the type of tumor. There are consistent data proving that MSCs from reproductive tissues, such as the uterus, umbilical cord or placenta, have potent anti-tumor effects and tropism towards tumor tissues. More interestingly, products derived from MSCs, such as secretome or extracellular vesicles, seem to reproduce the effects of their parental cells, showing a potential advantage for clinical treatments by avoiding the drawbacks associated with cell therapy. Given these perspectives, it appears necessary new research to optimize the production, safety and antitumor potency of the products derived from the MSCs suitable for oncological therapies.
Collapse
Affiliation(s)
- Noemi Eiro
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain.
| | - Maria Fraile
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain
| | - Silvia Fernández-Francos
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain
| | - Rosario Sánchez
- Department of Surgery, Fundación Hospital de Jove, 33290, Gijón, Asturias, Spain
| | - Luis A Costa
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain
| | - Francisco J Vizoso
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain. .,Department of Surgery, Fundación Hospital de Jove, 33290, Gijón, Asturias, Spain.
| |
Collapse
|
61
|
Suo F, Pan M, Li Y, Yan Q, Hu H, Hou L. Mesenchymal Stem Cells Cultured in 3D System Inhibit Non-Small Cell Lung Cancer Cells through p38 MAPK and CXCR4/AKT Pathways by IL-24 Regulating. Mol Biol 2021. [DOI: 10.1134/s0026893321030110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
62
|
Hassanzadeh A, Rahman HS, Markov A, Endjun JJ, Zekiy AO, Chartrand MS, Beheshtkhoo N, Kouhbanani MAJ, Marofi F, Nikoo M, Jarahian M. Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res Ther 2021; 12:297. [PMID: 34020704 PMCID: PMC8138094 DOI: 10.1186/s13287-021-02378-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, mesenchymal stem/stromal cells (MSCs) and their widespread biomedical applications have attracted great consideration from the scientific community around the world. However, reports have shown that the main populations of the transplanted MSCs are trapped in the liver, spleen, and lung upon administration, highlighting the importance of the development of cell-free therapies. Concerning rising evidence suggesting that the beneficial effects of MSC therapy are closely linked to MSC-released components, predominantly MSC-derived exosomes, the development of an MSC-based cell-free approach is of paramount importance. The exosomes are nano-sized (30100nm) lipid bilayer membrane vesicles, which are typically released by MSCs and are found in different body fluids. They include various bioactive molecules, such as messenger RNA (mRNA), microRNAs, proteins, and bioactive lipids, thus showing pronounced therapeutic competence for tissues recovery through the maintenance of their endogenous stem cells, the enhancement of regenerative phenotypic traits, inhibition of apoptosis concomitant with immune modulation, and stimulation of the angiogenesis. Conversely, the specific roles of MSC exosomes in the treatment of various tumors remain challenging. The development and clinical application of novel MSC-based cell-free strategies can be supported by better understanding their mechanisms, classifying the subpopulation of exosomes, enhancing the conditions of cell culture and isolation, and increasing the production of exosomes along with engineering exosomes to deliver drugs and therapeutic molecules to the target sites. In the current review, we deliver a brief overview of MSC-derived exosome biogenesis, composition, and isolation methods and discuss recent investigation regarding the therapeutic potential of MSC exosomes in regenerative medicine accompanied by their double-edged sword role in cancer.
Collapse
Affiliation(s)
- Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Sulaymaniyah, Iraq
| | | | - Judi Januadi Endjun
- Medical Faculty, UPN Veteran, Jakarta, Indonesia.,Gatot Soebroto Indonesia Army Hospital, Jakarta, Indonesia
| | | | | | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120, Heidelberg, Germany.
| |
Collapse
|
63
|
Tokunori Yamamoto, Gotoh M, Koide N, Funahashi Y, Shimizu S, Takei Y. Influence of human adipose stem cells on prostate cancer cell growth. NAGOYA JOURNAL OF MEDICAL SCIENCE 2021; 82:217-224. [PMID: 32581402 PMCID: PMC7276406 DOI: 10.18999/nagjms.82.2.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In a novel regenerative cell-based treatment developed by us for the patients with stress urinary incontinence, autologous adipose-derived stem cells (ASCs) are injected into the periurethral region and the external urethral sphincter. Since the candidates for this treatment included prostate cancer patients after radical prostatectomy, we investigated the effects of ASCs on prostate cancer cell proliferation in vitro and in vivo to confirm the feasibility of our therapeutic approach. The LNCaP (human prostate cancer cell line) cells and ASCs were co-cultured, and prostate-specific antigen (PSA) concentration in their culture medium supernatant was measured at 48 and 96 h. The PSA concentration significantly decreased in the coculture medium supernatant as compared to the culture medium with LNCaP cells alone. On the contrary, PSA concentrations in the culture medium of LNCaP cells were not affected by supplementation with ASC culture supernatant. After subcutaneous transplantation of LNCaP cells, with or without ASCs, in immunodeficient mice, tumor growth was compared. The growth of LNCaP xenograft tumor in immunodeficient mice was significantly suppressed by ASC addition. These results indicated that ASCs inhibit prostate cancer cell growth, without no proliferative effect on prostate cancer cells.
Collapse
Affiliation(s)
- Tokunori Yamamoto
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Momokazu Gotoh
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoshi Koide
- Division of Disease Models, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhito Funahashi
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinobu Shimizu
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yoshifumi Takei
- Division of Disease Models, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Medicinal Biochemistry, School of Pharmacy, Aichi Gakuin University, Nagoya, Japan
| |
Collapse
|
64
|
Tang Y, Zhou Y, Li HJ. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther 2021; 12:71. [PMID: 33468232 PMCID: PMC7814175 DOI: 10.1186/s13287-021-02138-7] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Stem cells can be used for regenerative medicine and as treatments for disease. The application of tissue engineering-related transplantation, stem cells, and local changes in the microenvironment is expected to solve major medical problems. Currently, most studies focus on tissue repair and regeneration, and mesenchymal stem cells (MSCs) are among the most common research topics. MSCs are applicable as seed cells, and they represent one of the current hot topics in regenerative medicine research. However, due to storage limitations and because cell senescence occurs during in vitro expansion, their clinical application is challenging. Exosomes, which are secreted by MSCs through paracrine signalling, not only have the same effects as MSCs, but they also have the advantages of targeted delivery, low immunogenicity, and high repairability. This article reviews the acquisition methods, characteristics, biological functions, and clinical applications of exosomes.
Collapse
Affiliation(s)
- Yaya Tang
- Key Laboratory of Vaccine Research and Development for Major Infectious Diseases of Yunnan Province, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118 People’s Republic of China
- Kunming Medical University, Kunming, 650500 People’s Republic of China
| | - Yan Zhou
- Key Laboratory of Vaccine Research and Development for Major Infectious Diseases of Yunnan Province, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118 People’s Republic of China
| | - Hong-Jun Li
- Key Laboratory of Vaccine Research and Development for Major Infectious Diseases of Yunnan Province, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118 People’s Republic of China
| |
Collapse
|
65
|
Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers (Basel) 2020; 13:cancers13010084. [PMID: 33396739 PMCID: PMC7795854 DOI: 10.3390/cancers13010084] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The aim of this review is to provide an overview of the current scientific evidence concerning the role played by exosomes in the pathogenesis, diagnosis and treatment of diseases. The potential use of exosomes as delivery vectors for small-molecule therapeutic agents will be discussed. In addition, a special emphasis will be placed on the involvement of exosomes in oncological diseases, as well as to their potential therapeutic application as liquid biopsy tools mainly in cancer diagnosis. A better understanding of exosome biology could improve the results of clinical interventions using exosomes as therapeutic agents. Abstract Exosomes are lipid bilayer particles released from cells into their surrounding environment. These vesicles are mediators of near and long-distance intercellular communication and affect various aspects of cell biology. In addition to their biological function, they play an increasingly important role both in diagnosis and as therapeutic agents. In this paper, we review recent literature related to the molecular composition of exosomes, paying special attention to their role in pathogenesis, along with their application as biomarkers and as therapeutic tools. In this context, we analyze the potential use of exosomes in biomedicine, as well as the limitations that preclude their wider application.
Collapse
|
66
|
Klyachko NL, Arzt CJ, Li SM, Gololobova OA, Batrakova EV. Extracellular Vesicle-Based Therapeutics: Preclinical and Clinical Investigations. Pharmaceutics 2020; 12:E1171. [PMID: 33271883 PMCID: PMC7760239 DOI: 10.3390/pharmaceutics12121171] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/24/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022] Open
Abstract
Drug nanoformulations hold remarkable promise for the efficient delivery of therapeutics to a disease site. Unfortunately, artificial nanocarriers, mostly liposomes and polymeric nanoparticles, show limited applications due to the unfavorable pharmacokinetics and rapid clearance from the blood circulation by the reticuloendothelial system (RES). Besides, many of them have high cytotoxicity, low biodegradability, and the inability to cross biological barriers, including the blood brain barrier. Extracellular vesicles (EVs) are novel candidates for drug delivery systems with high bioavailability, exceptional biocompatibility, and low immunogenicity. They provide a means for intercellular communication and the transmission of bioactive compounds to targeted tissues, cells, and organs. These features have made them increasingly attractive as a therapeutic platform in recent years. However, there are many obstacles to designing EV-based therapeutics. In this review, we will outline the main hurdles and limitations for therapeutic and clinical applications of drug loaded EV formulations and describe various attempts to solve these problems.
Collapse
Affiliation(s)
- Natalia L. Klyachko
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (N.L.K.); (O.A.G.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.J.A.); (S.M.L.)
| | - Camryn J. Arzt
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.J.A.); (S.M.L.)
| | - Samuel M. Li
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.J.A.); (S.M.L.)
| | - Olesia A. Gololobova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (N.L.K.); (O.A.G.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.J.A.); (S.M.L.)
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (N.L.K.); (O.A.G.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (C.J.A.); (S.M.L.)
| |
Collapse
|
67
|
Xing H, Liang C, Xu X, Sun H, Ma X, Jiang Z. Mesenchymal stroma/stem-like cells of GARP knockdown inhibits cell proliferation and invasion of mouse colon cancer cells (MC38) through exosomes. J Cell Mol Med 2020; 24:13984-13990. [PMID: 33155413 PMCID: PMC7753840 DOI: 10.1111/jcmm.16008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stroma/stem-like cells (MSCs) have antitumour activity, and MSC-derived exosomes play a role in the growth, metastasis and invasion of tumour cells. Additionally, glycoprotein A repetition predominant (GARP) promotes oncogenesis in breast cancer. Therefore, GARP is speculated to be a target gene for cancer therapy. We aimed to explore the therapy role of MSC-derived exosomes targeting GARP in mouse colon cancer cell MC38. We successfully established a GARP knockdown system using three kinds of siRNA-GARP in MSC cells. Exosomes were isolated from MSC and siGARP-MSC cells, and verified by the exosome surface protein markers CD9, CD63 and CD81. GARP expression was significantly decreased in siGARP-MSC exosomes compared with that of MSC exosomes. We found that siGARP-MSC exosomes inhibited cell proliferation, migration and invasion of MC38 cells, using CCK-8, colony formation, wound-healing and Transwell invasion assays. Furthermore, siGARP-MSC exosomes impeded IL-6 secretion and partly inactivated JAK1/STAT3 pathway, measured using ELISA and RT-qPCR. In conclusion, MSC-derived exosomes targeting GARP are a potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Haizhou Xing
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyan Liang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xintong Xu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Sun
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojun Ma
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
68
|
Massillo C, Duca RB, Lacunza E, Dalton GN, Farré PL, Taha N, Piccioni F, Scalise GD, Gardner K, De Siervi A. Adipose tissue from metabolic syndrome mice induces an aberrant miRNA signature highly relevant in prostate cancer development. Mol Oncol 2020; 14:2868-2883. [PMID: 32875710 PMCID: PMC7607170 DOI: 10.1002/1878-0261.12788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/11/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) remains an important public health concern in Western countries. Metabolic syndrome (MeS) is a cluster of pathophysiological disorders with increasing prevalence in the general population that is a risk factor for PCa. Several studies have determined that a crosstalk between white adipose tissue (WAT) and solid tumors favors cancer aggressiveness. In this work, our main goal was to investigate the interaction between WAT and PCa cells through microRNAs (miRNAs), in MeS mice. We developed a MeS‐like disease model using C57BL/6J mice chronically fed with high‐fat diet (HFD) that were inoculated with TRAMP‐C1 PCa cells. A group of five miRNAs (mmu‐miR‐221‐3p, 27a‐3p, 34a‐5p, 138‐5p, and 146a‐5p) were increased in gonadal WAT (gWAT), tumors, and plasma of MeS mice compared to control animals. Three of these five miRNAs were detected in the media from gWAT and TRAMP‐C1 cell cocultures, and significantly increased in MeS context. More importantly, hsa‐miR‐221‐3p, 146a‐5p, and 27a‐3p were increased in bloodstream of PCa patients compared to healthy donors. Using miRNA microarrays, we found that 121 miRNAs were differentially released to the coculture media between HFD‐gWAT and tumor cells compared to control diet‐gWAT and tumor cells. Target genes for the 66 most deregulated miRNAs were involved in common pathways, mainly related to fatty acid metabolism, ER protein processing, amino acid degradation, PI3K AKT signaling, and PCa. Our findings show for the first time a signature of five miRNAs as important players involved in the interaction between WAT and PCa in MeS mice. Further research will be necessary to track these miRNAs in the interaction between these tissues as well as their role in PCa patients with MeS.
Collapse
Affiliation(s)
- Cintia Massillo
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Rocío Belén Duca
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Ezequiel Lacunza
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Guillermo Nicolás Dalton
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Paula Lucía Farré
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Nicolás Taha
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Flavia Piccioni
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Georgina Daniela Scalise
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Kevin Gardner
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Adriana De Siervi
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
69
|
Wang T, Yu X, Lin J, Qin C, Bai T, Xu T, Wang L, Liu X, Li S. Adipose-Derived Stem Cells Inhibited the Proliferation of Bladder Tumor Cells by S Phase Arrest and Wnt/β-Catenin Pathway. Cell Reprogram 2020; 21:331-338. [PMID: 31809208 PMCID: PMC6918853 DOI: 10.1089/cell.2019.0047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ADSCs), which are present in most organs and tissues, were evaluated as a novel medium for stem cell therapy. In this study, we investigated the effects and underlying mechanisms of ADSCs in bladder tumor (BT) cells. SV-HUC, T24, and EJ cells were cultured with ADSCs and conditioned medium from ADSCs (ADSC-CM). We observed that in routine culture, ADSCs significantly inhibited the proliferation of T24 and EJ cells in a dose-dependent manner. In addition, ADSC-CM attenuated the viability of T24 and EJ cells in a dose-dependent manner. Cell cycle analysis indicated that ADSC-CM was capable of inducing T24 and EJ cells S phase arrest and downregulating the expression of CDK 1, whereas the expression of cyclin A was increased. ADSC-CM could induce apoptosis in T24 cells. The mechanism of this effect likely involved the caspase3/7 pathway and Wnt/β-catenin pathway. These findings demonstrated that ADSCs could inhibit the proliferation of BT cells via secretory factors.
Collapse
Affiliation(s)
- Tao Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xi Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Cong Qin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Bai
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Xu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shenglan Li
- Department of Radiography, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
70
|
Xunian Z, Kalluri R. Biology and therapeutic potential of mesenchymal stem cell-derived exosomes. Cancer Sci 2020; 111:3100-3110. [PMID: 32639675 PMCID: PMC7469857 DOI: 10.1111/cas.14563] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent stromal cells with the potential to differentiate into several cell types. MSC‐based therapy has emerged as a promising strategy for various diseases. Accumulating evidence suggests that the paracrine effects of MSC are partially exerted by the secretion of soluble factors, in particular exosomes. MSC‐derived exosomes are involved in intercellular communication through transfer of proteins, RNA, DNA and bioactive lipids, which might constitute a novel intercellular communication mode. This review illustrates the current knowledge on the composition and biological functions as well as the therapeutic potential of MSC‐derived exosomes in cancer, with a focus on clinical translation opportunities.
Collapse
Affiliation(s)
- Zhou Xunian
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Bioengineering, Rice University, Houston, Texas, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
71
|
Costa LA, Eiro N, Fraile M, Gonzalez LO, Saá J, Garcia-Portabella P, Vega B, Schneider J, Vizoso FJ. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: implications for further clinical uses. Cell Mol Life Sci 2020; 78:447-467. [PMID: 32699947 PMCID: PMC7375036 DOI: 10.1007/s00018-020-03600-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSC) are present in all organs and tissues. Several studies have shown the therapeutic potential effect of MSC or their derived products. However, the functional heterogeneity of MSC constitutes an important barrier for transferring these capabilities to the clinic. MSC heterogeneity depends on their origin (biological niche) or the conditions of potential donors (age, diseases or unknown factors). It is accepted that many culture conditions of the artificial niche to which they are subjected, such as O2 tension, substrate and extracellular matrix cues, inflammatory stimuli or genetic manipulations can influence their resulting phenotype. Therefore, to attain a more personalized and precise medicine, a correct selection of MSC is mandatory, based on their functional potential, as well as the need to integrate all the existing information to achieve an optimal improvement of MSC features in the artificial niche.
Collapse
Affiliation(s)
- Luis A Costa
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Noemi Eiro
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - María Fraile
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Luis O Gonzalez
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain.,Department of Anatomical Pathology, Fundación Hospital de Jove, Gijón, Spain
| | - Jorge Saá
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Pablo Garcia-Portabella
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Belén Vega
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - José Schneider
- Department of Obstetrics and Gynecology, University of Valladolid, Valladolid, Spain
| | - Francisco J Vizoso
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain.
| |
Collapse
|
72
|
Hu W, Liu C, Bi ZY, Zhou Q, Zhang H, Li LL, Zhang J, Zhu W, Song YYY, Zhang F, Yang HM, Bi YY, He QQ, Tan GJ, Sun CC, Li DJ. Comprehensive landscape of extracellular vesicle-derived RNAs in cancer initiation, progression, metastasis and cancer immunology. Mol Cancer 2020; 19:102. [PMID: 32503543 PMCID: PMC7273667 DOI: 10.1186/s12943-020-01199-1] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/15/2020] [Indexed: 01/18/2023] Open
Abstract
Extracellular vesicles (EVs), a class of heterogeneous membrane vesicles, are generally divided into exosomes and microvesicles on basis of their origination from the endosomal membrane or the plasma membrane, respectively. EV-mediated bidirectional communication among various cell types supports cancer cell growth and metastasis. EVs derived from different cell types and status have been shown to have distinct RNA profiles, comprising messenger RNAs and non-coding RNAs (ncRNAs). Recently, ncRNAs have attracted great interests in the field of EV-RNA research, and growing numbers of ncRNAs ranging from microRNAs to long ncRNAs have been investigated to reveal their specific functions and underlying mechanisms in the tumor microenvironment and premetastatic niches. Emerging evidence has indicated that EV-RNAs are essential functional cargoes in modulating hallmarks of cancers and in reciprocal crosstalk within tumor cells and between tumor and stromal cells over short and long distance, thereby regulating the initiation, development and progression of cancers. In this review, we discuss current findings regarding EV biogenesis, release and interaction with target cells as well as EV-RNA sorting, and highlight biological roles and molecular mechanisms of EV-ncRNAs in cancer biology.
Collapse
Affiliation(s)
- Wei Hu
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Cong Liu
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Zhuo-Yue Bi
- Hubei Provincial Key Laboratory for Applied Toxicology (Hubei Provincial Academy for Preventive Medicine), Wuhan, Hubei, 430079, People's Republic of China
| | - Qun Zhou
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Han Zhang
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Lin-Lin Li
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Jian Zhang
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Wei Zhu
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Yang-Yi-Yan Song
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Feng Zhang
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Hui-Min Yang
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Yong-Yi Bi
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Qi-Qiang He
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Gong-Jun Tan
- Department of Clinical Laboratory, Zhuhai Hospital, Jinan University, 79 Kangning Road, Zhuhai, Guangdong, 519000, People's Republic of China. .,Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Cheng-Cao Sun
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China. .,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - De-Jia Li
- Department of Preventive Medicine, School of Health Science, Wuhan University, No.115 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China. .,Population and Health Research Center, School of Health Sciences, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.
| |
Collapse
|
73
|
Liao Y, Xu K. Epigenetic regulation of prostate cancer: the theories and the clinical implications. Asian J Androl 2020; 21:279-290. [PMID: 30084432 PMCID: PMC6498736 DOI: 10.4103/aja.aja_53_18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epigenetics is the main mechanism that controls transcription of specific genes with no changes in the underlying DNA sequences. Epigenetic alterations lead to abnormal gene expression patterns that contribute to carcinogenesis and persist throughout disease progression. Because of the reversible nature, epigenetic modifications emerge as promising anticancer drug targets. Several compounds have been developed to reverse the aberrant activities of enzymes involved in epigenetic regulation, and some of them show encouraging results in both preclinical and clinical studies. In this article, we comprehensively review the up-to-date roles of epigenetics in the development and progression of prostate cancer. We especially focus on three epigenetic mechanisms: DNA methylation, histone modifications, and noncoding RNAs. We elaborate on current models/theories that explain the necessity of these epigenetic programs in driving the malignant phenotypes of prostate cancer cells. In particular, we elucidate how certain epigenetic regulators crosstalk with critical biological pathways, such as androgen receptor (AR) signaling, and how the cooperation dynamically controls cancer-oriented transcriptional profiles. Restoration of a "normal" epigenetic landscape holds promise as a cure for prostate cancer, so we concluded by highlighting particular epigenetic modifications as diagnostic and prognostic biomarkers or new therapeutic targets for treatment of the disease.
Collapse
Affiliation(s)
- Yiji Liao
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Kexin Xu
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
74
|
Parfejevs V, Sagini K, Buss A, Sobolevska K, Llorente A, Riekstina U, Abols A. Adult Stem Cell-Derived Extracellular Vesicles in Cancer Treatment: Opportunities and Challenges. Cells 2020; 9:cells9051171. [PMID: 32397238 PMCID: PMC7290929 DOI: 10.3390/cells9051171] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022] Open
Abstract
Adult stem cells (SCs) participate in tissue repair and homeostasis regulation. The relative ease of SC handling and their therapeutic effect has made of these cell popular candidates for cellular therapy. However, several problems interfere with their clinical application in cancer treatment, like safety issues, unpredictable pro-tumour effects, and tissue entrapment. Therefore cell-free therapies that exhibit SC properties are being investigated. It is now well known that adult SCs exhibit their therapeutic effect via paracrine mechanisms. In addition to secretory proteins, SCs also release extracellular vesicles (EV) that deliver their contents to the target cells. Cancer treatment is one of the most promising applications of SC-EVs. Moreover, SC-EVs could be modified to improve targeted drug delivery. The aim of the review is to summarise current knowledge of adult SC-EV application in cancer treatment and to emphasise future opportunities and challenges in cancer treatment.
Collapse
Affiliation(s)
- Vadims Parfejevs
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str 3, LV-1004 Riga, Latvia; (V.P.); (U.R.)
| | - Krizia Sagini
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway; (K.S.); (A.L.)
| | - Arturs Buss
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067 Riga, Latvia; (A.B.); (K.S.)
| | - Kristine Sobolevska
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067 Riga, Latvia; (A.B.); (K.S.)
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway; (K.S.); (A.L.)
| | - Una Riekstina
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str 3, LV-1004 Riga, Latvia; (V.P.); (U.R.)
| | - Arturs Abols
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067 Riga, Latvia; (A.B.); (K.S.)
- Correspondence:
| |
Collapse
|
75
|
Ha DH, Kim HK, Lee J, Kwon HH, Park GH, Yang SH, Jung JY, Choi H, Lee JH, Sung S, Yi YW, Cho BS. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020; 9:E1157. [PMID: 32392899 PMCID: PMC7290908 DOI: 10.3390/cells9051157] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nano-sized vesicles that serve as mediators for cell-to-cell communication. With their unique nucleic acids, proteins, and lipids cargo compositions that reflect the characteristics of producer cells, exosomes can be utilized as cell-free therapeutics. Among exosomes derived from various cellular origins, mesenchymal stem cell-derived exosomes (MSC-exosomes) have gained great attention due to their immunomodulatory and regenerative functions. Indeed, many studies have shown anti-inflammatory, anti-aging and wound healing effects of MSC-exosomes in various in vitro and in vivo models. In addition, recent advances in the field of exosome biology have enabled development of specific guidelines and quality control methods, which will ultimately lead to clinical application of exosomes. This review highlights recent studies that investigate therapeutic potential of MSC-exosomes and relevant mode of actions for skin diseases, as well as quality control measures required for development of exosome-derived therapeutics.
Collapse
Affiliation(s)
- Dae Hyun Ha
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Hyun-keun Kim
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Joon Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea;
| | | | - Gyeong-Hun Park
- Department of Dermatology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwasweong-si, Gyeonggi-do 18450, Korea;
| | | | | | | | - Jun Ho Lee
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Sumi Sung
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Yong Weon Yi
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Byong Seung Cho
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| |
Collapse
|
76
|
Shukla L, Yuan Y, Shayan R, Greening DW, Karnezis T. Fat Therapeutics: The Clinical Capacity of Adipose-Derived Stem Cells and Exosomes for Human Disease and Tissue Regeneration. Front Pharmacol 2020; 11:158. [PMID: 32194404 PMCID: PMC7062679 DOI: 10.3389/fphar.2020.00158] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Fat grafting is a well-established surgical technique used in plastic surgery to restore deficient tissue, and more recently, for its putative regenerative properties. Despite more frequent use of fat grafting, however, a scientific understanding of the mechanisms underlying either survival or remedial benefits of grafted fat remain lacking. Clinical use of fat grafts for breast reconstruction in tissues damaged by radiotherapy first provided clues regarding the clinical potential of stem cells to drive tissue regeneration. Healthy fat introduced into irradiated tissues appeared to reverse radiation injury (fibrosis, scarring, contracture and pain) clinically; a phenomenon since validated in several animal studies. In the quest to explain and enhance these therapeutic effects, adipose-derived stem cells (ADSCs) were suggested as playing a key role and techniques to enrich ADSCs in fat, in turn, followed. Stem cells - the body's rapid response 'road repair crew' - are on standby to combat tissue insults. ADSCs may exert influences either by releasing paracrine-signalling factors alone or as cell-free extracellular vesicles (EVs, exosomes). Alternatively, ADSCs may augment vital immune/inflammatory processes; or themselves differentiate into mature adipose cells to provide the 'building-blocks' for engineered tissue. Regardless, adipose tissue constitutes an ideal source for mesenchymal stem cells for therapeutic application, due to ease of harvest and processing; and a relative abundance of adipose tissue in most patients. Here, we review the clinical applications of fat grafting, ADSC-enhanced fat graft, fat stem cell therapy; and the latest evolution of EVs and nanoparticles in healing, cancer and neurodegenerative and multiorgan disease.
Collapse
Affiliation(s)
- Lipi Shukla
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia.,Department of Plastic Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Yinan Yuan
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia
| | - Ramin Shayan
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia.,Department of Plastic Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia.,Plastic, Hand and Faciomaxillary Surgery Unit, Alfred Hospital, Prahran, VIC, Australia.,Department of Plastic and Reconstructive Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Tara Karnezis
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia
| |
Collapse
|
77
|
Stem Cell Therapy for Hepatocellular Carcinoma: Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1237:97-119. [PMID: 31728916 DOI: 10.1007/5584_2019_441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of cancer and results in a high mortality rate worldwide. Unfortunately, most cases of HCC are diagnosed in an advanced stage, resulting in a poor prognosis and ineffective treatment. HCC is often resistant to both radiotherapy and chemotherapy, resulting in a high recurrence rate. Although the use of stem cells is evolving into a potentially effective approach for the treatment of cancer, few studies on stem cell therapy in HCC have been published. The administration of stem cells from bone marrow, adipose tissue, the amnion, and the umbilical cord to experimental animal models of HCC has not yielded consistent responses. However, it is possible to induce the apoptosis of cancer cells, repress angiogenesis, and cause tumor regression by administration of genetically modified stem cells. New alternative approaches to cancer therapy, such as the use of stem cell derivatives, exosomes or stem cell extracts, have been proposed. In this review, we highlight these experimental approaches for the use of stem cells as a vehicle for local drug delivery.
Collapse
|
78
|
Jurj A, Zanoaga O, Braicu C, Lazar V, Tomuleasa C, Irimie A, Berindan-Neagoe I. A Comprehensive Picture of Extracellular Vesicles and Their Contents. Molecular Transfer to Cancer Cells. Cancers (Basel) 2020; 12:cancers12020298. [PMID: 32012717 PMCID: PMC7072213 DOI: 10.3390/cancers12020298] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Critical processes such as growth, invasion, and metastasis of cancer cells are sustained via bidirectional cell-to-cell communication in tissue complex environments. Such communication involves the secretion of soluble factors by stromal cells and/or cancer cells within the tumor microenvironment (TME). Both stromal and cancer cells have been shown to export bilayer nanoparticles: encapsulated regulatory molecules that contribute to cell-to-cell communication. These nanoparticles are known as extracellular vesicles (EVs) being classified into exosomes, microvesicles, and apoptotic bodies. EVs carry a vast repertoire of molecules such as oncoproteins and oncopeptides, DNA fragments from parental to target cells, RNA species (mRNAs, microRNAs, and long non-coding RNA), and lipids, initiating phenotypic changes in TME. According to their specific cargo, EVs have crucial roles in several early and late processes associated with tumor development and metastasis. Emerging evidence suggests that EVs are being investigated for their implication in early cancer detection, monitoring cancer progression and chemotherapeutic response, and more relevant, the development of novel targeted therapeutics. In this study, we provide a comprehensive understanding of the biophysical properties and physiological functions of EVs, their implications in TME, and highlight the applicability of EVs for the development of cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Vladimir Lazar
- Worldwide Innovative Network for Personalized Cancer Therapy, 94800 Villejuif, France;
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
- Department of Hematology, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Alexandru Irimie
- 11th Department of Surgical Oncology and Gynaecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
- Department of Surgery, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
- Correspondence: (A.I.); (I.B.-N.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
- MEDFUTURE—Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
- Correspondence: (A.I.); (I.B.-N.)
| |
Collapse
|
79
|
Conti I, Varano G, Simioni C, Laface I, Milani D, Rimondi E, Neri LM. miRNAs as Influencers of Cell-Cell Communication in Tumor Microenvironment. Cells 2020; 9:cells9010220. [PMID: 31952362 PMCID: PMC7016744 DOI: 10.3390/cells9010220] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level, inducing the degradation of the target mRNA or translational repression. MiRNAs are involved in the control of a multiplicity of biological processes, and their absence or altered expression has been associated with a variety of human diseases, including cancer. Recently, extracellular miRNAs (ECmiRNAs) have been described as mediators of intercellular communication in multiple contexts, including tumor microenvironment. Cancer cells cooperate with stromal cells and elements of the extracellular matrix (ECM) to establish a comfortable niche to grow, to evade the immune system, and to expand. Within the tumor microenvironment, cells release ECmiRNAs and other factors in order to influence and hijack the physiological processes of surrounding cells, fostering tumor progression. Here, we discuss the role of miRNAs in the pathogenesis of multicomplex diseases, such as Alzheimer’s disease, obesity, and cancer, focusing on the contribution of both intracellular miRNAs, and of released ECmiRNAs in the establishment and development of cancer niche. We also review growing evidence suggesting the use of miRNAs as novel targets or potential tools for therapeutic applications.
Collapse
Affiliation(s)
- Ilaria Conti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Gabriele Varano
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Ilaria Laface
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Erika Rimondi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Luca M. Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
- LTTA—Electron Microscopy Center, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-455940
| |
Collapse
|
80
|
Ren XS, Tong Y, Qiu Y, Ye C, Wu N, Xiong XQ, Wang JJ, Han Y, Zhou YB, Zhang F, Sun HJ, Gao XY, Chen Q, Li YH, Kang YM, Zhu GQ. MiR155-5p in adventitial fibroblasts-derived extracellular vesicles inhibits vascular smooth muscle cell proliferation via suppressing angiotensin-converting enzyme expression. J Extracell Vesicles 2019; 9:1698795. [PMID: 31839907 PMCID: PMC6896498 DOI: 10.1080/20013078.2019.1698795] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 10/20/2019] [Accepted: 11/21/2019] [Indexed: 12/17/2022] Open
Abstract
Proliferation of vascular smooth muscle cells (VSMCs) plays crucial roles in vascular remodelling and stiffening in hypertension. Vascular adventitial fibroblasts are a key regulator of vascular wall function and structure. This study is designed to investigate the roles of adventitial fibroblasts-derived extracellular vesicles (EVs) in VSMC proliferation and vascular remodelling in normotensive Wistar-Kyoto rat (WKY) and spontaneously hypertensive rat (SHR), an animal model of human essential hypertension. EVs were isolated from aortic adventitial fibroblasts of WKY (WKY-EVs) and SHR (SHR-EVs). Compared with WKY-EVs, miR155-5p content was reduced, while angiotensin-converting enzyme (ACE) content was increased in SHR-EVs. WKY-EVs inhibited VSMC proliferation of SHR, which was prevented by miR155-5p inhibitor. SHR-EVs promoted VSMC proliferation of both strains, which was enhanced by miR155-5p inhibitor, but abolished by captopril or losartan. Dual luciferase reporter assay showed that ACE was a target gene of miR155-5p. MiR155-5p mimic or overexpression inhibited VSMC proliferation and ACE upregulation of SHR. WKY-EVs reduced ACE mRNA and protein expressions while SHR-EVs only increased ACE protein level in VSMCs of both strains. However, the SHR-EVs-derived from the ACE knockdown-treated adventitial fibroblasts lost the roles in promoting VSMC proliferation and ACE upregulation. Systemic miR155-5p overexpression reduced vascular ACE, angiotensin II and proliferating cell nuclear antigen levels, and attenuated hypertension and vascular remodelling in SHR. Repetitive intravenous injection of SHR-EVs increased blood pressure and vascular ACE contents, and promoted vascular remodelling in both strains, while WKY-EVs reduced vascular ACE contents and attenuated hypertension and vascular remodelling in SHR. We concluded that WKY-EVs-mediated miR155-5p transfer attenuates VSMC proliferation and vascular remodelling in SHR via suppressing ACE expression, while SHR-EVs-mediated ACE transfer promotes VSMC proliferation and vascular remodelling.
Collapse
Affiliation(s)
- Xing-Sheng Ren
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Tong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Qiu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chao Ye
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nan Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao-Qing Xiong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jue-Jin Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Han
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ye-Bo Zhou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hai-Jian Sun
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xing-Ya Gao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
81
|
Aoki M, Kakimoto K, Goto M, Higuchi K. Novel Therapeutic Approach Using Drug-loaded Adipose-derived Stem Cells for Pancreatic Cancer. Sci Rep 2019; 9:17971. [PMID: 31784549 PMCID: PMC6884490 DOI: 10.1038/s41598-019-53807-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022] Open
Abstract
We developed anticancer drug-conjugated biodegradable polymer-nanoparticle-loaded adipose-derived stem cells (AdSCs) as a tool for biodrug delivery systems for cancer therapy. Pirarubicin was conjugated in polylactic/glycolic acid (PLGA) followed by formation of nanoparticles (NPs), which were loaded with human AdSCs and cocultured. The pirarubicin-conjugated PLGA NP-loaded AdSCs (PirNP-AdSCs) were overall viable within 48 h and exhibited significantly enhanced migration activity. We confirmed that pirarubicin was gradually released into the culture medium from PirNP-AdSCs, and the conditioned medium significantly inhibited the proliferation activity and induced the apoptosis of human pancreatic cancer cells (KP1N). PirNP-AdSCs also significantly induced tumor cell apoptosis in an ex vivo culture system with KP1N-derived tumors, and there was increased invasion/migration of PirNP-AdSCs inside the tumor. Finally, we compared the therapeutic efficacy of the PirNP-AdSCs on KP1N-derived tumor growth with that of treatments of AdSCs alone, PirNPs alone or normal saline (control) in immunodeficient mice. Subcutaneous local administration of PirNP-AdSCs significantly inhibited tumor growth, inducing the apoptosis of tumor cells and vasculature compared with the other groups. The present therapeutic strategy might give rise to a novel cancer therapy minimizing the adverse side effects of anticancer drugs in patients who suffer from cancer.
Collapse
Affiliation(s)
- Masahiko Aoki
- Internal Medicine (II), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Kazuki Kakimoto
- Internal Medicine (II), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masahiro Goto
- Internal Medicine (II), Faculty of Medicine, Osaka Medical College, Osaka, Japan.
| | - Kazuhide Higuchi
- Internal Medicine (II), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| |
Collapse
|
82
|
Abdelrazik H, Giordano E, Barbanti Brodano G, Griffoni C, De Falco E, Pelagalli A. Substantial Overview on Mesenchymal Stem Cell Biological and Physical Properties as an Opportunity in Translational Medicine. Int J Mol Sci 2019; 20:5386. [PMID: 31671788 PMCID: PMC6862078 DOI: 10.3390/ijms20215386] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSC) have piqued worldwide interest for their extensive potential to treat a large array of clinical indications, their unique and controversial immunogenic and immune modulatory properties allowing ample discussions and debates for their possible applications. Emerging data demonstrating that the interaction of biomaterials and physical cues with MSC can guide their differentiation into specific cell lineages also provide new interesting insights for further MSC manipulation in different clinical applications. Moreover, recent discoveries of some regulatory molecules and signaling pathways in MSC niche that may regulate cell fate to distinct lineage herald breakthroughs in regenerative medicine. Although the advancement and success in the MSC field had led to an enormous increase in the amount of ongoing clinical trials, we still lack defined clinical therapeutic protocols. This review will explore the exciting opportunities offered by human and animal MSC, describing relevant biological properties of these cells in the light of the novel emerging evidence mentioned above while addressing the limitations and challenges MSC are still facing.
Collapse
Affiliation(s)
- Heba Abdelrazik
- Department of Clinical Pathology, Cairo University, Cairo 1137, Egypt.
- Department of Diagnosis, central laboratory department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16131 Genoa, Italy.
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, 47522 Cesena, Italy.
| | - Giovanni Barbanti Brodano
- Department of Oncological and Degenerative Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Cristiana Griffoni
- Department of Oncological and Degenerative Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
- Mediterranea Cardiocentro, 80122 Napoli, Italy.
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy.
- Institute of Biostructures and Bioimages (IBB), National Research Council (CNR), 80131 Naples, Italy.
| |
Collapse
|
83
|
Mohammadi S, Yousefi F, Shabaninejad Z, Movahedpour A, Mahjoubin Tehran M, Shafiee A, Moradizarmehri S, Hajighadimi S, Savardashtaki A, Mirzaei H. Exosomes and cancer: From oncogenic roles to therapeutic applications. IUBMB Life 2019; 72:724-748. [PMID: 31618516 DOI: 10.1002/iub.2182] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
Exosomes belong to extracellular vehicles that were produced and secreted from most eukaryotic cells and are involved in cell-to-cell communications. They are an effective delivery system for biological compounds such as mRNAs, microRNAs (miRNAs), proteins, lipids, saccharides, and other physiological compounds to target cells. In this way, they could influence on cellular pathways and mediate their physiological behaviors including cell proliferation, tumorigenesis, differentiation, and so on. Many research studies focused on their role in cancers and also on potentially therapeutic and biomarker applications. In the current study, we reviewed the exosomes' effects on cancer progression based on their cargoes including miRNAs, long noncoding RNAs, circular RNAs, DNAs, mRNAs, proteins, and lipids. Moreover, their therapeutic roles in cancer were considered. In this regard, we have given a brief overview of challenges and obstacles in using exosomes as therapeutic agents.
Collapse
Affiliation(s)
- Soheila Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Mahjoubin Tehran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alimohammad Shafiee
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Sanaz Moradizarmehri
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Sarah Hajighadimi
- Division of General Internal Medicine, Toronto General Hospital, Toronto, Canada
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
84
|
Park KS, Bandeira E, Shelke GV, Lässer C, Lötvall J. Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther 2019; 10:288. [PMID: 31547882 PMCID: PMC6757418 DOI: 10.1186/s13287-019-1398-3] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
After the initial investigations into applications of mesenchymal stem cells (MSCs) for cell therapy, there was increased interest in their secreted soluble factors. Following studies of MSCs and their secreted factors, extracellular vesicles (EVs) released from MSCs have emerged as a new mode of intercellular crosstalk. MSC-derived EVs have been identified as essential signaling mediators under both physiological and pathological conditions, and they appear to be responsible for many of the therapeutic effects of MSCs. In several in vitro and in vivo models, EVs have been observed to have supportive functions in modulating the immune system, mainly mediated by EV-associated proteins and nucleic acids. Moreover, stimulation of MSCs with biophysical or biochemical cues, including EVs from other cells, has been shown to influence the contents and biological activities of subsequent MSC-derived EVs. This review provides on overview of the contents of MSC-derived EVs in terms of their supportive effects, and it provides different perspectives on the manipulation of MSCs to improve the secretion of EVs and subsequent EV-mediated activities. In this review, we discuss the possibilities for manipulating MSCs for EV-based cell therapy and for using EVs to affect the expression of elements of interest in MSCs. In this way, we provide a clear perspective on the state of the art of EVs in cell therapy focusing on MSCs, and we raise pertinent questions and suggestions for knowledge gaps to be filled.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Elga Bandeira
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ganesh V Shelke
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
85
|
Gentile P, Garcovich S. Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. J Clin Med 2019; 8:855. [PMID: 31208047 PMCID: PMC6616456 DOI: 10.3390/jcm8060855] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been routinely used from several years in regenerative surgery without any definitive statement about their potential pro-oncogenic or anti-oncogenic role. ASCs has proven to favor tumor progression in several experimental cancer models, playing a central role in regulating tumor invasiveness and metastatic potential through several mechanisms, such as the paracrine release of exosomes containing pro-oncogenic molecules and the induction of epithelial-mesenchymal transition. However, the high secretory activity and the preferential tumor-targeting make also ASCs a potentially suitable vehicle for delivery of new anti-cancer molecules in tumor microenvironment. Nanotechnologies, viral vectors, drug-loaded exosomes, and micro-RNAs (MiR) represent additional new tools that can be applied for cell-mediated drug delivery in a tumor microenvironment. Recent studies revealed that the MiR play important roles in paracrine actions on adipose-resident macrophages, and their dysregulation has been implicated in the pathogenesis of obesity, diabetes, and diabetic complications as wounds. Numerous MiR are present in adipose tissues, actively participating in the regulation of adipogenesis, adipokine secretion, inflammation, and inter-cellular communications in the local tissues. These results provide important insights into Adipocyte-secreted exosomal microRNA (A-SE-MiR) function and they suggest evaluating the potential role of A-SE-MiR in tumor progression, the mechanisms underlying ASCs-cancer cell interplay and clinical safety of ASCs-based therapies.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery Unit, University of "Tor Vergata", 00133 Rome, Italy.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
86
|
Tong Y, Ye C, Ren XS, Qiu Y, Zang YH, Xiong XQ, Wang JJ, Chen Q, Li YH, Kang YM, Zhu GQ. Exosome-Mediated Transfer of ACE (Angiotensin-Converting Enzyme) From Adventitial Fibroblasts of Spontaneously Hypertensive Rats Promotes Vascular Smooth Muscle Cell Migration. Hypertension 2019; 72:881-888. [PMID: 30354715 DOI: 10.1161/hypertensionaha.118.11375] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Migration of vascular smooth muscle cells (VSMCs) is pivotal for vascular remodeling in hypertension. Vascular adventitial fibroblasts (AFs) are important in the homeostasis of vascular structure. This study is designed to investigate the roles of AF exosomes (AFE) in VSMC migration and underling mechanism. Primary VSMCs and AFs were obtained from the aorta of spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats. VSMC migration was evaluated with Boyden chamber assay and wound healing assay. AFE from WKY rats and SHR were isolated and identified. AFE from SHR promoted but AFE from WKY rats had no significant effect on VSMC migration. The effects of AFE on VSMC migration were prevented by an exosome inhibitor GW4869, an AT1R (Ang II [angiotensin II] type 1 receptor) antagonist losartan, or an inhibitor of ACE (angiotensin-converting enzyme) captopril. ACE contents and activity were much higher in AFE from SHR than those from WKY rats. There were no significant difference in Ang II and AT1R mRNA and protein levels between AFE from SHR and AFE from WKY rats. AFE from SHR increased Ang II and ACE contents and ACE activity in VSMCs of WKY rats and SHR. The changes of Ang II contents and ACE activity were prevented by captopril. ACE knockdown in AFs reduced ACE contents and activity in AFE from SHR and inhibited AFE-induced migration of VSMCs of WKY rats and those of SHR. These results indicate that exosomes from AFs of SHR transfer ACE to VSMCs, which increases Ang II levels and activates AT1R in VSMCs and thereby promotes VSMC migration.
Collapse
Affiliation(s)
- Ying Tong
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Chao Ye
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Xing-Sheng Ren
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Yun Qiu
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Ying-Hao Zang
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | - Xiao-Qing Xiong
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.)
| | | | - Qi Chen
- Department of Pathophysiology (Q.C., Y.-H.L., G.-Q.Z.), Nanjing Medical University, Jiangsu, China
| | - Yue-Hua Li
- Department of Pathophysiology (Q.C., Y.-H.L., G.-Q.Z.), Nanjing Medical University, Jiangsu, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, China (Y.-M.K.)
| | - Guo-Qing Zhu
- From the Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology (Y.T., C.Y., X.-S.R., Y.Q., Y.-H.Z., X.-Q.X., J.-J.W., G.-Q.Z.).,Department of Pathophysiology (Q.C., Y.-H.L., G.-Q.Z.), Nanjing Medical University, Jiangsu, China
| |
Collapse
|
87
|
Wong DE, Banyard DA, Santos PJF, Sayadi LR, Evans GRD, Widgerow AD. Adipose-derived stem cell extracellular vesicles: A systematic review ✰. J Plast Reconstr Aesthet Surg 2019; 72:1207-1218. [PMID: 30952587 DOI: 10.1016/j.bjps.2019.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/21/2019] [Accepted: 03/10/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are cell-secreted packages that deliver cargo to target cells to effect functional and phenotypic changes. They are secreted by many different cell types, including adipose-derived stem cells (ADSCs), which are a promising field of study in regenerative medicine. Our aim was to perform a systematic review of the literature to summarize the scientific work that has been conducted on ADSC EVs to date. METHODS The Pubmed database was queried with keywords (and variations of) "adipose derived stem cell," "stromal vascular fraction," and "extracellular vesicles." We excluded review papers, then manually screened articles based on title and abstract. Full-text articles were assessed for eligibility to include in final review. RESULTS While an extensive body of research exists on EVs, a much smaller proportion of that is original research on ADSC EVs. Of 44 manuscripts that met our database search criteria, 21 articles were selected for our systematic review. CONCLUSION ADSC EVs were found to exert effects on angiogenesis, cell survival and apoptosis, inflammation, tissue regeneration, and reduction of disease pathology. Further studies examine characteristics of ADSC EVs. Future work should aim to further detail the safety profiles of ADSC EVs given their potential for cell-based therapies. The body of research studies characterizing ADSC EVs continues to expand, and much work remains to be done before human pilot studies can be considered. To our knowledge, we offer the first systematic review summarizing the research on ADSC EVs and their determined roles to date.
Collapse
Affiliation(s)
- Daniel E Wong
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Derek A Banyard
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States; Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Pauline J F Santos
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Lohrasb R Sayadi
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Gregory R D Evans
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States
| | - Alan D Widgerow
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States; Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Orange, CA, United States.
| |
Collapse
|
88
|
Xiao Y, Geng F, Wang G, Li X, Zhu J, Zhu W. Bone marrow-derived mesenchymal stem cells-derived exosomes prevent oligodendrocyte apoptosis through exosomal miR-134 by targeting caspase-8. J Cell Biochem 2019; 120:2109-2118. [PMID: 30191592 DOI: 10.1002/jcb.27519] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023]
Abstract
Ischemic stroke causes severe brain damage and remains one of the leading causes of morbidity and mortality worldwide. The microRNA-134 (miR-134) is involved in regulating the process of ischemia injury in neural cells and brain with ischemia stroke. The role of miR-134 in ischemic stroke remains poorly understood. The purpose of the current study was to investigate the effect of bone marrow-derived mesenchymal stem cells (BMSCs)-derived exosomal miR-134 on rat oligodendrocytes (OLs) apoptosis and its underlying mechanism of action. The results demonstrated that levels of miR-134 in BMSCs-exosome decreased but increased incaspase-8 after oxygen-glucose deprivation (OGD) treatment. Exosomal miR-134 significantly inhibited apoptosis by decreasing caspase-8 expression and activity in OGD-treated group cultured with BMSCs-exosome and OLs. In addition, the miR-134 mimics decreased caspase-8 expression in OGD-treated OLs, whereas miR-134 inhibitors exacerbated the changes in the expression of the procaspase-8 and caspase-8 cleaved product proteins caused by OGD. The caspase-8 knockdown using caspase-8 small interfering RNA decreased OLs apoptosis, reversing the improvements that the miR-134 inhibited cells apoptosis by targeting caspase-8. Taken together, these results demonstrated that BMSCs-derived exosomes suppressed OLs apoptosis through exosomal miR-134 by negatively regulating the caspase-8-dependent apoptosis pathway and may, therefore, be a novel potential therapeutic target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Fengyang Geng
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Guifang Wang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Jianxin Zhu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Weijie Zhu
- Department of Neurosurgery, General Hospital of Jinan Military Region, Jinan, China
| |
Collapse
|
89
|
Zhou L, Feng Y, Dai J, Ouyang J. [Research progress of miRNA regulation in differentiation of adipose-derived stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:1506-1511. [PMID: 29806396 DOI: 10.7507/1002-1892.201706076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Objective To review the research progress of miRNA regulation in the differentiation of adipose-derived stem cells (ADSCs). Methods The recent literature associated with miRNAs and differentiation of ADSCs was reviewed. The regulatory mechanism was analyzed in detail and summarized. Results The results indicate that the expression of miRNAs changes during differentiation of ADSCs. In addition, miRNAs regulate the differentiation of ADSCs into adipocytes, osteoblasts, chondrocytes, neurons, and hepatocytes by regulating the signaling pathways involved in cell differentiation. Conclusion Through controlling the differentiation of ADSCs by miRNAs, the suitable seed cell for tissue engineering can be established. The review will provide a theoretical basis for molecular targeted therapy and stem cell therapy in clinic.
Collapse
Affiliation(s)
- Lanting Zhou
- Medical College, Hubei University of Arts and Science, Xiangyang Hubei, 441053, P.R.China;Department of Anatomy, Southern Medical University, Guangzhou Guangdong, 510515, P.R.China
| | - Yanting Feng
- Department of Anatomy, Southern Medical University, Guangzhou Guangdong, 510515, P.R.China
| | - Jingxing Dai
- Department of Anatomy, Southern Medical University, Guangzhou Guangdong, 510515, P.R.China
| | - Jun Ouyang
- Department of Anatomy, Southern Medical University, Guangzhou Guangdong, 510515,
| |
Collapse
|
90
|
Nawaz M. Extracellular vesicle-mediated transport of non-coding RNAs between stem cells and cancer cells: implications in tumor progression and therapeutic resistance. Stem Cell Investig 2017; 4:83. [PMID: 29167804 DOI: 10.21037/sci.2017.10.04] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
Recent years have witnessed intensive progress in studying extracellular vesicles (EVs), both for understanding their basic biology and contribution to variety of diseases, biomarker discovery, and their potential as gene delivery vectors and source of innovative therapies. As such, stem cell-derived EVs have contributed significant knowledge which led to the development of cell-free therapies in regenerative medicine. Although, the role of stem cell-derived EVs in maintaining stemness, differentiation and repairing tissue injuries is relatively well-understood; however, knowledge about the contribution of stem cell-derived EVs in cancer progression is just emerging. The aim of this review is, therefore, to discuss the recent developments in stem cell-derived EVs and tumor progression, placing a particular focus on non-coding RNA (ncRNA) mediated cancer progression and resistance against therapies. This includes the failure of normal hematopoiesis and the progression of myeloid neoplasms, enhanced capacity of cancer cells to proliferate and metastasize, and the conversion of normal cells into cancer cells, activation of angiogenic pathways and dormancy in cancer cells. These processes are shared by mesenchymal stem cells (MSCs), cancer stem like-cells and cancer cells in an intricate intratumoral network in order to create self-strengthening tumor niche. In this context, EV-ncRNAs serve as mediators to relay bystander effects of secreting cancer stem cells (CSCs) into recipient cells for priming a tumor permissive environment and relaying therapeutic resistance. Collectively, this knowledge will improve our understandings and approaches in finding new therapeutic targets in the context of CSCs, which could be benefited through engineering EVs for innovative therapies.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
91
|
Pan J, Ding M, Xu K, Yang C, Mao LJ. Exosomes in diagnosis and therapy of prostate cancer. Oncotarget 2017; 8:97693-97700. [PMID: 29228644 PMCID: PMC5722596 DOI: 10.18632/oncotarget.18532] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/02/2017] [Indexed: 01/16/2023] Open
Abstract
Exosomes are small vesicular bodies released by a variety of cells. Exosomes contain miRNAs, mRNAs and proteins with the potential to regulate signaling pathways in recipient cells. Exosomes deliver nucleic acids and proteins to mediate the communication between cancer cells and stroma cells. In this review, we summarize recent progress in our understanding of the role of exosomes in prostate cancer. The tumorigenesis, metastasis and drug resistance of prostate cancer are associated with the cargos of exosomes such as miRNAs, lncRNAs and proteins. In addition, prostate cancer cells modulate surrounding stromal cells via the exosomes. Affected stromal cells employ the exosomes to modulate microenvironment and promote tumor growth and metastasis. Exosomes derived from prostate cancer cells contribute to cancer chemoresistance. The lipid bilayer membrane of the exosomes makes them promising carriers of drugs and other therapeutic molecules targeting prostate cancer. Furthermore, exosomes can be detected and isolated from various body fluids for the diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Jun Pan
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Meng Ding
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Kai Xu
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Chunhua Yang
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.,Radiotherapy Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Li-Jun Mao
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| |
Collapse
|
92
|
Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med 2017; 49:e346. [PMID: 28620221 PMCID: PMC5519012 DOI: 10.1038/emm.2017.63] [Citation(s) in RCA: 436] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/03/2017] [Accepted: 01/12/2017] [Indexed: 12/13/2022] Open
Abstract
The administration of mesenchymal stem cells (MSCs) as a therapy for liver disease holds great promise. MSCs can differentiate into hepatocytes, reduce liver inflammation, promote hepatic regeneration and secrete protective cytokines. However, the risks of iatrogenic tumor formation, cellular rejection and infusional toxicity in MSC transplantation remain unresolved. Accumulating evidence now suggests that a novel cell-free therapy, MSC-secreted exosomes, might constitute a compelling alternative because of their advantages over the corresponding MSCs. They are smaller and less complex than their parent cells and, thus, easier to produce and store, they are devoid of viable cells, and they present no risk of tumor formation. Moreover, they are less immunogenic than their parent cells because of their lower content in membrane-bound proteins. This paper reviews the biogenesis of MSC exosomes and their physiological functions, and highlights the specific biochemical potential of MSC-derived exosomes in restoring tissue homeostasis. In addition, we summarize the recent advances in the role of exosomes in MSC therapy for various liver diseases, including liver fibrosis, acute liver injury and hepatocellular carcinoma. This paper also discusses the potential challenges and strategies in the use of exosome-based therapies for liver disease in the future.
Collapse
Affiliation(s)
- Guohua Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
93
|
Fatima F, Nawaz M. Vesiculated Long Non-Coding RNAs: Offshore Packages Deciphering Trans-Regulation between Cells, Cancer Progression and Resistance to Therapies. Noncoding RNA 2017; 3:ncrna3010010. [PMID: 29657282 PMCID: PMC5831998 DOI: 10.3390/ncrna3010010] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/10/2017] [Accepted: 02/16/2017] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized vesicles secreted from virtually all cell types and are thought to transport proteins, lipids and nucleic acids including non-coding RNAs (ncRNAs) between cells. Since, ncRNAs are central to transcriptional regulation during developmental processes; eukaryotes might have evolved novel means of post-transcriptional regulation by trans-locating ncRNAs between cells. EV-mediated transportation of regulatory elements provides a novel source of trans-regulation between cells. In the last decade, studies were mainly focused on microRNAs; however, functions of long ncRNA (lncRNA) have been much less studied. Here, we review the regulatory roles of EV-linked ncRNAs, placing a particular focus on lncRNAs, how they can foster dictated patterns of trans-regulation in recipient cells. This refers to envisaging novel mechanisms of epigenetic regulation, cellular reprogramming and genomic instability elicited in recipient cells, ultimately permitting the generation of cancer initiating cell phenotypes, senescence and resistance to chemotherapies. Conversely, such trans-regulation may introduce RNA interference in recipient cancer cells causing the suppression of oncogenes and anti-apoptotic proteins; thus favoring tumor inhibition. Collectively, understanding these mechanisms could be of great value to EV-based RNA therapeutics achieved through gene manipulation within cancer cells, whereas the ncRNA content of EVs from cancer patients could serve as non-invasive source of diagnostic biomarkers and prognostic indicators in response to therapies.
Collapse
Affiliation(s)
- Farah Fatima
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14049-900, Brazil.
| | - Muhammad Nawaz
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14049-900, Brazil.
| |
Collapse
|
94
|
Wen MM. Getting miRNA Therapeutics into the Target Cells for Neurodegenerative Diseases: A Mini-Review. Front Mol Neurosci 2016; 9:129. [PMID: 27920668 PMCID: PMC5118630 DOI: 10.3389/fnmol.2016.00129] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/08/2016] [Indexed: 11/29/2022] Open
Abstract
miRNAs play important roles in modulating gene expression in varying cellular processes and disease pathogenesis, including neurodegenerative diseases. Several miRNAs are expressed in the brain, control brain development and are identified as important biomarkers in the pathogenesis of motor—and neuro-cognitive diseases such as Alzheimer’s (AD), Huntington’s and Parkinson’s diseases (PD) and amyotrophic lateral sclerosis. These remarkable miRNAs could be used as diagnostic markers and therapeutic targeting potential for many stressful and untreatable progressive neurodegenerative diseases. To modulate these miRNA activities, there are currently two strategies involved; first one is to therapeutically restore the suppressed miRNA level by miRNA mimics (agonist), and the other one is to inhibit miRNA function by using anti-miR (antagonist) to repress overactive miRNA function. However, RNAi-based therapeutics often faces in vivo instability because naked nucleic acids are subject to enzyme degradation before reaching the target sites. Therefore, an effective, safe and stable bio-responsive delivery system is necessary to protect the nucleic acids from serum degradation and assist their entrance to the cells. Since neuronal cells are non-regenerating, to design engineered miRNAs to be delivered to the central nervous system (CNS) for long term gene expression and knockdown is representing an enormous challenge for scientists. This article provides an insight summary on some of the innovative strategies employed to deliver miRNA into target cells. These viral and non-viral carrier systems hold promise in RNA therapy delivery for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming Ming Wen
- Department of Pharmaceutics, Pharos University in Alexandria Alexandria, Egypt
| |
Collapse
|
95
|
Udensi UK, Tchounwou PB. Oxidative stress in prostate hyperplasia and carcinogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:139. [PMID: 27609145 PMCID: PMC5017015 DOI: 10.1186/s13046-016-0418-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
Abstract
Prostatic hyperplasia (PH) is a common urologic disease that affects mostly elderly men. PH can be classified as benign prostatic hyperplasia (BPH), or prostate cancer (PCa) based on its severity. Oxidative stress (OS) is known to influence the activities of inflammatory mediators and other cellular processes involved in the initiation, promotion and progression of human neoplasms including prostate cancer. Scientific evidence also suggests that micronutrient supplementation may restore the antioxidant status and hence improve the clinical outcomes for patients with BPH and PCa. This review highlights the recent studies on prostate hyperplasia and carcinogenesis, and examines the role of OS on the molecular pathology of prostate cancer progression and treatment.
Collapse
Affiliation(s)
- Udensi K Udensi
- NIH/NIMHD RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, 39217, USA
| | - Paul B Tchounwou
- NIH/NIMHD RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, 39217, USA.
| |
Collapse
|