201
|
The Role of Extracellular Vesicles in the Development of a Cancer Stem Cell Microenvironment Niche and Potential Therapeutic Targets: A Systematic Review. Cancers (Basel) 2021; 13:cancers13102435. [PMID: 34069860 PMCID: PMC8157362 DOI: 10.3390/cancers13102435] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Cancer stem cells (CSCs) are cancer cells that possess traits usually attributed to stem cells. An increase in CSCs can lead to more rapid cancer progression, treatment resistance and the increased likelihood of recurrence. To promote CSC survival and associated cancer progression, cancer cells enter into reciprocal crosstalk with the surrounding tissue environment, as well as with distant metastatic sites. This mechanism of communication relies, in part, on secreted factors, of which extracellular vesicles (EVs) are thought to have a critical role. This systematic review evaluates the current knowledge of cancer communication via EVs to alter the microenvironment to increase the survival and maintenance of CSCs. A total of 16 studies spanning the EV content, pathway alterations and CSC-targeting treatments provide new insights into how EVs mediate CSC traits and identify the gaps in our understanding of how modulation of the microenvironment plays a key role. Abstract Cancer stem cells (CSCs) have increasingly been shown to be a crucial element of heterogenous tumors. Although a relatively small component of the population, they increase the resistance to treatment and the likelihood of recurrence. In recent years, it has been shown, across multiple cancer types (e.g., colorectal, breast and prostate), that reciprocal communication between cancer and the microenvironment exists, which is, in part, facilitated by extracellular vesicles (EVs). However, the mechanisms of this method of communication and its influence on CSC populations is less well-understood. Therefore, the aim of this systematic review is to determine the evidence that supports the role of EVs in the manipulation of the tumor microenvironment to promote the survival of CSCs. Embase and PubMed were used to identify all studies on the topic, which were screened using PRISMA guidelines, resulting in the inclusion of 16 studies. These 16 studies reported on the EV content, pathways altered by EVs and therapeutic targeting of CSC through EV-mediated changes to the microenvironment. In conclusion, these studies demonstrated the role of EV-facilitated communication in maintaining CSCs via manipulation of the tumor microenvironment, demonstrating the potential of creating therapeutics to target CSCs. However, further works are needed to fully understand the targetable mechanisms upon which future therapeutics can be based.
Collapse
|
202
|
Guo M, Hao Y, Feng Y, Li H, Mao Y, Dong Q, Cui M. Microglial Exosomes in Neurodegenerative Disease. Front Mol Neurosci 2021; 14:630808. [PMID: 34045943 PMCID: PMC8148341 DOI: 10.3389/fnmol.2021.630808] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/09/2021] [Indexed: 12/20/2022] Open
Abstract
Microglia play an important role in neurodegenerative disease [i.e., Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS)]. These diseases share some similar pathological changes and several microglia-associated processes, including immune response, neuroinflammation, phagocytosis, elimination of synapses et al. Microglia in the central nervous system (CNS) has been described as having both destructive and protective effects in neurological disorders. Besides, considerable evidence also indicates that microglia play a significant role in neurogenesis, neuronal cell death, and synaptic interactions. The communication between microglia and neurons is of vital role in regulating complex functions which are key to appropriate the activity of the brain. Accumulating studies have also demonstrated that exosomes with sizes ranging from 40-100 nm, released by microglia, could serve as key mediators in intercellular signaling. These exosomes, identified in terms of cellular origin in many kinds of biological fluids, exert their effects by delivering specific cargos such as proteins, microRNAs (miRNAs), and mRNAs. It was shown that microglial exosomes could transport to and be uptake by neurons, which may either be beneficial or instead, detrimental to CNS diseases. The focus of this review is to summarize the involvement of microglial exosomes in critical pathologies associated with neurodegenerative disease and how they contribute to these disorders, including PD, AD, and ALS. We also review the application of microglia exosomes as potential biomarkers in monitoring disease progression, as well as focusing on their roles as drug delivery vehicles in treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yining Hao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haiqing Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiting Mao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
203
|
Gao Z, Han X, Zhu Y, Zhang H, Tian R, Wang Z, Cui Y, Wang Z, Niu R, Zhang F. Drug-resistant cancer cell-derived exosomal EphA2 promotes breast cancer metastasis via the EphA2-Ephrin A1 reverse signaling. Cell Death Dis 2021; 12:414. [PMID: 33879771 PMCID: PMC8058342 DOI: 10.1038/s41419-021-03692-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023]
Abstract
Tumor metastasis induced by drug resistance is a major challenge in successful cancer treatment. Nevertheless, the mechanisms underlying the pro-invasive and metastatic ability of drug resistance remain elusive. Exosome-mediated intercellular communications between cancer cells and stromal cells in tumor microenvironment are required for cancer initiation and progression. Recent reports have shown that communications between cancer cells also promote tumor aggression. However, little attention has been regarded on this aspect. Herein, we demonstrated that drug-resistant cell-derived exosomes promoted the invasion of sensitive breast cancer cells. Quantitative proteomic analysis showed that EphA2 was rich in exosomes from drug-resistant cells. Exosomal EphA2 conferred the invasive/metastatic phenotype transfer from drug-resistant cells to sensitive cells. Moreover, exosomal EphA2 activated ERK1/2 signaling through the ligand Ephrin A1-dependent reverse pathway rather than the forward pathway, thereby promoting breast cancer progression. Our findings indicate the key functional role of exosomal EphA2 in the transmission of aggressive phenotype between cancer cells that do not rely on direct cell-cell contact. Our study also suggests that the increase of EphA2 in drug-resistant cell-derived exosomes may be an important mechanism of chemotherapy/drug resistance-induced breast cancer progression.
Collapse
Affiliation(s)
- Zicong Gao
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Xingxing Han
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Yuying Zhu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Ran Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Yanfen Cui
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Zhaosong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
204
|
Plasma-Derived Exosomal microRNA-130a Serves as a Noninvasive Biomarker for Diagnosis and Prognosis of Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:5547911. [PMID: 33953745 PMCID: PMC8068531 DOI: 10.1155/2021/5547911] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/19/2021] [Accepted: 04/07/2021] [Indexed: 02/05/2023]
Abstract
Exosomal microRNAs (miRNAs) are considered as potential stable biomarkers in many types of human cancer, but investigations of plasma-derived exosomal miRNAs in oral squamous cell carcinoma (OSCC) are still lacking. The aim of this study is to evaluate the diagnostic and prognostic values of exosomal miR-130a in OSCC patients. Exosomes were isolated from plasma samples which were collected from 184 OSCC patients before surgery and 196 healthy individuals. Primary OSCC and paired adjacent noncancerous tissues were also obtained from 47 OSCC patients. The expression levels of miR-130a were analyzed by quantitative real-time PCR (qRT-PCR). Our results showed that the expression levels of exosomal miR-130a were significantly higher in OSCC patients than those of the healthy controls (p < 0.0001). Also, the expression of miR-130a was also significantly upregulated in OSCC tissues compared with paired adjacent noncancerous tissues (p < 0.0001). A significant positive correlation was found between exosomal miR-130a and tissue miR-130a levels. Receiver operating characteristic (ROC) analyses yielded an AUC value of 0.812 in discriminating OSCC patients from healthy controls. Furthermore, high levels of exosomal miR-130a were associated with the late T-stage (p=0.024), advanced TNM stage (p=0.003), and poorly differentiated OSCC (p=0.013). Patients with high exosomal miR-130a expression had significantly worse 3-year overall survival (OS) and recurrence-free survival (RFS). Multivariate analysis indicated that exosomal miR-130a was an independent prognostic factor for OS (p=0.001) and RFS (p=0.003). Our results suggest that exosomal miR-130a may serve as a promising diagnostic and prognostic biomarker for OSCC patients.
Collapse
|
205
|
Kletukhina SK, Neustroeva OA, Kurbangaleeva SV, Salafutdinov II, Rogov AM, James V, Rizvanov AA, Gomzikova MO. Storage stability and delivery potential of cytochalasin B induced membrane vesicles. ACTA ACUST UNITED AC 2021; 30:e00616. [PMID: 33996522 PMCID: PMC8090994 DOI: 10.1016/j.btre.2021.e00616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 11/27/2022]
Abstract
Cytochalasin B induced membrane vesicles are a promising tool for clinical application in regenerative medicine and antitumor therapy. Storage temperature is an important factor in maintaining the integrity and delivery potential of cytochalasin B induced membrane vesicles. Freezing at -20 °C in saline is optimal method of cytochalasin B induced membrane vesicles storage. The use of cryoprotectants may help to solve the problem of the aggregation of cytochalasin B induced membrane vesicles during freeze drying.
Cell-free therapies based on extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) are considered as a promising tool for stimulating regeneration and immunomodulation. The need to develop a practical approach for large-scale production of vesicles with homogenous content led to the implementation of cytochalasin B-induced to induce microvesicles (CIMVs) biogenesis. CIMVs mimic natural EVs in size and composition of the surrounding cytoplasmic membrane. Previously we observed that MSC derived CIMVs demonstrate the same therapeutic angiogenic and immunomodulatory activity as the parental MSCs, making them a potentially scalable cell-free therapeutic option. However, little is known about their storage stability and delivery potential. We determined that different storage conditions alter the protein concentration within the solution used to store CIMVs over time, this concided with a decrease in the amount of CIMVs due to gradual degradation. We established that freezing and storage CIMVs in saline at -20 °C reduces degredation and prolongs their shelf life. Additionally, we found that freeze-thawing preserved the CIMVs morphology better than freeze drying and subsequent rehydration which resulted in aggregation of CIMVs. Collectively our data demonstrates for the first time, that the most optimal method of CIMVs storage is freezing at -20 °C, to preserve the CIMVs in the maximum quantity and quality with retention of effective delivery. These findings will benefit the formation of standardized protocols for the use of CIMVs for both basic research and clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Kazan Federal University, Kazan, 420008, Russia.,M.M. Shemyakin-Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Marina O Gomzikova
- Kazan Federal University, Kazan, 420008, Russia.,M.M. Shemyakin-Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| |
Collapse
|
206
|
Lovisolo F, Carton F, Gino S, Migliario M, Renò F. Photobiomodulation induces microvesicle release in human keratinocytes: PI3 kinase-dependent pathway role. Lasers Med Sci 2021; 37:479-487. [PMID: 33826015 DOI: 10.1007/s10103-021-03285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 03/02/2021] [Indexed: 10/21/2022]
Abstract
Microvesicles (MVs, 100-1000 nm diameter) are released into the extracellular environment by mammalian cells. MVs interact with near or remote cells through different mechanisms; in particular, MVs from human keratinocytes accelerate wound healing. Photobiomodulation by laser improves wound healing, but no information is available about its effects on MV release from human keratinocyte. Human-immortalized keratinocytes (human adult low-calcium high-temperature, HaCaT) were starved for 24 h and then irradiated using a 980-nm energy density of 0, 16.2, 32.5, and 48.7 J/cm2. After 24 h, MVs released in the conditioned medium were isolated, stained, and quantified using flow cytometry. MVs were distinguished from exosomes on the basis of their volume (forward scatter signals). In some experiments, phosphatidylinositol 3-kinase (PI-3K) activity, involved in MV release and stimulated by laser light, was inhibited by pre-treating cells with Wortmannin (WRT, 10 μg/mL). MVs were observed in HaCaT-conditioned medium both in basal- and laser-stimulated conditions. Photobiomodulation therapy, also known as PBMT, was able to increase MV release from human keratinocytes reaching a maximum effect at 32.5 J/cm2 with a stimulation of (148.6 ±15.1)% of basal (p<0.001). PI-3K activity inhibition strongly reduced both basal- and laser-induced MV release; but PBMT by laser still increased MV release, compared to basal values in the presence of WRT. In vitro near infrared photobiomodulation increased the releasing of MVs from human keratinocytes, while Wortmannin, a PI-3K inhibitor, negatively affects both basal- and laser-induced releasing. Laser-induced MV release could be a new effect of biostimulation on the wound healing process.
Collapse
Affiliation(s)
- Flavia Lovisolo
- Innovative Research Laboratory for Wound Healing, Health Sciences Department, Università del Piemonte Orientale, via Solaroli 17, 28100, Novara, Italy
| | - Flavia Carton
- Innovative Research Laboratory for Wound Healing, Health Sciences Department, Università del Piemonte Orientale, via Solaroli 17, 28100, Novara, Italy
| | - Sarah Gino
- Innovative Research Laboratory for Wound Healing, Health Sciences Department, Università del Piemonte Orientale, via Solaroli 17, 28100, Novara, Italy
| | - Mario Migliario
- Dental Clinic, Health Sciences Department, Università del Piemonte Orientale, via Solaroli 17, 28100, Novara, Italy
| | - Filippo Renò
- Innovative Research Laboratory for Wound Healing, Health Sciences Department, Università del Piemonte Orientale, via Solaroli 17, 28100, Novara, Italy.
| |
Collapse
|
207
|
Ratajczak MZ, Ratajczak J. Innate Immunity Communicates Using the Language of Extracellular Microvesicles. Stem Cell Rev Rep 2021; 17:502-510. [PMID: 33634435 PMCID: PMC7906088 DOI: 10.1007/s12015-021-10138-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
The innate immunity system and extracellular microvesicles (ExMVs) both emerged early in the evolution of life, which is why its innate immunity cellular arm and its soluble-component arm learned, understood, and adapted to the "language" of ExMVs. This was most likely the first language of cell-cell communication during evolution, which existed before more specific intercellular crosstalk involving specific ligands and receptors emerged. ExMVs are involved in several processes in the body, including immune and coagulation responses, which are part of inflammation. In this review we will briefly highlight what is known about how ExMVs regulate the function of the cellular arm of innate immunity, including macrophages, monocytes, granulocytes, natural killer cells, and dendritic cells, and affect the soluble components of this system, which consists of the complement cascade (ComC) and soluble, circulating, pattern-recognition receptors (collectins, ficolins, and pentaxrins). These effects are direct, due to the fact that ExMVs affect the biological functions of innate immunity cells and may directly interact with soluble components of this system. Moreover, by activating coagulation proteases, ExMVs may also indirectly activate the ComC. In this review, we will use the term "extracellular microvesicles" (ExMVs) to refer to these small, spheroidal blebs of different sizes, which are surrounded by a membrane lipid layer. We will focus on the role of both ExMVs released during cell-surface membrane budding and smaller ExMVs, known as exosomes, which are derived from the budding of the endosomal membrane compartment. Finally, we will provide a brief update on the potential therapeutic applications of ExMVs, with a special emphasis on innate immunity.
Collapse
Affiliation(s)
- Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warszawa, Poland
| | - Janina Ratajczak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warszawa, Poland
| |
Collapse
|
208
|
Liu C, Li L, Wang M, Shui S, Yao H, Sui C, Zhang H. Endometrial extracellular vesicles of recurrent implantation failure patients inhibit the proliferation, migration, and invasion of HTR8/SVneo cells. J Assist Reprod Genet 2021; 38:825-833. [PMID: 33521905 PMCID: PMC8079592 DOI: 10.1007/s10815-021-02093-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/27/2021] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Endometrial extracellular vesicles are essential in regulating trophoblasts' function. This study aims to investigate whether endometrial extracellular vesicles (EVs) from recurrent implantation failure (RIF) patients inhibit the proliferation, invasion, and migration of HTR8/SVneo cells. METHODS Eighteen RIF patients and thirteen fertile women were recruited for endometria collection. Endometrial cells isolated from the endometria were cultured and modulated by hormones, and the conditioned medium was used for EV isolation. EVs secreted by the endometrial cells of RIF patients (RIF-EVs) or fertile women (FER-EVs) were determined by Western blotting, nanoparticle tracking analysis, and transmission electron microscopy. Fluorescence-labeled EVs were used to visualize internalization by HTR8/SVneo cells. RIF-EVs and FER-EVs were co-cultured with HTR8/SVneo cells. Cell Counting Kit-8, transwell invasion, and wound closure assays were performed to determine cellular proliferation, invasion, and migration, respectively, in different treatments. RESULTS RIF-EVs and FER-EVs were bilayer membrane vesicles, ranging from 100 to 150 nm in size, that expressed the classic EV markers Alix and CD9. RIF-EVs and FER-EVs were internalized by HTR8/SVneo cells within 2 h. The proliferation rate in the FER-EV group was significantly higher than that in the RIF-EV group at 20 μg/mL. Moreover, the invasion and migration capacity of trophoblast cells were decreased in the RIF-EV group relative to the FER-EV group at 20 μg/mL. CONCLUSION Endometrial EVs from RIF patients inhibited the functions of trophoblasts by decreasing their proliferation, migration, and invasive capacity. Such dysregulations induced by RIF-EVs may provide novel insights for better understanding the pathogenesis of implantation failure.
Collapse
Affiliation(s)
- Chang Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Linshuang Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Shike Shui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Haixia Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Cong Sui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
209
|
Standardized procedure to measure the size distribution of extracellular vesicles together with other particles in biofluids with microfluidic resistive pulse sensing. PLoS One 2021; 16:e0249603. [PMID: 33793681 PMCID: PMC8016234 DOI: 10.1371/journal.pone.0249603] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/19/2021] [Indexed: 01/15/2023] Open
Abstract
The particle size distribution (PSD) of extracellular vesicles (EVs) and other submicron particles in biofluids is commonly measured by nanoparticle tracking analysis (NTA) and tunable resistive pulse sensing (TRPS). A new technique for measuring the PSD is microfluidic resistive pulse sensing (MRPS). Because specific guidelines for measuring EVs together with other particles in biofluids with MRPS are lacking, we developed an operating procedure to reproducibly measure the PSD. The PSDs of particles in human plasma, conditioned medium of PC3 prostate cancer cell line (PC3 CM), and human urine were measured with MRPS (nCS1, Spectradyne LLC) to investigate: (i) the optimal diluent that reduces the interfacial tension of the sample while keeping EVs intact, (ii) the lower limit of detection (LoD) of particle size, (iii) the reproducibility of the PSD, (iv) the optimal dilution for measuring the PSD, and (v) the agreement in measured concentration between microfluidic cartridges with overlapping detection ranges. We found that the optimal diluent is 0.1% bovine serum albumin (w/v) in Dulbecco’s phosphate-buffered saline. Based on the shape of the PSD, which is expected to follow a power-law function within the full detection range, we obtained a lower LoD of 75 nm for plasma and PC3 CM and 65 nm for urine. Normalized PSDs are reproducible (R2 > 0.950) at dilutions between 10–100x for plasma, 5–20x for PC3 CM, and 2–4x for urine. Furthermore, sample dilution does not impact the dilution-corrected concentration when the microfluidic cartridges are operated within their specified concentration ranges. PSDs from microfluidic cartridges with overlapping detection ranges agreed well (R2 > 0.936) and when combined the overall PSD spanned 5 orders of magnitude of measured concentration. Based on these findings, we have developed operating guidelines to reproducibly measure the PSD of EVs together with other particles in biofluids with MRPS.
Collapse
|
210
|
Yokoi A, Ochiya T. Exosomes and extracellular vesicles: Rethinking the essential values in cancer biology. Semin Cancer Biol 2021; 74:79-91. [PMID: 33798721 DOI: 10.1016/j.semcancer.2021.03.032] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/17/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) such as exosomes are released by all living cells and contain diverse bioactive molecules, including nucleic acids, proteins, lipids, and metabolites. Accumulating evidence of EV-related functions has revealed that these tiny vesicles can mediate specific cell-to-cell communication. Within the tumor microenvironment, diverse cells are actively interacting with their surroundings via EVs facilitating tumor malignancy by regulating malignant cascades including angiogenesis, immune modulation, and metastasis. This review summarizes the recent studies of fundamental understandings of EVs from the aspect of EV heterogeneity and highlights the role of EVs in the various steps from oncogenic to metastatic processes. The recognition of EV subtypes is necessary to identify which pathways can be affected by EVs and which subtypes can be targeted in therapeutic approaches or liquid biopsies.
Collapse
Affiliation(s)
- Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
211
|
Vilcaes AA, Chanaday NL, Kavalali ET. Interneuronal exchange and functional integration of synaptobrevin via extracellular vesicles. Neuron 2021; 109:971-983.e5. [PMID: 33513363 PMCID: PMC7979516 DOI: 10.1016/j.neuron.2021.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 12/06/2020] [Accepted: 01/07/2021] [Indexed: 12/25/2022]
Abstract
Recent studies have investigated the composition and functional effects of extracellular vesicles (EVs) secreted by a variety of cell types. However, the mechanisms underlying the impact of these vesicles on neurotransmission remain unclear. Here, we isolated EVs secreted by rat and mouse hippocampal neurons and found that they contain synaptic-vesicle-associated proteins, in particular the vesicular SNARE (soluble N-ethylmaleimide-sensitive factor [NSF]-attachment protein receptor) synaptobrevin (also called VAMP). Using a combination of electrophysiology and live-fluorescence imaging, we demonstrate that this extracellular pool of synaptobrevins can rapidly integrate into the synaptic vesicle cycle of host neurons via a CD81-dependent process and selectively augment inhibitory neurotransmission as well as specifically rescue neurotransmission in synapses deficient in synaptobrevin. These findings uncover a novel means of interneuronal communication and functional coupling via exchange of vesicular SNAREs.
Collapse
Affiliation(s)
- A Alejandro Vilcaes
- CONICET, Universidad Nacional de Córdoba, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba X5000HUA, Argentina; Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Córdoba X5000HUA, Argentina; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Natali L Chanaday
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA.
| | - Ege T Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA.
| |
Collapse
|
212
|
Gao Y, Qin Y, Wan C, Sun Y, Meng J, Huang J, Hu Y, Jin H, Yang K. Small Extracellular Vesicles: A Novel Avenue for Cancer Management. Front Oncol 2021; 11:638357. [PMID: 33791224 PMCID: PMC8005721 DOI: 10.3389/fonc.2021.638357] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles are small membrane particles derived from various cell types. EVs are broadly classified as ectosomes or small extracellular vesicles, depending on their biogenesis and cargoes. Numerous studies have shown that EVs regulate multiple physiological and pathophysiological processes. The roles of small extracellular vesicles in cancer growth and metastasis remain to be fully elucidated. As endogenous products, small extracellular vesicles are an ideal drug delivery platform for anticancer agents. However, several aspects of small extracellular vesicle biology remain unclear, hindering the clinical implementation of small extracellular vesicles as biomarkers or anticancer agents. In this review, we summarize the utility of cancer-related small extracellular vesicles as biomarkers to detect early-stage cancers and predict treatment outcomes. We also review findings from preclinical and clinical studies of small extracellular vesicle-based cancer therapies and summarize interventional clinical trials registered in the United States Food and Drug Administration and the Chinese Clinical Trials Registry. Finally, we discuss the main challenges limiting the clinical implementation of small extracellular vesicles and recommend possible approaches to address these challenges.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
213
|
Camussi G. Exosomes and Microvesicles: from Stem Cell Biology to Translation in Human Diseases. Stem Cell Rev Rep 2021; 17:303-304. [PMID: 33713289 DOI: 10.1007/s12015-021-10148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
214
|
Willysson A, Ståhl AL, Karpman D. Isolation and Characterization of Shiga Toxin-Associated Microvesicles. Methods Mol Biol 2021; 2291:207-228. [PMID: 33704755 DOI: 10.1007/978-1-0716-1339-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Microvesicles are shed from cell surfaces during infectious or inflammatory conditions and may contribute to the pathogenesis of disease. During Shiga toxin-producing Escherichia coli (STEC) infection, microvesicles are released from blood cells. These microvesicles play a part in inflammation, thrombosis, hemolysis, and the transfer of the main virulence factor of STEC strains, Shiga toxin, to target organ cells. This chapter describes how to isolate blood cell- and cell culture-derived microvesicles from plasma or cell culture medium, respectively, and how to characterize these microvesicles by various methods, with special focus on Shiga toxin-associated microvesicles.
Collapse
Affiliation(s)
- Annie Willysson
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anne-Lie Ståhl
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
215
|
Biodistribution of surfactant-free poly(lactic-acid) nanoparticles and uptake by endothelial cells and phagocytes in zebrafish: Evidence for endothelium to macrophage transfer. J Control Release 2021; 331:228-245. [PMID: 33444668 DOI: 10.1016/j.jconrel.2021.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/16/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
In the development of therapeutic nanoparticles (NP), there is a large gap between in vitro testing and in vivo experimentation. Despite its prominence as a model, the mouse shows severe limitations for imaging NP and the cells with which they interact. Recently, the transparent zebrafish larva, which is well suited for high-resolution live-imaging, has emerged as a powerful alternative model to investigate the in vivo behavior of NP. Poly(D,L lactic acid) (PLA) is widely accepted as a safe polymer to prepare therapeutic NP. However, to prevent aggregation, many NP require surfactants, which may have undesirable biological effects. Here, we evaluate 'safe-by-design', surfactant-free PLA-NP that were injected intravenously into zebrafish larvae. Interaction of fluorescent NPs with different cell types labelled in reporter animals could be followed in real-time at high resolution; furthermore, by encapsulating colloidal gold into the matrix of PLA-NP we could follow their fate in more detail by electron microscopy, from uptake to degradation. The rapid clearance of fluorescent PLA-NP from the circulation coincided with internalization by endothelial cells lining the whole vasculature and macrophages. After 30 min, when no NP remained in circulation, we observed that macrophages continued to internalize significant amounts of NP. More detailed video-imaging revealed a new mechanism of NP transfer where NP are transmitted along with parts of the cytoplasm from endothelial cells to macrophages.
Collapse
|
216
|
Kidney Mesenchymal Stem Cell-derived Extracellular Vesicles Engineered to Express Erythropoietin Improve Renal Anemia in Mice with Chronic Kidney Disease. Stem Cell Rev Rep 2021; 18:980-992. [PMID: 33651336 DOI: 10.1007/s12015-021-10141-x] [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] [Accepted: 02/17/2021] [Indexed: 12/29/2022]
Abstract
Extracellular vesicles (EVs) shed from kidney mesenchymal stem cells (KMSCs) show protective effects against acute kidney injury and progressive kidney fibrosis via mRNA transfer. Previous studies report improvement of renal anemia following administration of genetically modified MSCs or peritoneal mesothelial cells that secrete erythropoietin (EPO). Here, we determined whether EPO-secreting KMSC-derived EVs (EPO(+)-EVs) can improve renal anemia in mouse models of chronic kidney disease (CKD). The mouse CKD and renal anemia model was induced by electrocoagulation of the right renal cortex and sequential left nephrectomy. At six weeks post-nephrectomy, we observed significantly lower hemoglobin (10.4 ± 0.2 vs. 13.2 ± 0.2 g/dL) and significantly higher blood urea nitrogen and serum creatinine levels in CKD mice relative to controls (60.5 ± 0.5 and 0.37 ± 0.09 mg/dL vs. 19.9 ± 0.5 and 0.12 ± 0.02 mg/dL, respectively). Genetically engineered EPO(+)-KMSCs secreted 71 IU/mL EPO/106 cells/24 h in vitro, and EPO(+)-EVs isolated by differential ultracentrifugation expressed EPO mRNA and horizontally transferred EPO mRNA into target cells in vitro and in vivo. Furthermore, at two weeks post-injection of EPO(+)-KMSCs or EPO(+)-EVs into CKD mice with renal anemia, we observed significant increases in hemoglobin levels (11.7 ± 0.2 and 11.5 ± 0.2 vs. 10.1 ± 0.2 g/dL, respectively) and significantly lower serum creatinine levels at eight weeks in comparison to mice receiving vehicle control (0.30 ± 0.00 and 0.23 ± 0.03 vs. 0.43 ± 0.06 mg/dL, respectively). These results demonstrate that intraperitoneal administration of EPO(+)-EVs significantly increased hemoglobin levels and renal function in CKD mice, suggesting the efficacy of these genetically engineered EVs as a promising novel strategy for the treatment of renal anemia.
Collapse
|
217
|
Yang ZJ, Zhang LL, Bi QC, Gan LJ, Wei MJ, Hong T, Tan RJ, Lan XM, Liu LH, Han XJ, Jiang LP. Exosomal connexin 43 regulates the resistance of glioma cells to temozolomide. Oncol Rep 2021; 45:44. [PMID: 33649836 PMCID: PMC7934218 DOI: 10.3892/or.2021.7995] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma is the most common and aggressive brain tumor and it is characterized by a high mortality rate. Temozolomide (TMZ) is an effective chemotherapy drug for glioblastoma, but the resistance to TMZ has come to represent a major clinical problem, and its underlying mechanism has yet to be elucidated. In the present study, the role of exosomal connexin 43 (Cx43) in the resistance of glioma cells to TMZ and cell migration was investigated. First, higher expression levels of Cx43 were detected in TMZ‑resistant U251 (U251r) cells compared with those in TMZ‑sensitive (U251s) cells. Exosomes from U251s or U251r cells (sExo and rExo, respectively) were isolated. It was found that the expression of Cx43 in rExo was notably higher compared with that in sExo, whereas treatment with rExo increased the expression of Cx43 in U251s cells. Additionally, exosomes stained with dioctadecyloxacarbocyanine (Dio) were used to visualized exosome uptake by glioma cells. It was observed that the uptake of Dio‑stained rExo in U251s cells was more prominent compared with that of Dio‑stained sExo, while 37,43Gap27, a gap junction mimetic peptide directed against Cx43, alleviated the rExo uptake by cells. Moreover, rExo increased the IC50 of U251s to TMZ, colony formation and Bcl‑2 expression, but decreased Bax and cleaved caspase‑3 expression in U251s cells. 37,43Gap27 efficiently inhibited these effects of rExo on U251s cells. Finally, the results of the wound healing and Transwell assays revealed that rExo significantly enhanced the migration of U251s cells, whereas 37,43Gap27 significantly attenuated rExo‑induced cell migration. Taken together, these results indicate the crucial role of exosomal Cx43 in chemotherapy resistance and migration of glioma cells, and suggest that Cx43 may hold promise as a therapeutic target for glioblastoma in the future.
Collapse
Affiliation(s)
- Zhang-Jian Yang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Le-Ling Zhang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiu-Chen Bi
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Jun Gan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Min-Jun Wei
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tao Hong
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ren-Jie Tan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xue-Mei Lan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Hua Liu
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Jian Han
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Ping Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
218
|
Tynecka M, Moniuszko M, Eljaszewicz A. Old Friends with Unexploited Perspectives: Current Advances in Mesenchymal Stem Cell-Based Therapies in Asthma. Stem Cell Rev Rep 2021; 17:1323-1342. [PMID: 33649900 PMCID: PMC7919631 DOI: 10.1007/s12015-021-10137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have a great regenerative and immunomodulatory potential that was successfully tested in numerous pre-clinical and clinical studies of various degenerative, hematological and inflammatory disorders. Over the last few decades, substantial immunoregulatory effects of MSC treatment were widely observed in different experimental models of asthma. Therefore, it is tempting to speculate that stem cell-based treatment could become an attractive means to better suppress asthmatic airway inflammation, especially in subjects resistant to currently available anti-inflammatory therapies. In this review, we discuss mechanisms accounting for potent immunosuppressive properties of MSCs and the rationale for their use in asthma. We describe in detail an intriguing interplay between MSCs and other crucial players in the immune system as well as lung microenvironment. Finally, we reveal the potential of MSCs in maintaining airway epithelial integrity and alleviating lung remodeling.
Collapse
Affiliation(s)
- Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
- Department of Allergology and Internal Medicine, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, Białystok, 15-276, Poland.
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
| |
Collapse
|
219
|
Di J, Yang M, Zhou H, Li M, Zhao J. MicroRNA-21-containing microvesicles from tubular epithelial cells promote cardiomyocyte hypertrophy. Ren Fail 2021; 43:391-400. [PMID: 33632070 PMCID: PMC7919913 DOI: 10.1080/0886022x.2021.1891098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Cardiomyocyte hypertrophy has been reported as one of the important mechanisms for cardiovascular disease (CVD) in patients with chronic kidney disease (CKD). MiroRNA-21(miR-21) was determined to play an important role in myocardial hypertrophy. However, the role of microvesicles (MVs) containing miR-21 in CKD-related cardiomyocyte hypertrophy remains largely unexplored. Methods Renal tubular epithelial cells were stimulated by transforming growth factor (TGF-β1), and the conditioned medium was extracted by differential centrifugation. Renal tubular epithelial cells were labeled with Dil-C18 dye and the recipient cardiomyocytes were observed by fluorescence microscope. MiR-21 level in MVs was detected by qRT-PCR, and the length and diameter of cardiomyocytes were measured by microscope. BCA protein kit and ANP kit were used to detect the content of cell protein and the level of ANP. MiR-21 inhibitor was transfected into cardiomyocytes to observe the effect of miR-21 on myocardial hypertrophy. Results TGF-β1 could induce donor renal tubular epithelial cells to produce MVs and delivered into cardiomyocytes, followed by the diameter, protein concentration and ANP content of cardiomyocytes significantly increased. Meanwhile, MiR-21 levels were markedly increased in MVs isolated from donor renal tubular epithelial cells and recipient cardiomyocytes. Pre-transfection of miR-21 inhibitors could inhibit MV-induced cardiomyocyte hypertrophy. Conclusion Tubular cells could secrete miR-21 by MVs and deliver it into recipient cardiomyocytes to induce cardiomyocyte hypertrophy. It might shed a new light on the mechanism and treatment of CKD-related cardiac dysfunction.
Collapse
Affiliation(s)
- Jia Di
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Min Yang
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Hua Zhou
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Min Li
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Jiabi Zhao
- Department of Pathology, The Second People's Hospital of Changzhou, Changzhou, China
| |
Collapse
|
220
|
Nassir CMNCM, Ghazali MM, Hashim S, Idris NS, Yuen LS, Hui WJ, Norman HH, Gau CH, Jayabalan N, Na Y, Feng L, Ong LK, Abdul Hamid H, Ahamed HN, Mustapha M. Diets and Cellular-Derived Microparticles: Weighing a Plausible Link With Cerebral Small Vessel Disease. Front Cardiovasc Med 2021; 8:632131. [PMID: 33718454 PMCID: PMC7943466 DOI: 10.3389/fcvm.2021.632131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
Cerebral small vessel disease (CSVD) represents a spectrum of pathological processes of various etiologies affecting the brain microcirculation that can trigger neuroinflammation and the subsequent neurodegenerative cascade. Prevalent with aging, CSVD is a recognized risk factor for stroke, vascular dementia, Alzheimer disease, and Parkinson disease. Despite being the most common neurodegenerative condition with cerebrocardiovascular axis, understanding about it remains poor. Interestingly, modifiable risk factors such as unhealthy diet including high intake of processed food, high-fat foods, and animal by-products are known to influence the non-neural peripheral events, such as in the gastrointestinal tract and cardiovascular stress through cellular inflammation and oxidation. One key outcome from such events, among others, includes the cellular activations that lead to elevated levels of endogenous cellular-derived circulating microparticles (MPs). MPs can be produced from various cellular origins including leukocytes, platelets, endothelial cells, microbiota, and microglia. MPs could act as microthrombogenic procoagulant that served as a plausible culprit for the vulnerable end-artery microcirculation in the brain as the end-organ leading to CSVD manifestations. However, little attention has been paid on the potential role of MPs in the onset and progression of CSVD spectrum. Corroboratively, the formation of MPs is known to be influenced by diet-induced cellular stress. Thus, this review aims to appraise the body of evidence on the dietary-related impacts on circulating MPs from non-neural peripheral origins that could serve as a plausible microthrombosis in CSVD manifestation as a precursor of neurodegeneration. Here, we elaborate on the pathomechanical features of MPs in health and disease states; relevance of dietary patterns on MP release; preclinical studies pertaining to diet-based MPs contribution to disease; MP level as putative surrogates for early disease biomarkers; and lastly, the potential of MPs manipulation with diet-based approach as a novel preventive measure for CSVD in an aging society worldwide.
Collapse
Affiliation(s)
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Sabarisah Hashim
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Nur Suhaila Idris
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Lee Si Yuen
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Wong Jia Hui
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Haziq Hazman Norman
- Anatomy Unit, International Medical School (IMS), Management and Science University (MSU), Shah Alam, Malaysia
| | - Chuang Huei Gau
- Department of Psychology and Counselling, Faculty of Arts and Social Science, Universiti Tunku Abdul Rahman (UTAR), Kampar, Malaysia
| | - Nanthini Jayabalan
- Translational Neuroscience Lab, University of Queensland (UQ), Centre for Clinical Research, The University of Queensland, Herston, QLD, Australia
| | - Yuri Na
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Linqing Feng
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, National Health and Medical Research Council (NHMRC), Heidelberg, VIC, Australia
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Haja Nazeer Ahamed
- Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, India
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Hospital Universiti Sains Malaysia, Jalan Raja Perempuan Zainab II, Kubang Kerian, Malaysia
| |
Collapse
|
221
|
Lorico A, Lorico-Rappa M, Karbanová J, Corbeil D, Pizzorno G. CD9, a tetraspanin target for cancer therapy? Exp Biol Med (Maywood) 2021; 246:1121-1138. [PMID: 33601913 DOI: 10.1177/1535370220981855] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present minireview, we intend to provide a brief history of the field of CD9 involvement in oncogenesis and in the metastatic process of cancer, considering its potential value as a tumor-associated antigenic target. Over the years, CD9 has been identified as a favorable prognostic marker or predictor of metastatic potential depending on the cancer type. To understand its implications in cancer beside its use as an antigenic biomarker, it is essential to know its physiological functions, including its molecular partners in a given cell system. Moreover, the discovery that CD9 is one of the most specific and broadly expressed markers of extracellular membrane vesicles, nanometer-sized entities that are released into extracellular space and various physiological body fluids and play a role in intercellular communication under physiological and pathological conditions, notably the establishment of cancer metastases, has added a new dimension to our knowledge of CD9 function in cancer. Here, we will discuss these issues as well as the possible cancer therapeutic implications of CD9, their limitations, and pitfalls.
Collapse
Affiliation(s)
- Aurelio Lorico
- Touro University College of Medicine, Henderson, NV 89014, USA.,Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | | | - Jana Karbanová
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Denis Corbeil
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Giuseppe Pizzorno
- University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Erlanger Health System, Chattanooga, TN 37403 , USA
| |
Collapse
|
222
|
Altschuler J, Stockert JA, Kyprianou N. Non-Coding RNAs Set a New Phenotypic Frontier in Prostate Cancer Metastasis and Resistance. Int J Mol Sci 2021; 22:ijms22042100. [PMID: 33672595 PMCID: PMC7924036 DOI: 10.3390/ijms22042100] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) mortality remains a significant public health problem, as advanced disease has poor survivability due to the development of resistance in response to both standard and novel therapeutic interventions. Therapeutic resistance is a multifaceted problem involving the interplay of a number of biological mechanisms including genetic, signaling, and phenotypic alterations, compounded by the contributions of a tumor microenvironment that supports tumor growth, invasiveness, and metastasis. The androgen receptor (AR) is a primary regulator of prostate cell growth, response and maintenance, and the target of most standard PCa therapies designed to inhibit AR from interacting with androgens, its native ligands. As such, AR remains the main driver of therapeutic response in patients with metastatic castration-resistant prostate cancer (mCRPC). While androgen deprivation therapy (ADT), in combination with microtubule-targeting taxane chemotherapy, offers survival benefits in patients with mCRPC, therapeutic resistance invariably develops, leading to lethal disease. Understanding the mechanisms underlying resistance is critical to improving therapeutic outcomes and also to the development of biomarker signatures of predictive value. The interconversions between epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET) navigate the prostate tumor therapeutic response, and provide a novel targeting platform in overcoming therapeutic resistance. Both microRNA (miRNA)- and long non-coding RNA (lncRNA)-mediated mechanisms have been associated with epigenetic changes in prostate cancer. This review discusses the current evidence-based knowledge of the role of the phenotypic transitions and novel molecular determinants (non-coding RNAs) as contributors to the emergence of therapeutic resistance and metastasis and their integrated predictive value in prostate cancer progression to advanced disease.
Collapse
Affiliation(s)
- Joshua Altschuler
- Department of Urology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.A.); (J.A.S.)
| | - Jennifer A. Stockert
- Department of Urology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.A.); (J.A.S.)
| | - Natasha Kyprianou
- Department of Urology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.A.); (J.A.S.)
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence:
| |
Collapse
|
223
|
Jayasinghe MK, Tan M, Peng B, Yang Y, Sethi G, Pirisinu M, Le MTN. New approaches in extracellular vesicle engineering for improving the efficacy of anti-cancer therapies. Semin Cancer Biol 2021; 74:62-78. [PMID: 33609665 DOI: 10.1016/j.semcancer.2021.02.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/11/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Cancer is a disease that evolves continuously with unpredictable outcomes. Although conventional chemotherapy can display significant antitumor effects, the lack of specificity and poor bioavailability remain major concerns in cancer therapy. Moreover, with the advent of novel anti-cancer gene therapies, there is an urgent need for drug delivery vectors capable of bypassing cellular barriers and efficiently transferring therapeutic cargo to recipient cells. A number of drug delivery systems have been proposed to overcome these limitations, but their successful clinical translation has been hampered by the onset of unexpected side effects and associated toxicities. The application of extracellular vesicles (EVs), a class of naturally released, cell-derived particles, as drug delivery vectors presents a breakthrough in nanomedicine, taking into account their biocompatibility and natural role in intercellular communication. Combining the advantageous intrinsic properties of EVs with surface functionalization and the encapsulation of drugs allows for a new class of engineered EVs that serve as effective therapeutic carriers. Here, we describe the various successful approaches involving the application of engineered EVs as bio-derived drug delivery vectors in cancer therapy. The latest and most effective strategies of engineering EVs to improve drug loading, stealth properties and tumour targeting capabilities of EVs are debated. Finally, current obstacles and future perspectives of smart engineered EVs are discussed.
Collapse
Affiliation(s)
- Migara Kavishka Jayasinghe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore; Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong
| | - Melissa Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Boya Peng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Yuqi Yang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marco Pirisinu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong.
| | - Minh T N Le
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
224
|
Wang X, HuangFu C, Zhu X, Liu J, Gong X, Pan Q, Ma X. Exosomes and Exosomal MicroRNAs in Age-Associated Stroke. Curr Vasc Pharmacol 2021; 19:587-600. [PMID: 33563154 DOI: 10.2174/1570161119666210208202621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/04/2021] [Accepted: 01/18/2021] [Indexed: 11/22/2022]
Abstract
Aging has been considered to be the most important non-modifiable risk factor for stroke and death. Changes in circulation factors in the systemic environment, cellular senescence and artery hypertension during human ageing have been investigated. Exosomes are nanosize membrane vesicles that can regulate target cell functions via delivering their carried bioactive molecules (e.g. protein, mRNA, and microRNAs). In the central nervous system, exosomes and exosomal microRNAs play a critical role in regulating neurovascular function, and are implicated in the initiation and progression of stroke. MicroRNAs are small non-coding RNAs that have been reported to play critical roles in various biological processes. Recently, evidence has shown that microRNAs are packaged into exosomes and can be secreted into the systemic and tissue environment. Circulating microRNAs participate in cellular senescence and contribute to age-associated stroke. Here, we provide an overview of current knowledge on exosomes and their carried microRNAs in the regulation of cellular and organismal ageing processes, demonstrating the potential role of exosomes and their carried microRNAs in age-associated stroke.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| | - Changmei HuangFu
- Department of Geriatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| | - Xiudeng Zhu
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| | - Jiehong Liu
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| | - Xinqin Gong
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| | - Qunwen Pan
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| | - Xiaotang Ma
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, . China
| |
Collapse
|
225
|
Nederveen JP, Warnier G, Di Carlo A, Nilsson MI, Tarnopolsky MA. Extracellular Vesicles and Exosomes: Insights From Exercise Science. Front Physiol 2021; 11:604274. [PMID: 33597890 PMCID: PMC7882633 DOI: 10.3389/fphys.2020.604274] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
The benefits of exercise on health and longevity are well-established, and evidence suggests that these effects are partially driven by a spectrum of bioactive molecules released into circulation during exercise (e.g., exercise factors or 'exerkines'). Recently, extracellular vesicles (EVs), including microvesicles (MVs) and exosomes or exosome-like vesicles (ELVs), were shown to be secreted concomitantly with exerkines. These EVs have therefore been proposed to act as cargo carriers or 'mediators' of intercellular communication. Given these findings, there has been a rapidly growing interest in the role of EVs in the multi-systemic, adaptive response to exercise. This review aims to summarize our current understanding of the effects of exercise on MVs and ELVs, examine their role in the exercise response and long-term adaptations, and highlight the main methodological hurdles related to blood collection, purification, and characterization of ELVs.
Collapse
Affiliation(s)
- Joshua P Nederveen
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Geoffrey Warnier
- Institut of Neuroscience, UCLouvain, Université catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Alessia Di Carlo
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Mats I Nilsson
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada.,Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| |
Collapse
|
226
|
Barok M, Puhka M, Yazdi N, Joensuu H. Extracellular vesicles as modifiers of antibody-drug conjugate efficacy. J Extracell Vesicles 2021; 10:e12070. [PMID: 33613875 PMCID: PMC7881363 DOI: 10.1002/jev2.12070] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of anti-cancer drugs that consist of a monoclonal antibody, a highly potent small-molecule cytotoxic drug, and a chemical linker between the two. ADCs can selectively deliver cytotoxic drugs to cancer cells leading to a reduced systemic exposure and a wider therapeutic window. To date, nine ADCs have received marketing approval, and over 100 are being investigated in nearly 600 clinical trials. The target antigens of at least eight out of the nine approved anti-cancer ADCs and of 69 investigational ADCs are present on extracellular vesicles (EVs) (tiny particles produced by almost all types of cells) that may carry their contents into local and distant cells. Therefore, the EVs have a potential to mediate both the anti-cancer effects and the adverse effects of ADCs. In this overview, we discuss the mechanisms of action of ADCs and the resistance mechanisms to them, the EV-mediated resistance mechanisms to small molecule anti-cancer drugs and anti-cancer monoclonal antibodies, and the EVs as modifiers of ADC efficacy and safety.
Collapse
Affiliation(s)
- Mark Barok
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Laboratory of Molecular OncologyUniversity of HelsinkiBiomedicumHelsinkiFinland
| | - Maija Puhka
- Institute for Molecular Medicine FIMMEV and HiPrep CoreUniversity of HelsinkiHelsinkiFinland
| | - Narjes Yazdi
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Laboratory of Molecular OncologyUniversity of HelsinkiBiomedicumHelsinkiFinland
| | - Heikki Joensuu
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Laboratory of Molecular OncologyUniversity of HelsinkiBiomedicumHelsinkiFinland
| |
Collapse
|
227
|
Li D, Jia H, Zhang H, Lv M, Liu J, Zhang Y, Huang T, Huang B. TLR4 signaling induces the release of microparticles by tumor cells that regulate inflammatory cytokine IL-6 of macrophages via microRNA let-7b. Oncoimmunology 2021; 1:687-693. [PMID: 22934260 PMCID: PMC3429572 DOI: 10.4161/onci.19854] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tumor cells expressing TLRs is generally recognized to mediate tumor inflammation. However, whether and how tumor TLR signaling pathways negatively regulate tumor inflammation remains unclear. In this report, we find that TLR4 signaling of H22 hepatocarcinoma tumor cells is transduced through MyD88 pathway to actin cytoskeletons, leading to the release of microparticles (MPs), the cellular membrane-derived vesicles. As a result, tumor macrophages take up MPs and acquire MP-contained microRNA let-7b, which attenuates tumor inflammation by targeting proinflammatory cytokine IL-6. Thus, tumor TLR signaling, contrary to the original promoting effect, may play an opposite role in downregulating tumor inflammation through MP pathways.
Collapse
Affiliation(s)
- Dapeng Li
- Department of Biochemistry and Molecular Biology; Tongji Medical College; Huazhong University of Science and Technology; Wuhan, China ; Department of Breast and Thyroid Surgery; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
228
|
Affiliation(s)
- Peng Shi
- Department of Biomedical Engineering The Pennsylvania State University University Park PA 16802 USA
| | - Yong Wang
- Department of Biomedical Engineering The Pennsylvania State University University Park PA 16802 USA
| |
Collapse
|
229
|
Shi P, Wang Y. Synthetic DNA for Cell-Surface Engineering. Angew Chem Int Ed Engl 2021; 60:11580-11591. [PMID: 33006229 DOI: 10.1002/anie.202010278] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/29/2020] [Indexed: 12/14/2022]
Abstract
The cell membrane is not only a physical barrier, but also a functional organelle that regulates the communication between a cell and its environment. The ability to functionalize the cell membrane with synthetic molecules or nanostructures would advance cellular functions beyond what evolution has provided. The aim of this Minireview is to introduce recent progress in using synthetic DNA and DNA-based nanostructures for cell-surface engineering. We first introduce chemical conjugation and physical binding methods for monovalent and polyvalent surface engineering. We then introduce the application of these methods for either the promotion or inhibition of cell-environment communication in numerous applications, including the promotion of cell-cell recognition, regulation of intracellular pathways, protection of therapeutic cells, and sensing of the intracellular and extracellular microenvironments. Lastly, we summarize current challenges existing in this area and potential solutions to solve these challenges.
Collapse
Affiliation(s)
- Peng Shi
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
230
|
Boosting anti-PD-1 therapy with metformin-loaded macrophage-derived microparticles. Nat Commun 2021; 12:440. [PMID: 33469052 PMCID: PMC7815730 DOI: 10.1038/s41467-020-20723-x] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
The main challenges for programmed cell death 1(PD-1)/PD-1 ligand (PD-L1) checkpoint blockade lie in a lack of sufficient T cell infiltration, tumor immunosuppressive microenvironment, and the inadequate tumor accumulation and penetration of anti-PD-1/PD-L1 antibody. Resetting tumor-associated macrophages (TAMs) is a promising strategy to enhance T-cell antitumor immunity and ameliorate tumor immunosuppression. Here, mannose-modified macrophage-derived microparticles (Man-MPs) loading metformin (Met@Man-MPs) are developed to efficiently target to M2-like TAMs to repolarize into M1-like phenotype. Met@Man-MPs-reset TAMs remodel the tumor immune microenvironment by increasing the recruitment of CD8+ T cells into tumor tissues and decreasing immunosuppressive infiltration of myeloid-derived suppressor cells and regulatory T cells. More importantly, the collagen-degrading capacity of Man-MPs contributes to the infiltration of CD8+ T cells into tumor interiors and enhances tumor accumulation and penetration of anti-PD-1 antibody. These unique features of Met@Man-MPs contribute to boost anti-PD-1 antibody therapy, improving anticancer efficacy and long-term memory immunity after combination treatment. Our results support Met@Man-MPs as a potential drug to improve tumor resistance to anti-PD-1 therapy. Durable response rate to anti-PD-1/PD-L1 therapy remains relatively low in patients with cancer. Here the authors show that metformin-loaded mannose-modified macrophage-derived microparticles reprogram the tumor immune microenvironment and improve responses to anti-PD-1 therapy.
Collapse
|
231
|
Altered microRNA Profiles of Extracellular Vesicles Secreted by Endometrial Cells from Women with Recurrent Implantation Failure. Reprod Sci 2021; 28:1945-1955. [PMID: 33432533 DOI: 10.1007/s43032-020-00440-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022]
Abstract
Recurrent implantation failure (RIF) is characterized by repeated embryo transfers without pregnancy. To date, the etiology of RIF remains poorly understood. Accumulating evidence indicates a beneficial role of endometrial extracellular vesicles (EVs) during the implantation by delivering signaling molecules to embryos, especially miRNAs. However, whether EVs secreted by RIF patients' endometria have a similar miRNA expression profile of endometrial EVs of fertile women has not been investigated. Therefore, in this study, we compared the miRNA expression profiles between the endometrial EVs of RIF patients (RIF-EVs) and fertile women (FER-EVs). Endometrial tissues from fifteen RIF patients and nine fertile women were collected and digested to cells for culture. Endometrial cells were modulated by estrogen and progesterone to mimic the secretory phase, and the conditioned medium was collected for EV isolation. EVs were determined by western blotting, nanoparticle tracking analysis, and transmission electronic microscopy (TEM). Three pairs of EV samples from two groups were used for miRNA sequencing, and twelve RIF-EV samples and six FER-EV samples were used for validation using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results showed that a total of 11 miRNAs were differently expressed in the RIF-EVs. Besides, four of the differently expressed miRNAs were validated using qRT-PCR. Target genes of the differently expressed miRNAs were predicted, and the functional analysis was performed. Besides, we proved that the most significantly different miRNA, 6131, inhibited the growth and invasion of HTR8/SVneo cells. Our study suggested that the altered miRNAs in the RIF-EVs might be involved in the pathogenesis of RIF.
Collapse
|
232
|
Berger A, Araújo-Filho I, Piffoux M, Nicolás-Boluda A, Grangier A, Boucenna I, Real CC, Marques FLN, de Paula Faria D, do Rego ACM, Broudin C, Gazeau F, Wilhelm C, Clément O, Cellier C, Buchpiguel CA, Rahmi G, Silva AKA. Local administration of stem cell-derived extracellular vesicles in a thermoresponsive hydrogel promotes a pro-healing effect in a rat model of colo-cutaneous post-surgical fistula. NANOSCALE 2021; 13:218-232. [PMID: 33326529 DOI: 10.1039/d0nr07349k] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Extracellular vesicles (EVs), especially from stem/stromal cells (SCs), represent a cell-free alternative in regenerative medicine holding promises to promote tissue healing while providing safety and logistic advantages in comparison to cellular counterparts. Herein, we hypothesize that SC EVs, administered locally in a thermoresponsive gel, is a therapeutic strategy for managing post-surgical colo-cutaneous fistulas. This disease is a neglected and challenging condition associated to low remission rates and high refractoriness. Herein, EVs from a murine SC line were produced by a high-yield scalable method in bioreactors. The post-surgical intestinal fistula model was induced via a surgical cecostomy communicating the cecum and the skin in Wistar rats. Animals were treated just after cecostomy with PBS, thermoresponsive Pluronic F-127 hydrogel alone or containing SC EVs. A PET-monitored biodistribution investigation of SC EVs labelled with 89Zr was performed. Fistula external orifice and output assessment, probe-based confocal laser endomicroscopy, MRI and histology were carried out for therapy follow-up. The relevance of percutaneous EV administration embedded in the hydrogel vehicle was indicated by the PET-biodistribution study. Local administration of SC EVs in the hydrogel reduced colo-cutaneous fistula diameter, output, fibrosis and inflammation while increasing the density of neo-vessels when compared to the PBS and gel groups. This multi-modal investigation pointed-out the therapeutic potential of SC EVs administered locally and in a thermoresponsive hydrogel for the management of challenging post-surgical colon fistulas in a minimally-invasive cell-free strategy.
Collapse
Affiliation(s)
- Arthur Berger
- Laboratoire Imagerie de l'Angiogénèse, Plateforme d'Imagerie du Petit Animal, PARCC, INSERM U970, Université de Paris, 75015, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Hyperactivation of P2X7 receptors as a culprit of COVID-19 neuropathology. Mol Psychiatry 2021; 26:1044-1059. [PMID: 33328588 PMCID: PMC7738776 DOI: 10.1038/s41380-020-00965-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022]
Abstract
Scientists and health professionals are exhaustively trying to contain the coronavirus disease 2019 (COVID-19) pandemic by elucidating viral invasion mechanisms, possible drugs to prevent viral infection/replication, and health cares to minimize individual exposure. Although neurological symptoms are being reported worldwide, neural acute and long-term consequences of SARS-CoV-2 are still unknown. COVID-19 complications are associated with exacerbated immunoinflammatory responses to SARS-CoV-2 invasion. In this scenario, pro-inflammatory factors are intensely released into the bloodstream, causing the so-called "cytokine storm". Both pro-inflammatory factors and viruses may cross the blood-brain barrier and enter the central nervous system, activating neuroinflammatory responses accompanied by hemorrhagic lesions and neuronal impairment, which are largely described processes in psychiatric disorders and neurodegenerative diseases. Therefore, SARS-CoV-2 infection could trigger and/or worse brain diseases. Moreover, patients with central nervous system disorders associated to neuroimmune activation (e.g. depression, Parkinson's and Alzheimer's disease) may present increased susceptibility to SARS-CoV-2 infection and/or achieve severe conditions. Elevated levels of extracellular ATP induced by SARS-CoV-2 infection may trigger hyperactivation of P2X7 receptors leading to NLRP3 inflammasome stimulation as a key mediator of neuroinvasion and consequent neuroinflammatory processes, as observed in psychiatric disorders and neurodegenerative diseases. In this context, P2X7 receptor antagonism could be a promising strategy to prevent or treat neurological complications in COVID-19 patients.
Collapse
|
234
|
Nazari M, Javandoost E, Talebi M, Movassaghpour A, Soleimani M. Platelet Microparticle Controversial Role in Cancer. Adv Pharm Bull 2021; 11:39-55. [PMID: 33747851 PMCID: PMC7961228 DOI: 10.34172/apb.2021.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are a group of micrometer-scale extracellular vesicles released by platelets upon activation that are responsible for the majority of microvesicles found in plasma. PMPs' physiological properties and functions have long been investigated by researchers. In this regard, a noticeable area of studies has been devoted to evaluating the potential roles and effects of PMPs on cancer progression. Clinical and experimental evidence conflictingly implicates supportive and suppressive functions for PMPs regarding cancer. Many of these functions could be deemed as a cornerstone for future considerations of PMPs usage in cancer targeted therapy. This review discusses what is currently known about PMPs and provides insights for new and possible research directions for further grasping the intricate interplay between PMPs and cancer.
Collapse
Affiliation(s)
- Mahnaz Nazari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Javandoost
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. Introduction
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
235
|
Li X, Jiang W, Gan Y, Zhou W. The Application of Exosomal MicroRNAs in the Treatment of Pancreatic Cancer and Its Research Progress. Pancreas 2021; 50:12-16. [PMID: 33370018 DOI: 10.1097/mpa.0000000000001713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
ABSTRACT Despite great progress in the treatment of pancreatic cancer under the efforts of researchers, the survival time of patients with pancreatic cancer is shorter than that of patients with other cancers. Thus, we have a great need for innovative therapeutic methods, including research and development of new drugs and innovation of administration methods. Exosomes are a type of extracellular vesicles wrapped by a lipid bilayer. Thanks to the low clearance ratio and strong specificity of exosomes in circulation, together with in-depth research on the surface protein of exosomes and a targeted modification method, there is a strong potential to apply exosomes in the transfer and even targeted delivery of chemotherapeutics, RNA, and natural products. Particularly, exosomes carrying microRNA show good application prospects in cancer therapy. This article is intended to summarize the progress of research relating to the treatment of pancreatic cancer via exosomal microRNAs.
Collapse
Affiliation(s)
- Xin Li
- From the The First Clinical Medical College, Lanzhou University
| | - Wenkai Jiang
- From the The First Clinical Medical College, Lanzhou University
| | - Yu Gan
- From the The First Clinical Medical College, Lanzhou University
| | | |
Collapse
|
236
|
Ramírez-Bajo MJ, Banon-Maneus E, Rovira J, Campistol JM, Diekmann F. Isolation of Extracellular Vesicles Derived from Mesenchymal Stromal Cells by Ultracentrifugation. Bio Protoc 2020; 10:e3860. [PMID: 33855106 DOI: 10.21769/bioprotoc.3860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/25/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membranous vesicles that differ on their biogenesis and release pathways, such as exosomes, microvesicles and apoptotic bodies. They are involved in cell-to-cell communication delivering signal molecules (proteins, nucleic acids, lipids, etc.) that can regulate different physiological processes, as well as the development and progression of several diseases. There are different methods and commercial kits to isolate EVs and depending on the methodology one could obtain EVs with different degrees of efficiency, purity and it can be more or less time-consuming. Then, the choice has to be according to the different advantages and disadvantages, and their use for downstream applications. Here, we describe the EVs isolation method from mesenchymal stromal cells by ultracentrifugation. This EVs isolation can be performed using common media and buffers, and only with the requirement of an analytical ultracentrifuge. Moreover, this method can be used to obtain large quantity of EVs with a good reproducibility for developing in vitro and in vivo experiments and studying their biological actions.
Collapse
Affiliation(s)
- María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi iSunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Elisenda Banon-Maneus
- Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi iSunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Josep M Campistol
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi iSunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain.,Departament de Nefrologia i Trasplantament Renal, ICNU, Hospital Clínic, Barcelona, Spain
| | - Fritz Diekmann
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi iSunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain.,Departament de Nefrologia i Trasplantament Renal, ICNU, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
237
|
Multifaceted Functions of Platelets in Cancer: From Tumorigenesis to Liquid Biopsy Tool and Drug Delivery System. Int J Mol Sci 2020; 21:ijms21249585. [PMID: 33339204 PMCID: PMC7765591 DOI: 10.3390/ijms21249585] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Platelets contribute to several types of cancer through plenty of mechanisms. Upon activation, platelets release many molecules, including growth and angiogenic factors, lipids, and extracellular vesicles, and activate numerous cell types, including vascular and immune cells, fibroblasts, and cancer cells. Hence, platelets are a crucial component of cell-cell communication. In particular, their interaction with cancer cells can enhance their malignancy and facilitate the invasion and colonization of distant organs. These findings suggest the use of antiplatelet agents to restrain cancer development and progression. Another peculiarity of platelets is their capability to uptake proteins and transcripts from the circulation. Thus, cancer-patient platelets show specific proteomic and transcriptomic expression patterns, a phenomenon called tumor-educated platelets (TEP). The transcriptomic/proteomic profile of platelets can provide information for the early detection of cancer and disease monitoring. Platelet ability to interact with tumor cells and transfer their molecular cargo has been exploited to design platelet-mediated drug delivery systems to enhance the efficacy and reduce toxicity often associated with traditional chemotherapy. Platelets are extraordinary cells with many functions whose exploitation will improve cancer diagnosis and treatment.
Collapse
|
238
|
Al Sharif S, Pinto DO, Mensah GA, Dehbandi F, Khatkar P, Kim Y, Branscome H, Kashanchi F. Extracellular Vesicles in HTLV-1 Communication: The Story of an Invisible Messenger. Viruses 2020; 12:E1422. [PMID: 33322043 PMCID: PMC7763366 DOI: 10.3390/v12121422] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) infects 5-10 million people worldwide and is the causative agent of adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) as well as other inflammatory diseases. A major concern is that the most majority of individuals with HTLV-1 are asymptomatic carriers and that there is limited global attention by health care officials, setting up potential conditions for increased viral spread. HTLV-1 transmission occurs primarily through sexual intercourse, blood transfusion, intravenous drug usage, and breast feeding. Currently, there is no cure for HTLV-1 infection and only limited treatment options exist, such as class I interferons (IFN) and Zidovudine (AZT), with poor prognosis. Recently, small membrane-bound structures, known as extracellular vesicles (EVs), have received increased attention due to their potential to carry viral cargo (RNA and proteins) in multiple pathogenic infections (i.e., human immunodeficiency virus type I (HIV-1), Zika virus, and HTLV-1). In the case of HTLV-1, EVs isolated from the peripheral blood and cerebral spinal fluid (CSF) of HAM/TSP patients contained the viral transactivator protein Tax. Additionally, EVs derived from HTLV-1-infected cells (HTLV-1 EVs) promote functional effects such as cell aggregation which enhance viral spread. In this review, we present current knowledge surrounding EVs and their potential role as immune-modulating agents in cancer and other infectious diseases such as HTLV-1 and HIV-1. We discuss various features of EVs that make them prime targets for possible vehicles of future diagnostics and therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fatah Kashanchi
- Laboratory of Molecular Virology, George Mason University, Manassas, VA 20110, USA; (S.A.S.); (D.O.P.); (G.A.M.); (F.D.); (P.K.); (Y.K.); (H.B.)
| |
Collapse
|
239
|
Characterization and Transcriptome Analysis of Exosomal and Nonexosomal RNAs in Bovine Adipocytes. Int J Mol Sci 2020; 21:ijms21239313. [PMID: 33297338 PMCID: PMC7730049 DOI: 10.3390/ijms21239313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022] Open
Abstract
Exosomes are endosome-derived extracellular vesicles that allow intercellular communication. However, the biological significance of adipocyte exosomal RNAs remains unclear. To determine the role of RNAs from bovine adipocytes and exosomes in bovine adipogenesis, exosomal and nonexosomal RNAs were extracted from three bovine primary white adipocyte samples and then profiles were generated using DNBSEQ/BGISEQ-500 technology. The RNAome of adipocytes consisted of 12,082 mRNAs, 8589 lncRNAs, and 378 miRNAs for a higher complexity that that detected in exosomes, with 1083 mRNAs, 105 lncRNAs, and 48 miRNAs. Exosomal miRNA-mRNA and lncRNA–miRNA–mRNA networks were constructed and enrichment analysis was performed to predict functional roles and regulatory mechanisms. Our study provides the first characterization of RNAs from bovine adipocyte and exosomes. The findings reveal that some RNAs are specifically packaged in adipocyte-derived exosomes, potentially enabling crosstalk between adipocytes and/or other cells that is mediated by exosomes. Our results greatly expand our understanding of exosomal RNAs from bovine adipocytes, and provide a reference for future functional investigations of adipocyte exosomal RNAs under normal physiological conditions.
Collapse
|
240
|
Ratajczak MZ, Ratajczak J. Extracellular microvesicles/exosomes: discovery, disbelief, acceptance, and the future? Leukemia 2020; 34:3126-3135. [PMID: 32929129 PMCID: PMC7685969 DOI: 10.1038/s41375-020-01041-z] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 08/18/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
There are concepts in science that need time to overcome initial disbelief before finally arriving at the moment when they are embraced by the research community. One of these concepts is the biological meaning of the small, spheroidal vesicles released from cells, which are described in the literature as microparticles, microvesicles, or exosomes. In the beginning, this research was difficult, as it was hard to distinguish these small vesicles from cell debris or apoptotic bodies. However, they may represent the first language of cell-cell communication, which existed before a more specific intercellular cross-talk between ligands and receptors emerged during evolution. In this review article, we will use the term "extracellular microvesicles" (ExMVs) to refer to these small spheroidal blebs of different sizes surrounded by a lipid layer of membrane. We have accepted an invitation from the Editor-in-Chief to write this review in observance of the 20th anniversary of the 2001 ASH Meeting when our team demonstrated that, by horizontal transfer of several bioactive molecules, including mRNA species and proteins, ExMVs harvested from embryonic stem cells could modify hematopoietic stem/progenitor cells and expand them ex vivo. Interestingly, the result that moved ExMV research forward was published first in 2005 in Leukemia, having been previously rejected by other major scientific journals out of simple disbelief. Therefore, the best judge of a new concept is the passage of time, although the speed of its adoption is aided by perseverance and confidence in one's own data. In this perspective article, we will provide a brief update on the current status of, hopes for, and likely future of ExMV research as well as therapeutic and diagnostic applications, with a special emphasis on hematopoiesis.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland.
| | - Janina Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
241
|
Weiss C, Kornicka-Grabowska K, Mularczyk M, Siwinska N, Marycz K. Extracellular Microvesicles (MV's) Isolated from 5-Azacytidine-and-Resveratrol-Treated Cells Improve Viability and Ameliorate Endoplasmic Reticulum Stress in Metabolic Syndrome Derived Mesenchymal Stem Cells. Stem Cell Rev Rep 2020; 16:1343-1355. [PMID: 32880856 PMCID: PMC7667134 DOI: 10.1007/s12015-020-10035-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs), a spherical membrane fragments including exosomes, are released from several cell types, including mesenchymal stromal cells (MSCs), constitutively or under stimulation. As MVs cargo include DNA, RNA, miRNA, lipids and proteins their have gain special attention in the field of regenerative medicine. Depending on the type of transferred molecules, MVs may exert wide range of biological effects in recipient cells including pro-inflammatory and anti-apoptotic action. In presented paper, we isolated MVs form adipose derived mesenchymal stem cells (ASC) which underwent stimulation with 5-azacytydine and resveratrol (AZA/RES) in order to improve their therapeutic potential. Then, isolated MVs were applied to ASC with impaired cytophysiological properties, isolated from equine metabolic syndrome diagnosed animals. Using RT-PCR, immunofluorescence, ELISA, confocal microscopy and western blot, we have evaluated the effects of MVs on recipient cells. We have found, that MVs derived from AZA/RES treated ASC ameliorates apoptosis, senescence and endoplasmic reticulum (ER) stress in deteriorated cells, restoring their proper functions. The work indicates, that cells treated with AZA/RES through their paracrine action can rejuvenate recipient cells. However, further research needs to be performed in order to fully understand the molecular mechanisms of these bioactive factors action. Graphical Abstract Graphical abstract of presented study.
Collapse
Affiliation(s)
- C Weiss
- PferdePraxis Dr. Med. Vet. Daniel Weiss, Postmatte 14, CH-8807, Freienbach, Switzerland
| | - K Kornicka-Grabowska
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B street, A7 building, 50-375, Wroclaw, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland
| | - M Mularczyk
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B street, A7 building, 50-375, Wroclaw, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland
| | - N Siwinska
- Department of Internal Medicine and Clinic of Diseases of Horses, Dogs and Cats, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Pl. Grunwaldzki 47, 50-366, Wroclaw, Poland
| | - K Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B street, A7 building, 50-375, Wroclaw, Poland.
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany.
| |
Collapse
|
242
|
Exosomes and Extracellular Vesicles as Emerging Theranostic Platforms in Cancer Research. Cells 2020; 9:cells9122569. [PMID: 33271820 PMCID: PMC7761021 DOI: 10.3390/cells9122569] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/19/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Exosomes are endosome-derived nanovesicles produced by healthy as well as diseased cells. Their proteic, lipidic and nucleic acid composition is related to the cell of origin, and by vehiculating bioactive molecules they are involved in cell-to-cell signaling, both in healthy and pathologic conditions. Being nano-sized, non-toxic, biocompatible, scarcely immunogenic, and possessing targeting ability and organotropism, exosomes have been proposed as nanocarriers for their potential application in diagnosis and therapy. Among the different techniques exploited for exosome isolation, the sequential ultracentrifugation/ultrafiltration method seems to be the gold standard; alternatively, commercially available kits for exosome selective precipitation from cell culture media are frequently employed. To load a drug or a detectable agent into exosomes, endogenous or exogenous loading approaches have been developed, while surface engineering procedures, such as click chemistry, hydrophobic insertion and exosome display technology, allow for obtaining actively targeted exosomes. This review reports on diagnostic or theranostic platforms based on exosomes or exosome-mimetic vesicles, highlighting the diverse preparation, loading and surface modification methods applied, and the results achieved so far.
Collapse
|
243
|
Shi J, Zhang Y, Yao B, Sun P, Hao Y, Piao H, Zhao X. Role of Exosomes in the Progression, Diagnosis, and Treatment of Gliomas. Med Sci Monit 2020; 26:e924023. [PMID: 33245712 PMCID: PMC7706139 DOI: 10.12659/msm.924023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gliomas are the most common primary malignant brain tumors associated with a low survival rate. Even after surgery, radiotherapy, and chemotherapy, gliomas still have a poor prognosis. Extracellular vesicles are a heterogeneous group of cell-derived membranous structures. Exosomes are a type of extracellular vesicles, their size ranges from 30 nm to 100 nm. Recent studies have proved that glioma cells could release numerous exosomes; therefore, exosomes have gained increasing attention in glioma-related research. Recent studies have confirmed the importance of extracellular vesicles, particularly exosomes, in the development of brain tumors, including gliomas. Exosomes mediate intercellular communication in the tumor microenvironment by transporting biomolecules (proteins, lipids, deoxyribonucleic acid, and ribonucleic acid); thereby playing a prominent role in tumor proliferation, differentiation, metastasis, and resistance to chemotherapy or radiation. Given their nanoscale size, exosomes can traverse the blood-brain barrier and promote tumor progression by modifying the tumor microenvironment. Based on their structural and functional characteristics, exosomes are demonstrating their value not only as diagnostic and prognostic markers, but also as tools in therapies specifically targeting glioma cells. Therefore, exosomes are a promising therapeutic target for the diagnosis, prognosis, and treatment of malignant gliomas. More research will be needed before exosomes can be used in clinical applications. Here, we describe the exosomes, their morphology, and their roles in the diagnosis and progression of gliomas. In addition, we discuss the potential of exosomes as a therapeutic target/drug delivery system for patients with gliomas.
Collapse
Affiliation(s)
- Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Ye Zhang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Bing Yao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Peixin Sun
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yuanyuan Hao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xi Zhao
- Department of Anesthesia, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
244
|
Automated enumeration and phenotypic characterization of CTCs and tdEVs in patients with metastatic castration resistant prostate cancer. Prostate Cancer Prostatic Dis 2020; 24:499-506. [PMID: 33230201 PMCID: PMC8134056 DOI: 10.1038/s41391-020-00304-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
Background Although most patients with metastatic castration-resistant prostate cancer (mCRPC) initially benefit from treatment with androgen receptor signaling inhibitors (ARSi), resistance inevitably occurs. Hence, we investigated the prognostic value of automated circulating tumor cell (CTC) and tumor-derived extracellular vesicle (tdEV) enumeration and their dynamics, in patients with mCRPC in the context of the initiation of treatment with ARSi. Furthermore, we hypothesize that CTC phenotypic heterogeneity might serve as a measurable biomarker under these circumstances. Methods Using an image analysis tool, we reanalyzed all CellSearch images previously acquired in the context of a prospective, multicenter clinical study for patients with mCRPC (n = 170) starting a new line of ARSi, for CTC and tdEV detection and enumeration. CTC (n = 19 129) phenotypic diversity was quantified by the Shannon index (SI). Progression-free survival (PFS) and overall survival (OS) were compared between groups of patients stratified according to CTC, tdEV, and SI levels. Results Automated CTC enumeration provided similar clinical prognostication compared with operator-based counts. Patients demonstrating high CTC phenotypic heterogeneity before therapy had a shorter median PFS (4.82 vs. 8.49 months, HR 1.79; P = 0.03) and OS (12.6 months vs. not reached, HR 2.32; P = 0.03), compared to patients with low diversity, irrespective of CTC level. Multivariable analysis showed how the prognostic value of the baseline SI was lost by pretreatment chemotherapy status, CTC counts, and PSA levels. Conclusions Automated CTC counts are a reliable substitute for reviewer-based enumeration, as they are equally informative for prognosis assessment in patients with mCRPC. Beyond enumeration, we demonstrated the added value of studying CTC phenotypic diversity for patient prognostication, warranting future investigation.
Collapse
|
245
|
Bazzoni R, Takam Kamga P, Tanasi I, Krampera M. Extracellular Vesicle-Dependent Communication Between Mesenchymal Stromal Cells and Immune Effector Cells. Front Cell Dev Biol 2020; 8:596079. [PMID: 33240892 PMCID: PMC7677193 DOI: 10.3389/fcell.2020.596079] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells residing in the stromal tissues of the body and capable of promoting tissue repair and attenuating inflammatory processes through their immunomodulatory properties. Preclinical and clinical observations revealed that not only direct intercellular communication mediates MSC properties; in fact, a pivotal role is also played by the release of soluble and bioactive factors, such as cytokines, growth factor and extracellular vesicles (EVs). EVs are membrane-coated vesicles containing a large variety of bioactive molecules, including lipids, proteins, and nucleic acids, such as RNA. EVs release their contents into target cells, thus influencing cell fate through the control of intracellular processes. In addition, MSC-derived EVs can mediate modulatory effects toward different effector cells belonging to both innate and adaptive immunity. In this review, we will discuss the literature data concerning MSC-derived EVs, including the current standardized methods for their isolation and characterization, the mechanisms supporting their immunoregulatory properties, and their potential clinical application as alternative to MSC-based therapy for inflammatory reactions, such as graft-versus-host disease (GvHD).
Collapse
Affiliation(s)
- Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
- EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Ilaria Tanasi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
246
|
Chen T, Moscvin M, Bianchi G. Exosomes in the Pathogenesis and Treatment of Multiple Myeloma in the Context of the Bone Marrow Microenvironment. Front Oncol 2020; 10:608815. [PMID: 33251153 PMCID: PMC7674795 DOI: 10.3389/fonc.2020.608815] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/12/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematological malignancy, is an incurable cancer of plasma cells. MM cells diffusely involves the bone marrow (BM) and establish a close interaction with the BM niche that in turn supports MM survival, proliferation, dissemination and drug resistance. In spite of remarkable progress in understanding MM biology and developing drugs targeting MM in the context of the BM niche, acquisition of multi-class drug resistance is almost universally inevitable. Exosomes are small, secreted vesicles that have been shown to mediate bidirectional transfer of proteins, lipids, and nucleic acids between BM microenvironment and MM, supporting MM pathogenesis by promoting angiogenesis, osteolysis, and drug resistance. Exosome content has been shown to differ between MM patients and healthy donors and could potentially serve as both cancer biomarker and target for novel therapies. Furthermore, the natural nanostructure and modifiable surface properties of exosomes make them good candidates for drug delivery or novel immunomodulatory therapy. In this review we will discuss the current knowledge regarding exosome's role in MM pathogenesis and its potential role as a novel biomarker and therapeutic tool in MM.
Collapse
Affiliation(s)
- Tianzeng Chen
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Maria Moscvin
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Giada Bianchi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| |
Collapse
|
247
|
Strohbach A, Böhm A, Mahajan-Thakur S, Wirtz C, Wetzel H, Busch MC, Felix SB, Rauch BH, Busch R. Platelet apelin receptor expression is reduced in patients with acute myocardial infarction. Vascul Pharmacol 2020; 136:106808. [PMID: 33130016 DOI: 10.1016/j.vph.2020.106808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/06/2020] [Accepted: 10/26/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND The G-protein-coupled apelin receptor and its apelin ligand are an emerging regulatory system of the vascular homeostasis. To date, the implications of the apelin/apelin receptor system in athero-thrombosis are not completely clarified yet. This study determines the expression of the apelin receptor on human platelets, the effect of different apelin isoforms on platelet aggregation and the potential role of the apelin/apelin receptor system in acute myocardial infarction. METHODS We applied immunofluorescence staining, Western Blot analysis, aggregometry, and flow cytometry to elucidate the role of the apelin receptor in activated platelets. Furthermore, in an observational pilot study, we assessed platelet apelin recpetor expression and apelin-17 plasma levels in patients with acute myocardial infarction (AMI, n = 27). RESULTS Immunofluorescence staining indicates that the apelin receptor is located at the cell membrane in resting platelets and diminishes upon activation with a selective thrombin receptor-activating peptide (AP1, 3 to 100 μM). Western Blot analyses of AP1-activated platelets and their supernatants suggest that the apelin receptor is not predominantly internalized but is released from activated platelets. The isoform apelin-17 attenuated AP-1-induced platelet activation in-vitro, presumably via a NO-dependent mechanism. Furthermore, platelet apelin receptor expression was significantly reduced in patients with AMI (n = 27) compared to age-matched controls (n = 14; p < 0.05) and inversely correlated with troponin I plasma levels (r = -0.46; p = 0.03). Besides that, circulating apelin-17 was significantly reduced in MI patients compared to the control group. CONCLUSION Taken together, our data support a crucial role of the platelet apelinergic system assuming an antithrombotic effect and therefore holding a potential diagnostic and therapeutic impact.
Collapse
Affiliation(s)
- Anne Strohbach
- Department of Internal Medicine B (Cardiology), University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner site, Greifswald, Germany.
| | - Andreas Böhm
- DZHK (German Centre for Cardiovascular Research), Partner site, Greifswald, Germany; Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), Ernst-Moritz-Arndt University, Felix-Hausdorff-Strasse 3, 17487 Greifswald, Germany
| | - Shailaja Mahajan-Thakur
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), Ernst-Moritz-Arndt University, Felix-Hausdorff-Strasse 3, 17487 Greifswald, Germany
| | - Christopher Wirtz
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), Ernst-Moritz-Arndt University, Felix-Hausdorff-Strasse 3, 17487 Greifswald, Germany
| | - Hanno Wetzel
- Department of Internal Medicine B (Cardiology), University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany
| | - Mathias C Busch
- Department of Internal Medicine B (Cardiology), University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner site, Greifswald, Germany
| | - Stephan B Felix
- Department of Internal Medicine B (Cardiology), University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner site, Greifswald, Germany
| | - Bernhard H Rauch
- DZHK (German Centre for Cardiovascular Research), Partner site, Greifswald, Germany; Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), Ernst-Moritz-Arndt University, Felix-Hausdorff-Strasse 3, 17487 Greifswald, Germany
| | - Raila Busch
- Department of Internal Medicine B (Cardiology), University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), Partner site, Greifswald, Germany.
| |
Collapse
|
248
|
Qin B, Zhang Q, Hu XM, Mi TY, Yu HY, Liu SS, Zhang B, Tang M, Huang JF, Xiong K. How does temperature play a role in the storage of extracellular vesicles? J Cell Physiol 2020; 235:7663-7680. [PMID: 32324279 DOI: 10.1002/jcp.29700] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) contain specific proteins, lipids, and nucleic acids that can be passed to other cells as signal molecules to alter their function. However, there are many problems and challenges in the conversion and clinical application of EVs. Storage and protection of EVs is one of the issues that need further research. To adapt to potential clinical applications, this type of problem must be solved. This review summarizes the storage practices of EVs in recent years, and explains the impact of temperature on the quality and stability of EVs during storage based on current research, and explains the potential mechanisms involved in this effect as much as possible.
Collapse
Affiliation(s)
- Bo Qin
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qi Zhang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xi-Min Hu
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Tuo-Yang Mi
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hai-Yang Yu
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shen-Shen Liu
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Bin Zhang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Mu Tang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ju-Fang Huang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China
| | - Kun Xiong
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China
| |
Collapse
|
249
|
Ma J, Zhang H, Tang K, Huang B. Tumor-derived microparticles in tumor immunology and immunotherapy. Eur J Immunol 2020; 50:1653-1662. [PMID: 32976623 PMCID: PMC7702100 DOI: 10.1002/eji.202048548] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/11/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Microvesicles or microparticles, a type of cytoplasm membrane-derived extracellular vesicles, can be released by cancer cells or normal cell types. Alteration of F-actin cytoskeleton by various signals may lead to the cytoplasm membrane encapsulating cellular contents to form microparticles, which contain various messenger molecules, including enzymes, RNAs and even DNA fragments, and are released to extracellular space. The release of microparticles by tumor cells (T-MPs) is a very common event in tumor microenvironments. As a result, T-MPs not only influence tumor cell biology but also profoundly forge tumor immunology. Moreover, T-MPs can act as a natural vehicle that delivers therapeutic drugs to tumor cells and immune cells, thus, remodeling tumor microenvironments and resetting antitumor immune responses, thus, conferring T-MPs a potential role in tumor immunotherapies and tumor vaccines. In this review, we focus on the double-edged sword role of T-MPs in tumor immunology, specifically in TAMs and DCs, and emphasize the application of drug-packaging T-MPs in cancer patients. We aim to provide a new angle to understand immuno-oncology and new strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, P. R. China
| | - Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, P. R. China
| | - Ke Tang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, P. R. China
| | - Bo Huang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, P. R. China.,Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, P. R. China.,Clinical Immunology Center, CAMS, Beijing, P. R. China
| |
Collapse
|
250
|
Role of extracellular vesicles in tumour microenvironment. Cell Commun Signal 2020; 18:163. [PMID: 33081785 PMCID: PMC7574205 DOI: 10.1186/s12964-020-00643-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
In recent years, it has been demonstrated that extracellular vesicles (EVs) can be released by almost all cell types, and detected in most body fluids. In the tumour microenvironment (TME), EVs serve as a transport medium for lipids, proteins, and nucleic acids. EVs participate in various steps involved in the development and progression of malignant tumours by initiating or suppressing various signalling pathways in recipient cells. Although tumour-derived EVs (T-EVs) are known for orchestrating tumour progression via systemic pathways, EVs from non-malignant cells (nmEVs) also contribute substantially to malignant tumour development. Tumour cells and non-malignant cells typically communicate with each other, both determining the progress of the disease. In this review, we summarise the features of both T-EVs and nmEVs, tumour progression, metastasis, and EV-mediated chemoresistance in the TME. The physiological and pathological effects involved include but are not limited to angiogenesis, epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) remodelling, and immune escape. We discuss potential future directions of the clinical application of EVs, including diagnosis (as non-invasive biomarkers via liquid biopsy) and therapeutic treatment. This may include disrupting EV biogenesis and function, thus utilising the features of EVs to repurpose them as a therapeutic tool in immunotherapy and drug delivery systems. We also discuss the overall findings of current studies, identify some outstanding issues requiring resolution, and propose some potential directions for future research. Video abstract.
Collapse
|