201
|
Extracellular Vesicles and Matrix Remodeling Enzymes: The Emerging Roles in Extracellular Matrix Remodeling, Progression of Diseases and Tissue Repair. Cells 2018; 7:cells7100167. [PMID: 30322133 PMCID: PMC6210724 DOI: 10.3390/cells7100167] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 09/17/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane enclosed micro- and nano-sized vesicles that are secreted from almost every species, ranging from prokaryotes to eukaryotes, and from almost every cell type studied so far. EVs contain repertoire of bioactive molecules such as proteins (including enzymes and transcriptional factors), lipids, carbohydrates and nucleic acids including DNA, coding and non-coding RNAs. The secreted EVs are taken up by neighboring cells where they release their content in recipient cells, or can sail through body fluids to reach distant organs. Since EVs transport bioactive cargo between cells, they have emerged as novel mediators of extra- and intercellular activities in local microenvironment and inter-organ communications distantly. Herein, we review the activities of EV-associated matrix-remodeling enzymes such as matrix metalloproteinases, heparanases, hyaluronidases, aggrecanases, and their regulators such as extracellular matrix metalloproteinase inducers and tissue inhibitors of metalloproteinases as novel means of matrix remodeling in physiological and pathological conditions. We discuss how such EVs act as novel mediators of extracellular matrix degradation to prepare a permissive environment for various pathological conditions such as cancer, cardiovascular diseases, arthritis and metabolic diseases. Additionally, the roles of EV-mediated matrix remodeling in tissue repair and their potential applications as organ therapies have been reviewed. Collectively, this knowledge could benefit the development of new approaches for tissue engineering.
Collapse
|
202
|
Schlesinger M. Role of platelets and platelet receptors in cancer metastasis. J Hematol Oncol 2018; 11:125. [PMID: 30305116 PMCID: PMC6180572 DOI: 10.1186/s13045-018-0669-2] [Citation(s) in RCA: 366] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/25/2018] [Indexed: 01/15/2023] Open
Abstract
The interaction of tumor cells with platelets is a prerequisite for successful hematogenous metastatic dissemination. Upon tumor cell arrival in the blood, tumor cells immediately activate platelets to form a permissive microenvironment. Platelets protect tumor cells from shear forces and assault of NK cells, recruit myeloid cells by secretion of chemokines, and mediate an arrest of the tumor cell platelet embolus at the vascular wall. Subsequently, platelet-derived growth factors confer a mesenchymal-like phenotype to tumor cells and open the capillary endothelium to expedite extravasation in distant organs. Finally, platelet-secreted growth factors stimulate tumor cell proliferation to micrometastatic foci. This review provides a synopsis on the current literature on platelet-mediated effects in cancer metastasis and particularly focuses on platelet adhesion receptors and their role in metastasis. Immunoreceptor tyrosine-based activation motif (ITAM) and hemi ITAM (hemITAM) comprising receptors, especially, glycoprotein VI (GPVI), FcγRIIa, and C-type lectin-like-2 receptor (CLEC-2) are turned in the spotlight since several new mechanisms and contributions to metastasis have been attributed to this family of platelet receptors in the last years.
Collapse
|
203
|
Wang W, Luo J, Wang S. Recent Progress in Isolation and Detection of Extracellular Vesicles for Cancer Diagnostics. Adv Healthc Mater 2018; 7:e1800484. [PMID: 30009550 DOI: 10.1002/adhm.201800484] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/20/2018] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are emerging as one of the many new and promising biomarkers for liquid biopsy of cancer due to their loading capability of some specific proteins and nucleic acids that are closely associated with cancer states. As such, the isolation and detection of cancer-derived EVs offer important information in noninvasive diagnosis of early-stage cancer and real-time monitoring of cancer development. In light of the importance of EVs, over the last decade, researchers have made remarkable innovations to advance the development of EV isolation and detection methods by taking advantage of microfluidics, biomolecule probes, nanomaterials, surface plasmon, optics, and so on. This review introduces the basic properties of EVs and common cancer-derived EV ingredients, and provides a comprehensive overview of EV isolation and detection strategies, with emphasis on liquid biopsies of EVs for cancer diagnostics.
Collapse
Affiliation(s)
- Wenshuo Wang
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science; Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Jing Luo
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science; Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science; Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| |
Collapse
|
204
|
Gulei D, Petrut B, Tigu AB, Onaciu A, Fischer-Fodor E, Atanasov AG, Ionescu C, Berindan-Neagoe I. Exosomes at a glance - common nominators for cancer hallmarks and novel diagnosis tools. Crit Rev Biochem Mol Biol 2018; 53:564-577. [PMID: 30247075 DOI: 10.1080/10409238.2018.1508276] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer represents a heterogeneous disease with multiple levels of regulation and a dynamic environment that sustains the evolution of the malignant mass. This dynamic is in part sustained by a class of extracellular vesicles termed exosomes that are able to imprint the pathological state by incorporating differential cargos in order to facilitate cell-to-cell communication. Exosomes are stable within the extracellular medium and function as shuttles secreted by healthy or pathological cells, being further taken by the accepting cell with direct effects on its phenotype. The exosomal trafficking is deeply involved in multiple levels of cancer development with roles in all cancer hallmarks. Nowadays, studies are constantly exploring the ability of exosomes to sustain the malignant progression in order to attack this pathological trafficking and impair the ability of the tumor mass to expand within the organisms. As important, the circulatory characteristics of exosomes represent a steady advantage regarding the possibility of using them as minimally invasive diagnosis tools, where cancer patients' present modified exosomal profiles compared to the healthy ones. This last characteristic, as novel diagnosis tools, has the advantage of a possible rapid transition within the clinic, compared to the studies that evaluate the therapeutic meaning.
Collapse
Affiliation(s)
- Diana Gulei
- a MEDFUTURE - Research Center for Advanced Medicine "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Bogdan Petrut
- b Department of Urology , The Oncology Institute "Prof Dr. Ion Chiricuta" , Cluj-Napoca , Romania
| | - Adrian Bogdan Tigu
- a MEDFUTURE - Research Center for Advanced Medicine "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Anca Onaciu
- a MEDFUTURE - Research Center for Advanced Medicine "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Eva Fischer-Fodor
- a MEDFUTURE - Research Center for Advanced Medicine "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania.,c Tumor Biology Department , Ion Chiricuta Oncology Institute , Cluj-Napoca , Romania
| | - Atanas G Atanasov
- d Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzȩbiec, Magdalenka , Poland.,e Department of Pharmacognosy , University of Vienna , Vienna , Austria
| | - Calin Ionescu
- f 5th Surgical Department , Municipal Hospital , Cluj-Napoca , Romania.,g "Iuliu Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Ioana Berindan-Neagoe
- a MEDFUTURE - Research Center for Advanced Medicine "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania.,h Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania.,i Department of Functional Genomics and Experimental Pathology , "Prof. Dr. Ion Chiricuta" Oncology Institute , Cluj-Napoca , Romania
| |
Collapse
|
205
|
Jurj A, Pop L, Petrushev B, Pasca S, Dima D, Frinc I, Deak D, Desmirean M, Trifa A, Fetica B, Gafencu G, Selicean S, Moisoiu V, Micu WT, Berce C, Sacu A, Moldovan A, Colita A, Bumbea H, Tanase A, Dascalescu A, Zdrenghea M, Stiufiuc R, Leopold N, Tetean R, Burzo E, Tomuleasa C, Berindan-Neagoe I. Exosome-carried microRNA-based signature as a cellular trigger for the evolution of chronic lymphocytic leukemia into Richter syndrome. Crit Rev Clin Lab Sci 2018; 55:501-515. [PMID: 30238808 DOI: 10.1080/10408363.2018.1499707] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Even if considered a cumulative and not a proliferative CD5+ B-cell neoplasm, chronic lymphocytic leukemia (CLL) has a proliferation rate higher than that recognized earlier, especially in the lymphoid tissues. Some patients with CLL develop a clinical syndrome entitled Richter syndrome (RS). Understanding CLL genetics and epigenetics may help to elucidate the molecular basics of the clinical heterogeneity of this type of malignancy. In the present project we aimed to identify a microRNA species that can predict the evolution of therapy-resistant CLL towards RS. In the first phase of our study, microRNA-19b was identified as a possible target, and in the second phase, we transfected three different CLL cell lines with microRNA-19b mimic and inhibitor and assessed the potential role on leukemia cells in vitro. The mechanism by which miR-19b acts were identified as the upregulation of Ki67 and downregulation of p53. This was further supported through RT-PCR and western blotting on CLL cell lines, as well as by next generation sequencing on two patients diagnosed with CLL that evolved into RS.
Collapse
Affiliation(s)
- Ancuta Jurj
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Laura Pop
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Bobe Petrushev
- b Department of Pathology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Sergiu Pasca
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Delia Dima
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Ioana Frinc
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Dalma Deak
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Minodora Desmirean
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Adrian Trifa
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Bogdan Fetica
- b Department of Pathology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Grigore Gafencu
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Sonia Selicean
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Vlad Moisoiu
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Wilhelm-Thomas Micu
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Cristian Berce
- e Center for Experimental Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Alexandra Sacu
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Alin Moldovan
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania.,f Department of Hematology , Carol Davilla University of Medicine and Pharmacy , Bucharest , Romania
| | - Andrei Colita
- g Department of Hematology , Coltea Hospital , Bucharest , Romania
| | - Horia Bumbea
- f Department of Hematology , Carol Davilla University of Medicine and Pharmacy , Bucharest , Romania.,h Department of Hematology , University Hospital , Bucharest , Romania
| | - Alina Tanase
- h Department of Hematology , University Hospital , Bucharest , Romania.,i Department of Hematology , Fundeni Clinical Hospital , Bucharest , Romania
| | - Angela Dascalescu
- j Department of Hematology , Grigore T. Popa University of Medicine and Pharmacy , Iasi , Romania.,k Department of Hematology , Regional Institute of Oncology , Iasi , Romania
| | - Mihnea Zdrenghea
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Rares Stiufiuc
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Nicolae Leopold
- l Department of Physics , Babes Bolyai University , Cluj Napoca , Romania
| | - Romulus Tetean
- l Department of Physics , Babes Bolyai University , Cluj Napoca , Romania
| | - Emil Burzo
- l Department of Physics , Babes Bolyai University , Cluj Napoca , Romania.,m Romanian Academy , Romania
| | - Ciprian Tomuleasa
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania.,c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Ioana Berindan-Neagoe
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| |
Collapse
|
206
|
Naderi-Meshkin H, Ahmadiankia N. Cancer metastasis versus stem cell homing: Role of platelets. J Cell Physiol 2018; 233:9167-9178. [PMID: 30105746 DOI: 10.1002/jcp.26937] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
One of the major obstacles in achieving a successful stem cell therapy is insufficient homing of transplanted cells. To overcome this obstacle, understanding the underlying mechanisms of stem cell homing is of obvious importance. Central to this review is the concept that cancer metastasis can be viewed as a role model to build up a comprehensive concept of stem cell homing. In this novel perspective, the prosurvival choices of the cancerous cells in the bloodstream, their arrest, extravasation, and proliferation at the secondary site can be exploited in favor of targeted stem cell homing. To date, tumor cells have been found to employ a wide variety of strategies to promote metastasis. One of these strategies is through their ability to activate platelets and subsequently activated platelets serve cancer cell survival and metastasis. Accordingly, in the first part of this review the roles of platelets in cancer metastasis as well as stem cell homing are discussed. Next, we provide some lessons learned from cancer metastasis in favor of developing strategies for improvement of stem cell homing with emphasis on the role of platelets. Based on direct or indirect evidence from metastasis, strategies such as manipulation of stem cells to enhance interaction with platelets, preconditioning-pretreatment of stem cells with platelets in vitro, and coinjection of both stem cells and platelets are proposed to improve stem cell homing.
Collapse
Affiliation(s)
- Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Naghmeh Ahmadiankia
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
207
|
Hafiane A, Daskalopoulou SS. Extracellular vesicles characteristics and emerging roles in atherosclerotic cardiovascular disease. Metabolism 2018; 85:213-222. [PMID: 29727628 DOI: 10.1016/j.metabol.2018.04.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/06/2018] [Accepted: 04/25/2018] [Indexed: 01/08/2023]
Abstract
The term extracellular vesicles (EVs) describes membrane vesicles released into the extracellular space by most cell types. EVs have been recognized to play an important role in cell-to-cell communication. They are known to contain various bioactive molecules, including proteins, lipids, and nucleic acids. Although the nomenclature of EVs is not entirely standardized, they are considered to include exosomes, microparticles or microvesicles and apoptotic bodies. EVs are believed to play important roles in a wide range of biological processes. Although the pathogenic roles of EVs are largely documented, their protective roles are not as well established. Cardiovascular disease represents one of the most relevant and rapidly growing areas of the EV research. Circulating EVs released from platelets, erythrocytes, leukocytes, and endothelial cells may contain potentially valuable biological information for biomarker development in cardiovascular disease and could serve as a vehicle for therapeutic use. Herein, we provide an overview of the current knowledge in EV in cardiovascular disease, including a discussion on challenges in EV research, EV properties in various cell types, and their importance in atherosclerotic disease.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Stella S Daskalopoulou
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
208
|
Abstract
Platelet-derived microvesicles (pMVs) are small, heterogeneous vesicles released from platelet membranes as a result of activation. These microvesicles possess a wide range of properties, including prothrombotic, proatherogenic, proinflammatory, immunomodulatory, and even anticoagulant activity. The elevated release of these microvesicles has been observed in various metabolic, inflammatory, thrombotic, and vascular diseases, including ischemic heart disease, stroke, hypertension, diabetes, and connective tissue disease. Modulation of both pMV generation and the expression of their surface molecules may have beneficial clinical implications and could become a novel therapeutic target. However, mechanisms by which pharmacological agents can modify pMV formation are elusive. The purpose of this review is to discuss the effects of drugs routinely used in primary and secondary prevention of vascular disease on the release of pMV and expression of their surface procoagulant and proinflammatory molecules.
Collapse
Affiliation(s)
- Justyna Rosińska
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Maria Łukasik
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| |
Collapse
|
209
|
Best MG, Wesseling P, Wurdinger T. Tumor-Educated Platelets as a Noninvasive Biomarker Source for Cancer Detection and Progression Monitoring. Cancer Res 2018; 78:3407-3412. [PMID: 29921699 DOI: 10.1158/0008-5472.can-18-0887] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/17/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022]
Abstract
Liquid biopsies represent a potential revolution in cancer diagnostics as a noninvasive method for detecting and monitoring diseases, complementary to or even replacing current tissue biopsy approaches. Several blood-based biosources and biomolecules, such as cell-free DNA and RNA, proteins, circulating tumor cells, and extracellular vesicles, have been explored for molecular test development. We recently discovered the potential of tumor-educated blood platelets (TEP) as a noninvasive biomarker trove for RNA biomarker panels. TEPs are involved in the progression and spread of several solid tumors, and spliced TEP RNA surrogate signatures can provide specific information on the presence, location, and molecular characteristics of cancers. So far, TEP samples from patients with different tumor types, including lung, brain, and breast cancers, have been tested, and it has been shown that TEPs from patients with cancer are distinct from those with inflammatory and other noncancerous diseases. It remains to be investigated how platelets are "educated," which mechanisms cause intraplatelet RNA splicing, and whether the relative contribution of specific platelet subpopulations changes in patients with cancer. Ultimately, TEP RNA may complement currently used biosources and biomolecules employed for liquid biopsy diagnosis, potentially enhancing the detection of cancer in an early stage and facilitating noninvasive disease monitoring. Cancer Res; 78(13); 3407-12. ©2018 AACR.
Collapse
Affiliation(s)
- Myron G Best
- Department of Neurosurgery, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Pieter Wesseling
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, the Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
210
|
Xu WC, Qian G, Liu AQ, Li YQ, Zou HQ. Urinary Extracellular Vesicle: A Potential Source of Early Diagnostic and Therapeutic Biomarker in Diabetic Kidney Disease. Chin Med J (Engl) 2018; 131:1357-1364. [PMID: 29786051 PMCID: PMC5987509 DOI: 10.4103/0366-6999.232801] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE Diabetic kidney disease (DKD) has become one of the major causes of end-stage renal disease. Urinary extracellular vesicles (uEVs) contain rich biological information which could be the ideal source for noninvasive biomarkers of DKD. This review discussed the potential early diagnostic and therapeutic values of proteins and microRNAs in uEVs in DKD. DATA SOURCES This review was based articles published in PubMed, Embase, Cochrane, and Google Scholar databases up to November 20, 2017, with the following keywords: "Diabetic kidney disease", "Extracellular vesicle", and "Urine". STUDY SELECTION Relevant articles were carefully reviewed, with no exclusions applied to the study design and publication type. RESULTS There is no "gold standard" technology to separate and/or purify uEVs. The uEVs contain a variety of proteins and RNAs and participate in the physiological and pathological processes of the kidney. UEVs, especially urinary exosomes, may be useful biomarkers for early diagnosis and treatment to DKD. Furthermore, the uEVs has been used as a therapeutic target for DKD. CONCLUSION Proteins and nucleic acids in uEVs represent promising biomarker for the diagnosis and treatment of DKD.
Collapse
Affiliation(s)
- Wei-Cheng Xu
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Ge Qian
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Ai-Qun Liu
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Yong-Qiang Li
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - He-Qun Zou
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
211
|
Rackov G, Garcia-Romero N, Esteban-Rubio S, Carrión-Navarro J, Belda-Iniesta C, Ayuso-Sacido A. Vesicle-Mediated Control of Cell Function: The Role of Extracellular Matrix and Microenvironment. Front Physiol 2018; 9:651. [PMID: 29922170 PMCID: PMC5996101 DOI: 10.3389/fphys.2018.00651] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) — including exosomes, microvesicles and apoptotic bodies — have received much scientific attention last decade as mediators of a newly discovered cell-to-cell communication system, acting at short and long distances. EVs carry biologically active molecules, thus providing signals that influence a spectrum of functions in recipient cells during various physiological and pathological processes. Recent findings point to EVs as very attractive immunomodulatory therapeutic agents, vehicles for drug delivery and diagnostic and prognostic biomarkers in liquid biopsies. In addition, EVs interact with and regulate the synthesis of extracellular matrix (ECM) components, which is crucial for organ development and wound healing, as well as bone and cardiovascular calcification. EVs carrying matrix metalloproteinases (MMPs) are involved in ECM remodeling, thus modifying tumor microenvironment and contributing to premetastatic niche formation and angiogenesis. Here we review the role of EVs in control of cell function, with emphasis on their interaction with ECM and microenvironment in health and disease.
Collapse
Affiliation(s)
| | | | - Susana Esteban-Rubio
- Fundación de Investigación HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad CEU San Pablo, Madrid, Spain
| | | | | | - Angel Ayuso-Sacido
- IMDEA Nanoscience Institute, Madrid, Spain.,Fundación de Investigación HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad CEU San Pablo, Madrid, Spain
| |
Collapse
|
212
|
Sato S, Weaver AM. Extracellular vesicles: important collaborators in cancer progression. Essays Biochem 2018; 62:149-163. [PMID: 29666212 PMCID: PMC6377252 DOI: 10.1042/ebc20170080] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are membrane vesicles that are released from cells and mediate cell-cell communication. EVs carry protein, lipid, and nucleic acid cargoes that interact with recipient cells to alter their phenotypes. Evidence is accumulating that tumor-derived EVs can play important roles in all steps of cancer progression. Here, we review recent studies reporting critical roles for EVs in four major areas of cancer progression: promotion of cancer invasiveness and motility, enhancement of angiogenesis and vessel permeability, conditioning premetastatic niches, and immune suppression.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| |
Collapse
|
213
|
Yeung V, Webber JP, Dunlop EA, Morgan H, Hutton J, Gurney M, Jones E, Falcon-Perez J, Tabi Z, Errington R, Clayton A. Rab35-dependent extracellular nanovesicles are required for induction of tumour supporting stroma. NANOSCALE 2018; 10:8547-8559. [PMID: 29693684 DOI: 10.1039/c8nr02417k] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Communication between diseased cells and the microenvironment is a complex yet crucial element in progression of varied pathological processes. Recent studies in cancer highlight an important role for small extracellular nanovesicles secreted by cancer cells as modulators of cancer-associated stroma, leading to enhanced angiogenesis and metastatic priming. The intrinsic factors regulating extracellular nanovesicle biogenesis and secretion are therefore relevant in studies of nano-communication in the cancer milieu. We generated prostate cancer cells bearing stable knockdown of several candidate vesicle regulating factors and examined the impact on cell health, vesicle secretion and on communication with fibroblastic stromal cells. We highlight that RAB11B and RAB35 regulate phenotypically distinct nanovesicle populations, each accounting for only around 20% of the total. Depleting RAB35, but not RAB11B leaves a remaining population of vesicles whose phenotype is insufficient for driving fibroblast to myofibroblast differentiation, leading to attenuated motile behaviours in 3D in vitro models. Co-implantation of tumour cells with stromal fibroblasts in xenografts similarly showed that RAB11B knockdown had little effect on growth rates in vivo. In contrast, significant attenuation in growth, and attenuation of myofibroblasts at the tumour site was evident when using RAB35-knockdown cells. The study concludes that a RAB35 regulated nanovesicle sub-population is particularly important for communication between cancer and stromal cells, and is required for generating a tumour-supportive microenvironment.
Collapse
Affiliation(s)
- V Yeung
- Division of Cancer & Genetics, School of Medicine, Cardiff University, Heath Park Heath Park, Cardiff CF14 4XN, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Extracellular vesicles – biogenesis, composition, function, uptake and therapeutic applications. Biologia (Bratisl) 2018. [DOI: 10.2478/s11756-018-0047-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
215
|
Pedersen KW, Kierulf B, Neurauter A. Specific and Generic Isolation of Extracellular Vesicles with Magnetic Beads. Methods Mol Biol 2018; 1660:65-87. [PMID: 28828649 DOI: 10.1007/978-1-4939-7253-1_7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This chapter covers magnetic bead-based isolation and analysis of the smallest members of extracellular vesicles (EVs), the exosomes (30-150 nm), generally regarded to originate from the multivesicular bodies (MVBs). Also included, are descriptions of how to prepare samples prior to isolations. The magnetic bead-based isolation workflow is dramatically shortened both by omitting the pre-enrichment step and providing an option for a very short capture time. Three direct exosome isolation strategies are described: (1) "Specific and Direct," (2) "Semi Generic and Direct" and (3) "Generic and Direct" as well as exosome release from the magnetic beads. Detailed description of downstream exosome analysis is included covering flow cytometry, Western blot and electron microscopy. Finally, a description of exosome isolation from more complex starting material including urine and serum/plasma is discussed.
Collapse
Affiliation(s)
- Ketil W Pedersen
- Thermo Fisher Scientific, Ullernchausseen 52, PO Box 114, Smestad, 0379, Oslo, Norway.
| | - Bente Kierulf
- Thermo Fisher Scientific, Ullernchausseen 52, PO Box 114, Smestad, 0379, Oslo, Norway
| | - Axl Neurauter
- Thermo Fisher Scientific, Ullernchausseen 52, PO Box 114, Smestad, 0379, Oslo, Norway
| |
Collapse
|
216
|
Exosomes: Definition, Role in Tumor Development and Clinical Implications. CANCER MICROENVIRONMENT 2018; 11:13-21. [PMID: 29721824 DOI: 10.1007/s12307-018-0211-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 04/12/2018] [Indexed: 12/21/2022]
Abstract
Exosomes are microvesicles released by cells in both physiological and pathological situations. They are surrounded by a lipid bilayer with proteins derived from the origin cell, and contain a variety of molecules, such as nucleic acids. They represent an emerging mechanism of intercellular communication, and they play an important role in the pathogenesis of cancer, stimulating proliferation and aggressiveness of cancer cells, inducing a microenvironment favorable to tumor development and controlling immune responses. Because of the growing understanding of the potential implications of extracellular vesicles in the development of malignancies, research on exosomes, and its role as a diagnostic and therapeutic tool, constitutes nowadays a very exciting and promising field.
Collapse
|
217
|
Platelet microparticle delivered microRNA-Let-7a promotes the angiogenic switch. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2633-2643. [PMID: 29684582 DOI: 10.1016/j.bbadis.2018.04.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 01/14/2023]
Abstract
Platelet microparticle (PMP)-induced angiogenesis plays a key role in tumour metastasis and has been proposed to contribute towards cardiovascular disease by enhancing atherosclerotic plaque vulnerability. However, the mechanisms underlying PMP induced angiogenesis are ill defined. Recent reports demonstrate that PMPs deliver micro-RNAs (miRNAs) to recipient cells, controlling gene expression. We therefore evaluated whether miRNA transfer was a key regulator of PMP-induced angiogenesis. Co-culturing PMPs with human umbilical vein endothelial cells (HUVEC) on extracellular matrix gel induced robust capillary like structure formation. PMP treatment altered the release of angiogenesis modulators from HUVEC, including significantly reducing production of anti-angiogenic thrombospondin-1 (THBS-1). Both functional responses were abrogated by treating PMPs with RNase, suggesting the transfer of PMP-derived RNA was a critical event. PMPs were an abundant source of miRNA Let-7a, which was transferred to HUVEC following co-incubation. Using luciferase reporter assays we have shown that Let-7a directly targets the 3'UTR of the THBS-1 mRNA. HUVEC transfection with a Let-7a anti-sense oligonucleotide reduced the ability of PMPs to inhibit THBS-1 release, and significantly decreased PMP induced in vitro angiogenesis. Antibody neutralisation of THBS-1 reversed the anti-angiogenic effect of let-7a inhibition in PMP treated HUVEC, highlighting Let-7a dependent translational repression of THBS-1 drives angiogenesis. Importantly, plasmid overexpression of Let-7a in HUVEC alone induced robust tubule formation on extracellular matrix gel. These data reveal a new role for Let-7a in promoting angiogenesis and show for the first time PMPs induced angiogenic responses occur through miRNA regulation of HUVEC.
Collapse
|
218
|
Kanikarla-Marie P, Lam M, Sorokin AV, Overman MJ, Kopetz S, Menter DG. Platelet Metabolism and Other Targeted Drugs; Potential Impact on Immunotherapy. Front Oncol 2018; 8:107. [PMID: 29732316 PMCID: PMC5919962 DOI: 10.3389/fonc.2018.00107] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/27/2018] [Indexed: 12/13/2022] Open
Abstract
The role of platelets in cancer progression has been well recognized in the field of cancer biology. Emerging studies are elaborating further the additional roles and added extent that platelets play in promoting tumorigenesis. Platelets release factors that support tumor growth and also form heterotypic aggregates with tumor cells, which can provide an immune-evasive advantage. Their most critical role may be the inhibition of immune cell function that can negatively impact the body’s ability in preventing tumor establishment and growth. This review summarizes the importance of platelets in tumor progression, therapeutic response, survival, and finally the notion of immunotherapy modulation being likely to benefit from the inclusion of platelet inhibitors.
Collapse
Affiliation(s)
- Preeti Kanikarla-Marie
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Lam
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexey V Sorokin
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J Overman
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
219
|
Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018. [PMID: 29519806 DOI: 10.1182/blood-2017-05-743187] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Platelets have long been recognized as key players in hemostasis and thrombosis; however, growing evidence suggests that they are also significantly involved in cancer, the second leading cause of mortality worldwide. Preclinical and clinical studies showed that tumorigenesis and metastasis can be promoted by platelets through a wide variety of crosstalk between platelets and cancer cells. For example, cancer changes platelet behavior by directly inducing tumor-platelet aggregates, triggering platelet granule and extracellular vesicle release, altering platelet phenotype and platelet RNA profiles, and enhancing thrombopoiesis. Reciprocally, platelets reinforce tumor growth with proliferation signals, antiapoptotic effect, and angiogenic factors. Platelets also activate tumor invasion and sustain metastasis via inducing an invasive epithelial-mesenchymal transition phenotype of tumor cells, promoting tumor survival in circulation, tumor arrest at the endothelium, and extravasation. Furthermore, platelets assist tumors in evading immune destruction. Hence, cancer cells and platelets maintain a complex, bidirectional communication. Recently, aspirin (acetylsalicylic acid) has been recognized as a promising cancer-preventive agent. It is recommended at daily low dose by the US Preventive Services Task Force for primary prevention of colorectal cancer. The exact mechanisms of action of aspirin in chemoprevention are not very clear, but evidence has emerged that suggests a platelet-mediated effect. In this article, we will introduce how cancer changes platelets to be more cancer-friendly and highlight advances in the modes of action for aspirin in cancer prevention. We also discuss the opportunities, challenges, and opposing viewpoints on applying aspirin and other antiplatelet agents for cancer prevention and treatment.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - George M Yousef
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada; and
- Department of Medicine and
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
220
|
Alipoor SD, Mortaz E, Varahram M, Movassaghi M, Kraneveld AD, Garssen J, Adcock IM. The Potential Biomarkers and Immunological Effects of Tumor-Derived Exosomes in Lung Cancer. Front Immunol 2018; 9:819. [PMID: 29720982 PMCID: PMC5915468 DOI: 10.3389/fimmu.2018.00819] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/04/2018] [Indexed: 12/21/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths worldwide. Despite considerable achievements in lung cancer diagnosis and treatment, the global control of the disease remains problematic. In this respect, greater understanding of the disease pathology is crucially needed for earlier diagnosis and more successful treatment to be achieved. Exosomes are nano-sized particles secreted from most cells, which allow cross talk between cells and their surrounding environment via transferring their cargo. Tumor cells, just like normal cells, also secrete exosomes that are termed Tumor-Derived Exosome or tumor-derived exosome (TEX). TEXs have gained attention for their immuno-modulatory activities, which strongly affect the tumor microenvironment and antitumor immune responses. The immunological activity of TEX influences both the innate and adaptive immune systems including natural killer cell activity and regulatory T-cell maturation as well as numerous anti-inflammatory responses. In the context of lung cancer, TEXs have been studied in order to better understand the mechanisms underlying tumor metastasis and progression. As such, TEX has the potential to act both as a biomarker for lung cancer diagnosis as well as the response to therapy.
Collapse
Affiliation(s)
- Shamila D Alipoor
- Molecular Medicine Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Varahram
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Movassaghi
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aletta D Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| | - Ian M Adcock
- Airways Disease Section, Imperial College London, National Heart & Lung Institute, London, United Kingdom.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
221
|
Kulichkova VA, Selenina AV, Tomilin AN, Tsimokha AS. Establishment of the HeLa Cell Line with Stable Expression of CD63 Exosome Marker Fused with Fluorescent Protein TagRFP and HTBH Tag. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s1990519x18020049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
222
|
Computational Approach to Investigating Key GO Terms and KEGG Pathways Associated with CNV. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8406857. [PMID: 29850576 PMCID: PMC5925134 DOI: 10.1155/2018/8406857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 12/25/2022]
Abstract
Choroidal neovascularization (CNV) is a severe eye disease that leads to blindness, especially in the elderly population. Various endogenous and exogenous regulatory factors promote its pathogenesis. However, the detailed molecular biological mechanisms of CNV have not been fully revealed. In this study, by using advanced computational tools, a number of key gene ontology (GO) terms and KEGG pathways were selected for CNV. A total of 29 validated genes associated with CNV and 17,639 nonvalidated genes were encoded based on the features derived from the GO terms and KEGG pathways by using the enrichment theory. The widely accepted feature selection method-maximum relevance and minimum redundancy (mRMR)-was applied to analyze and rank the features. An extensive literature review for the top 45 ranking features was conducted to confirm their close associations with CNV. Identifying the molecular biological mechanisms of CNV as described by the GO terms and KEGG pathways may contribute to improving the understanding of the pathogenesis of CNV.
Collapse
|
223
|
Rossi M, Battafarano G, D'Agostini M, Del Fattore A. The Role of Extracellular Vesicles in Bone Metastasis. Int J Mol Sci 2018; 19:ijms19041136. [PMID: 29642618 PMCID: PMC5979436 DOI: 10.3390/ijms19041136] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple types of cancer have the specific ability to home to the bone microenvironment and cause metastatic lesions. Despite being the focus of intense investigation, the molecular and cellular mechanisms that regulate the metastasis of disseminated tumor cells still remain largely unknown. Bone metastases severely impact quality of life since they are associated with pain, fractures, and bone marrow aplasia. In this review, we will summarize the recent discoveries on the role of extracellular vesicles (EV) in the regulation of bone remodeling activity and bone metastasis occurrence. Indeed, it was shown that extracellular vesicles, including exosomes and microvesicles, released from tumor cells can modify the bone microenvironment, allowing the formation of osteolytic, osteosclerotic, and mixed mestastases. In turn, bone-derived EV can stimulate the proliferation of tumor cells. The inhibition of EV-mediated crosstalk between cancer and bone cells could represent a new therapeutic target for bone metastasis.
Collapse
Affiliation(s)
- Michela Rossi
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, 00165 Rome, Italy.
| | - Giulia Battafarano
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, 00165 Rome, Italy.
| | - Matteo D'Agostini
- Clinical Laboratory, Bambino Gesù Children's Hospital, 00165 Rome, Italy.
| | - Andrea Del Fattore
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, 00165 Rome, Italy.
| |
Collapse
|
224
|
Abstract
There has been remarkable insight into the importance of platelets in a wide range of pathophysiologic events, including inflammation and cancer progression. Thrombocytosis in cancer patients is a common finding. Tumor cells induce platelet activation and subsequent aggregation through direct and indirect mechanisms. Platelets are recognized to contribute to metastatic dissemination. There is plenty of evidence that components of the hemostatic system contribute to the process of angiogenesis. Furthermore, there are accumulated data on the substantial influence of blood platelets in the process of blood vessel formation during malignancy. Platelets appear to be the main physiologic transporters of proangiogenic and antiangiogenic factors. Moreover, they influence the process of angiogenesis through platelet-derived microparticles, microRNA, lipids, and variety of surface receptors. Platelets contribute to early and late stages of angiogenesis. Available data support the overall stimulatory effect of platelets on tumor angiogenesis. It raises the possibility that interfering with platelet function may be an effective antineoplastic treatment strategy.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University, 12 Ogrodowa St., 15-027, Bialystok, Poland. .,Department of Clinical Oncology, Comprehensive Cancer Center, Bialystok, Poland.
| | - Ewa Sierko
- Department of Oncology, Medical University, 12 Ogrodowa St., 15-027, Bialystok, Poland.,Department of Clinical Oncology, Comprehensive Cancer Center, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center, Bialystok, Poland
| | - Dominika Hempel
- Department of Oncology, Medical University, 12 Ogrodowa St., 15-027, Bialystok, Poland.,Department of Clinical Oncology, Comprehensive Cancer Center, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center, Bialystok, Poland
| | - Stephanie C Tucker
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Detroit, MI, USA
| | - Kenneth V Honn
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Detroit, MI, USA.,Department of Chemistry, Wayne State University, Detroit, MI, USA.,Department of Oncology, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
225
|
Nogués L, Benito-Martin A, Hergueta-Redondo M, Peinado H. The influence of tumour-derived extracellular vesicles on local and distal metastatic dissemination. Mol Aspects Med 2018; 60:15-26. [PMID: 29196097 PMCID: PMC5856602 DOI: 10.1016/j.mam.2017.11.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/18/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are key mediators of intercellular communication that have been ignored for decades. Tumour cells benefit from the secretion of vesicles as they can influence the behaviour of neighbouring tumour cells within the tumour microenvironment. Several studies have shown that extracellular vesicles play an active role in pre-metastatic niche formation and importantly, they are involved in the metastatic organotropism of different tumour types. Tumour-derived EVs carry and transfer molecules to recipient cells, modifying their behaviour through a process defined as "EV-driven education". EVs favour metastasis to sentinel lymph nodes and distal organs by reinforcing angiogenesis, inflammation and lymphangiogenesis. Hence, in this review we will summarize the main mechanisms by which tumour-derived EVs regulate lymph node and distal organ metastasis. Moreover, since some cancers metastasize through the lymphatic system, we will discuss recent discoveries about the presence and function of tumour EVs in the lymph. Finally, we will address the potential value of tumour EVs as prognostic biomarkers in liquid biopsies, specially blood and lymphatic fluid, and the use of these tools as early detectors of metastases.
Collapse
Affiliation(s)
- Laura Nogués
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Alberto Benito-Martin
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Marta Hergueta-Redondo
- Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain
| | - Héctor Peinado
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA; Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain.
| |
Collapse
|
226
|
Effects of platelets on cancer progression. Thromb Res 2018; 164 Suppl 1:S40-S47. [DOI: 10.1016/j.thromres.2018.01.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/21/2022]
|
227
|
PKH26 labeling of extracellular vesicles: Characterization and cellular internalization of contaminating PKH26 nanoparticles. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1350-1361. [PMID: 29551275 DOI: 10.1016/j.bbamem.2018.03.013] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 02/14/2018] [Accepted: 03/13/2018] [Indexed: 12/21/2022]
Abstract
PKH lipophilic dyes are highly fluorescent and stain membranes by intercalating their aliphatic portion into the exposed lipid bilayer. They have established use in labeling and tracking of cells in vivo and in vitro. Despite wide use of PKH-labeled extracellular vesicles (EVs) in cell targeting and functional studies, nonEV-associated fluorescent structures have never been examined systematically, nor was their internalization by cells. Here, we have characterized PKH26-positive particles in lymphoblastoid B exosome samples and exosome-free controls stained by ultracentrifugation, filtration, and sucrose-cushion-based and sucrose-gradient-based procedures, using confocal imaging and asymmetric-flow field-flow fractionation coupled to multi-angle light-scattering detector analysis. We show for the first time that numerous PKH26 nanoparticles (nine out of ten PKH26-positive particles) are formed during ultracentrifugation-based exosome staining, which are almost indistinguishable from PKH26-labeled exosomes in terms of size, surface area, and fluorescence intensity. When PKH26-labeled exosomes were purified through sucrose, PKH26 nanoparticles were differentiated from PKH26-labeled exosomes based on their reduced size. However, PKH26 nanoparticles were only physically removed from PKH26-labeled exosomes when separated on a sucrose gradient, and at the expense of low PKH26-labeled exosome recovery. Overall, low PKH26-positive particle recovery is characteristic of filtration-based exosome staining. Importantly, PKH26 nanoparticles are internalized by primary astrocytes into similar subcellular compartments as PKH26-labeled exosomes. Altogether, PKH26 nanoparticles can result in false-positive signals for stained EVs that can compromise the interpretation of EV internalization. Thus, for use in EV uptake and functional studies, sucrose-gradient-based isolation should be the method of choice to obtain PKH26-labeled exosomes devoid of PKH26 nanoparticles.
Collapse
|
228
|
Lazar S, Goldfinger LE. Platelet Microparticles and miRNA Transfer in Cancer Progression: Many Targets, Modes of Action, and Effects Across Cancer Stages. Front Cardiovasc Med 2018; 5:13. [PMID: 29564336 PMCID: PMC5850852 DOI: 10.3389/fcvm.2018.00013] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
Platelet-derived microparticles (PMPs) have long been known to increase in circulation in the presence of cancer, and have been considered to be cancer promoting by multiple mechanisms including shrouding of circulating tumor cells allowing immune evasion, inducing a procoagulant state associated with increased risk for venous thromboembolic events in cancer patients, and supporting metastatic dissemination by establishment of niches for anchorage of circulating tumor cells. These modes of PMP-enhanced progression of late stage cancer are generally based on the adhesive and procoagulant surfaces of PMPs. However, it is now clear that PMPs can also act as intercellular signaling vesicles, by fusion with target cells and transfer of a broad array of platelet-derived molecular contents including growth factors, angiogenic modulators, second messengers, lipids, and nucleic acids. It is also now well established that PMPs are major repositories of microRNAs (miRNAs). In recent years, new roles of PMPs in cancer have begun emerging, primarily reflecting their ability to transfer miRNA contents and modulate gene expression in target cells, allowing PMPs to affect cancer development at many stages. PMPs have been shown to interact with and transfer miRNAs to various blood vascular cells including endothelium, macrophages and neutrophils. As each of these contributes to cancer progression, PMP-mediated miRNA transfer can affect immune response, NETosis, tumor angiogenesis, and likely other cancer-associated processes. Recently, PMP miRNA transfer was found to suppress primary tumor growth, via PMP infiltration in solid tumors, anchorage to tumor cells and direct miRNA transfer, resulting in tumor cell gene suppression and inhibition of tumor growth. This mini-review will summarize current knowledge of PMP-miRNA interactions with cancer-associated cells and effects in cancer progression, and will indicate new research directions for understanding platelet-cancer interactions.
Collapse
Affiliation(s)
- Sophia Lazar
- The Sol Sherry Thrombosis Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Lawrence E Goldfinger
- The Sol Sherry Thrombosis Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| |
Collapse
|
229
|
Polymer-Mediated Inhibition of Pro-invasive Nucleic Acid DAMPs and Microvesicles Limits Pancreatic Cancer Metastasis. Mol Ther 2018; 26:1020-1031. [PMID: 29550075 DOI: 10.1016/j.ymthe.2018.02.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/16/2018] [Accepted: 02/18/2018] [Indexed: 12/16/2022] Open
Abstract
Nucleic acid binding polymers (NABPs) have been extensively used as vehicles for DNA and RNA delivery. More recently, we discovered that a subset of these NABPs can also serve as anti-inflammatory agents by capturing pro-inflammatory extracellular nucleic acids and associated protein complexes that promote activation of toll-like receptors (TLRs) in diseases such as lupus erythematosus. Nucleic-acid-mediated TLR signaling also facilitates tumor progression and metastasis in several cancers, including pancreatic cancer (PC). In addition, extracellular DNA and RNA circulate on or within lipid microvesicles, such as microparticles or exosomes, which also promote metastasis by inducing pro-tumorigenic signaling in cancer cells and pre-conditioning secondary sites for metastatic establishment. Here, we explore the use of an NABP, the 3rd generation polyamidoamine dendrimer (PAMAM-G3), as an anti-metastatic agent. We show that PAMAM-G3 not only inhibits nucleic-acid-mediated activation of TLRs and invasion of PC tumor cells in vitro, but can also directly bind extracellular microvesicles to neutralize their pro-invasive effects as well. Moreover, we demonstrate that PAMAM-G3 dramatically reduces liver metastases in a syngeneic murine model of PC. Our findings identify a promising therapeutic application of NABPs for combating metastatic disease in PC and potentially other malignancies.
Collapse
|
230
|
Maia J, Caja S, Strano Moraes MC, Couto N, Costa-Silva B. Exosome-Based Cell-Cell Communication in the Tumor Microenvironment. Front Cell Dev Biol 2018. [PMID: 29515996 PMCID: PMC5826063 DOI: 10.3389/fcell.2018.00018] [Citation(s) in RCA: 452] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tumors are not isolated entities, but complex systemic networks involving cell-cell communication between transformed and non-transformed cells. The milieu created by tumor-associated cells may either support or halt tumor progression. In addition to cell-cell contact, cells communicate through secreted factors via a highly complex system involving characteristics such as ligand concentration, receptor expression and integration of diverse signaling pathways. Of these, extracellular vesicles, such as exosomes, are emerging as novel cell-cell communication mediators in physiological and pathological scenarios. Exosomes, membrane vesicles of endocytic origin released by all cells (both healthy and diseased), ranging in size from 30 to 150 nm, transport all the main biomolecules, including lipids, proteins, DNAs, messenger RNAs and microRNA, and perform intercellular transfer of components, locally and systemically. By acting not only in tumor cells, but also in tumor-associated cells such as fibroblasts, endothelium, leukocytes and progenitor cells, tumor- and non-tumor cells-derived exosomes have emerged as new players in tumor growth and invasion, tumor-associated angiogenesis, tissue inflammation and immunologic remodeling. In addition, due to their property of carrying molecules from their cell of origin to the peripheral circulation, exosomes have been increasingly studied as sources of tumor biomarkers in liquid biopsies. Here we review the current literature on the participation of exosomes in the communication between tumor and tumor-associated cells, highlighting the role of this process in the setup of tumor microenvironments that modulate tumor initiation and metastasis.
Collapse
Affiliation(s)
- Joana Maia
- Systems Oncology Group, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Sergio Caja
- Systems Oncology Group, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | | | - Nuno Couto
- Systems Oncology Group, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
231
|
Gutkin A, Uziel O, Beery E, Nordenberg J, Pinchasi M, Goldvaser H, Henick S, Goldberg M, Lahav M. Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 2018; 7:59173-59188. [PMID: 27385095 PMCID: PMC5312303 DOI: 10.18632/oncotarget.10384] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 06/06/2016] [Indexed: 11/25/2022] Open
Abstract
Exosomes are small (30-100nm) vesicles secreted from all cell types serving as inter-cell communicators and affecting biological processes in “recipient” cells upon their uptake. The current study demonstrates for the first time that hTERT mRNA, the transcript of the enzyme telomerase, is shuttled from cancer cells via exosomes into telomerase negative fibroblasts, where it is translated into a fully active enzyme and transforms these cells into telomerase positive, thus creating a novel type of cells; non malignant cells with telomerase activity. All tested telomerase positive cells, including cancer cells and non malignant cells with overexpressed telomerase secreted exosomal hTERT mRNA in accordance with the endogenous levels of their hTERT mRNA and telomerase activity. Similarly exosomes isolated from sera of patients with pancreatic and lung cancer contained hTERT mRNA as well. Telomerase activity induced phenotypic changes in the recipient fibroblasts including increased proliferation, extension of life span and postponement of senescence. In addition, telomerase activity protected the fibroblasts from DNA damage induced by phleomycin and from apoptosis, indicating that also telomerase “extracurricular” activities are manifested in the recipient cells. The shuttle of telomerase from cancer cells into fibroblasts and the induction of these changes may contribute to the alterations of cancer microenvironment and its role in cancer. The described process has an obvious therapeutic potential which will be explored in further studies.
Collapse
Affiliation(s)
- Anna Gutkin
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Orit Uziel
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Einat Beery
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Jardena Nordenberg
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Maria Pinchasi
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Hadar Goldvaser
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel.,Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Steven Henick
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Michal Goldberg
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Petah Tikva, Israel
| | - Meir Lahav
- The Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel.,Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
232
|
Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin Exp Metastasis 2018; 35:269-284. [PMID: 29307118 DOI: 10.1007/s10585-017-9870-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Tumors often overcome the cytotoxic effects of chemotherapy through either acquired or environment-mediated drug resistance. In addition, signals from the microenvironment obfuscate the beneficial effects of chemotherapy and may facilitate progression and metastatic dissemination. Seminal mediators in chemotherapy-induced metastasis appear to be a wide range of hematopoietic, mesenchymal and immune progenitor cells, originating from the bone marrow. The actual purpose of these cells is to orchestrate the repair response to the cytotoxic damage of chemotherapy. However, these repair responses are exploited by tumor cells at every step of the metastatic cascade, ranging from tumor cell invasion, intravasation and hematogenous dissemination to extravasation and effective colonization at the metastatic site. A better understanding of the mechanistic underpinnings of chemotherapy-induced metastasis will allow us to better predict which patients are more likely to exhibit pro-metastatic responses to chemotherapy and will help develop new therapeutic strategies to neutralize chemotherapy-driven prometastatic changes.
Collapse
|
233
|
Zhao H, Achreja A, Iessi E, Logozzi M, Mizzoni D, Di Raimo R, Nagrath D, Fais S. The key role of extracellular vesicles in the metastatic process. Biochim Biophys Acta Rev Cancer 2018; 1869:64-77. [PMID: 29175553 PMCID: PMC5800973 DOI: 10.1016/j.bbcan.2017.11.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/16/2017] [Accepted: 11/22/2017] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs), including exosomes, have a key role in the paracrine communication between organs and compartments. EVs shuttle virtually all types of biomolecules such as proteins, lipids, nucleic acids, metabolites and even pharmacological compounds. Their ability to transfer their biomolecular cargo into target cells enables EVs to play a key role in intercellular communication that can regulate cellular functions such as proliferation, apoptosis and migration. This has led to the emergence of EVs as a key player in tumor growth and metastasis through the formation of "tumor niches" in target organs. Recent data have also been shown that EVs may transform the microenvironment of primary tumors thus favoring the selection of cancer cells with a metastatic behavior. The release of EVs from resident non-malignant cells may contribute to the metastatic processes as well. However, cancer EVs may induce malignant transformation in resident mesenchymal stem cells, suggesting that the metastatic process is not exclusively due to circulating tumor cells. In this review, we outline and discuss evidence-based roles of EVs in actively regulating multiple steps of the metastatic process and how we can leverage EVs to impair metastasis.
Collapse
Affiliation(s)
- Hongyun Zhao
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Abhinav Achreja
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Elisabetta Iessi
- Department of Oncology and Molecular Medicine, National Institute of Health, viale Regina Elena 299, 00161, Rome, Italy.
| | - Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, National Institute of Health, viale Regina Elena 299, 00161, Rome, Italy.
| | - Davide Mizzoni
- Department of Oncology and Molecular Medicine, National Institute of Health, viale Regina Elena 299, 00161, Rome, Italy.
| | - Rossella Di Raimo
- Department of Oncology and Molecular Medicine, National Institute of Health, viale Regina Elena 299, 00161, Rome, Italy
| | - Deepak Nagrath
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, National Institute of Health, viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
234
|
Clinical and biological significance of HAX-1 overexpression in nasopharyngeal carcinoma. Oncotarget 2017; 7:12505-24. [PMID: 26871467 PMCID: PMC4914301 DOI: 10.18632/oncotarget.7274] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/23/2016] [Indexed: 12/22/2022] Open
Abstract
HS1-associated protein X-1 (HAX-1) is an important marker in many types of cancers and contributes to cancer progression and metastasis. We examined the expression of HAX-1 in nasopharyngeal carcinoma (NPC) and experimentally manipulated its expression. We observed that HAX-1 expression is elevated in NPC and is correlated with lymph node metastasis, M classification, clinical stage, and poor prognosis. In addition, overexpression of HAX-1 promoted NPC proliferation both in vitro and in vivo. Exosomes are potential carriers of pro-tumorigenic factors that participate in oncogenesis. We found that NPC-derived exosomes are enriched in HAX-1 and accelerate NPC tumor growth and angiogenesis in vitro and in vivo. Furthermore, we demonstrated that oncogenic HAX-1 facilitates the growth of NPC when it is transferred via exosomes to recipient human umbilical vein endothelial cells (HUVECs). Oncogenic HAX-1 also increases the proliferation, migration, and angiogenic activity of HUVECs. Our findings provide unique insight into the pathogenesis of NPC and underscore the need to explore novel therapeutic targets such as HAX-1 to improve NPC treatment.
Collapse
|
235
|
Badimon L, Suades R, Arderiu G, Peña E, Chiva-Blanch G, Padró T. Microvesicles in Atherosclerosis and Angiogenesis: From Bench to Bedside and Reverse. Front Cardiovasc Med 2017; 4:77. [PMID: 29326946 PMCID: PMC5741657 DOI: 10.3389/fcvm.2017.00077] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/22/2017] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis (AT) is a progressive chronic disease involving lipid accumulation, fibrosis, and inflammation in medium and large-sized arteries, and it is the main cause of cardiovascular disease (CVD). AT is caused by dyslipidemia and mediated by both innate and adaptive immune responses. Despite lipid-lowering drugs have shown to decrease the risk of cardiovascular events (CVEs), there is a significant burden of AT-related morbidity and mortality. Identification of subjects at increased risk for CVE as well as discovery of novel therapeutic targets for improved treatment strategies are still unmet clinical needs in CVD. Microvesicles (MVs), small extracellular plasma membrane particles shed by activated and apoptotic cells have been widely linked to the development of CVD. MVs from vascular and resident cells by facilitating exchange of biological information between neighboring cells serve as cellular effectors in the bloodstream and play a key role in all stages of disease progression. This article reviews the current knowledge on the role of MVs in AT and CVD. Attention is focused on novel aspects of MV-mediated regulatory mechanisms from endothelial dysfunction, vascular wall inflammation, oxidative stress, and apoptosis to coagulation and thrombosis in the progression and development of atherothrombosis. MV contribution to vascular remodeling is also discussed, with a particular emphasis on the effect of MVs on the crosstalk between endothelial cells and smooth muscle cells, and their role regulating the active process of AT-driven angiogenesis and neovascularization. This review also highlights the latest findings and main challenges on the potential prognostic, diagnostic, and therapeutic value of cell-derived MVs in CVD. In summary, MVs have emerged as new regulators of biological functions in atherothrombosis and might be instrumental in cardiovascular precision medicine; however, significant efforts are still needed to translate into clinics the latest findings on MV regulation and function.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Rosa Suades
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Gemma Arderiu
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Esther Peña
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| |
Collapse
|
236
|
Nana-Sinkam SP, Acunzo M, Croce CM, Wang K. Extracellular Vesicle Biology in the Pathogenesis of Lung Disease. Am J Respir Crit Care Med 2017; 196:1510-1518. [PMID: 28678586 PMCID: PMC5754438 DOI: 10.1164/rccm.201612-2457pp] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 06/29/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Serge P. Nana-Sinkam
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Mario Acunzo
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, James Comprehensive Cancer Center, Ohio State University, Columbus, Ohio; and
| | - Kai Wang
- Institutes for Systems Biology, Seattle, Washington
| |
Collapse
|
237
|
Zarà M, Guidetti GF, Boselli D, Villa C, Canobbio I, Seppi C, Visconte C, Canino J, Torti M. Release of Prometastatic Platelet-Derived Microparticles Induced by Breast Cancer Cells: A Novel Positive Feedback Mechanism for Metastasis. TH OPEN 2017; 1:e155-e163. [PMID: 31249921 PMCID: PMC6524851 DOI: 10.1055/s-0037-1613674] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/08/2017] [Indexed: 12/16/2022] Open
Abstract
Circulating platelets and platelet-derived microparticles are regulators of cancer metastasis. In this study, we show that breast cancer cells induce platelet aggregation and lead to the release of platelet-derived microparticles. Although able to cause comparable aggregation, the highly aggressive MDA-MB-231 cells were more potent than the poorly aggressive MCF7 cells in inducing platelet-derived microparticles release, which was comparable to that promoted by thrombin. MDA-MB-231 cells were able to bind and internalize both MCF7- and MDA-MB-231-induced platelet-derived microparticles with comparable efficiency. By contrast, MCF7 cells did not interact with either type of platelet-derived microparticles. Upon internalization, only platelet-derived microparticles released by platelet stimulation with MDA-MB-231 cells, but not those released upon stimulation with MCF7 cells, caused activation of MDA-MB-231 cells and promoted the phosphorylation of selected signaling proteins, including p38MAPK and myosin light chain. Accordingly, MDA-MB-231-induced, but not MCF7-induced, platelet-derived microparticles dose-dependently stimulated migration and invasion of targeted MDA-MB-231 cells. These results identify a novel paracrine positive feedback mechanism initiated by aggressive breast cancer cell types to potentiate their invasive phenotype through the release of platelet-derived microparticles.
Collapse
Affiliation(s)
- Marta Zarà
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | | | - Chiara Villa
- FRACTAL - San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Claudio Seppi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Caterina Visconte
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Jessica Canino
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
238
|
Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC. Paracrine Mechanisms of Mesenchymal Stem Cells in Tissue Repair. Methods Mol Biol 2017; 1416:123-46. [PMID: 27236669 DOI: 10.1007/978-1-4939-3584-0_7] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue regeneration from transplanted mesenchymal stromal cells (MSC) either through transdifferentiation or cell fusion was originally proposed as the principal mechanism underlying their therapeutic action. However, several studies have now shown that both these mechanisms are very inefficient. The low MSC engraftment rate documented in injured areas also refutes the hypothesis that MSC repair tissue damage by replacing cell loss with newly differentiated cells. Indeed, despite evidence of preferential homing of MSC to the site of myocardial ischemia, exogenously administered MSC show poor survival and do not persist in the infarcted area. Therefore, it has been proposed that the functional benefits observed after MSC transplantation in experimental models of tissue injury might be related to the secretion of soluble factors acting in a paracrine fashion. This hypothesis is supported by pre-clinical studies demonstrating equal or even improved organ function upon infusion of MSC-derived conditioned medium (MSC-CM) compared with MSC transplantation. Identifying key MSC-secreted factors and their functional role seems a reasonable approach for a rational design of nextgeneration MSC-based therapeutics. Here, we summarize the major findings regarding both different MSC-mediated paracrine actions and the identification of paracrine mediators.
Collapse
Affiliation(s)
- Massimiliano Gnecchi
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy. .,Department of Cardiothoracic and Vascular Sciences - Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy. .,Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy. .,Department of Medicine, University of Cape Town, Cape Town, South Africa.
| | - Patrizia Danieli
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences - Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giuseppe Malpasso
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences - Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Chiara Ciuffreda
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences - Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
239
|
Vogel R, Pal AK, Jambhrunkar S, Patel P, Thakur SS, Reátegui E, Parekh HS, Saá P, Stassinopoulos A, Broom MF. High-Resolution Single Particle Zeta Potential Characterisation of Biological Nanoparticles using Tunable Resistive Pulse Sensing. Sci Rep 2017; 7:17479. [PMID: 29234015 PMCID: PMC5727177 DOI: 10.1038/s41598-017-14981-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/17/2017] [Indexed: 12/25/2022] Open
Abstract
Physicochemical properties of nanoparticles, such as size, shape, surface charge, density, and porosity play a central role in biological interactions and hence accurate determination of these characteristics is of utmost importance. Here we propose tunable resistive pulse sensing for simultaneous size and surface charge measurements on a particle-by-particle basis, enabling the analysis of a wide spectrum of nanoparticles and their mixtures. Existing methodologies for measuring zeta potential of nanoparticles using resistive pulse sensing are significantly improved by including convection into the theoretical model. The efficacy of this methodology is demonstrated for a range of biological case studies, including measurements of mixed anionic, cationic liposomes, extracellular vesicles in plasma, and in situ time study of DNA immobilisation on the surface of magnetic nanoparticles. The high-resolution single particle size and zeta potential characterisation will provide a better understanding of nano-bio interactions, positively impacting nanomedicine development and their regulatory approval.
Collapse
Affiliation(s)
- Robert Vogel
- School of Mathematics and Physics, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Anoop K Pal
- Izon Science US Limited, 85 Bolton Street, STE 108, Cambridge, MA, 02140, USA
| | - Siddharth Jambhrunkar
- Mucosal Diseases Group, Translational Research Institute, The University of Queensland, 37 Kent St., Woolloongabba, QLD 4102, Australia
| | - Pragnesh Patel
- Izon Science US Limited, 85 Bolton Street, STE 108, Cambridge, MA, 02140, USA
| | - Sachin S Thakur
- School of Pharmacy, The University of Queensland, 20 Cornwall St., Woolloongabba, QLD 4102, Australia
| | - Eduardo Reátegui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Harendra S Parekh
- School of Pharmacy, The University of Queensland, 20 Cornwall St., Woolloongabba, QLD 4102, Australia
| | - Paula Saá
- Scientific Affairs, American Red Cross, Rockville, MD, 20877, USA
| | | | - Murray F Broom
- Izon Science Limited, 8C Homersham Place, PO Box 39168, Burnside, Christchurch 8053, New Zealand
| |
Collapse
|
240
|
Nurden A. Platelets, inflammation and tissue regeneration. Thromb Haemost 2017; 105 Suppl 1:S13-33. [DOI: 10.1160/ths10-11-0720] [Citation(s) in RCA: 469] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 02/04/2011] [Indexed: 12/20/2022]
Abstract
SummaryBlood platelets have long been recognised to bring about primary haemostasis with deficiencies in platelet production and function manifesting in bleeding while upregulated function favourises arterial thrombosis. Yet increasing evidence indicates that platelets fulfil a much wider role in health and disease. First, they store and release a wide range of biologically active substances including the panoply of growth factors, chemokines and cytokines released from α-granules. Membrane budding gives rise to microparticles (MPs), another active participant within the blood stream. Platelets are essential for the innate immune response and combat infection (viruses, bacteria, micro-organisms). They help maintain and modulate inflammation and are a major source of pro-inflammatory molecules (e.g. P-selectin, tissue factor, CD40L, metalloproteinases). As well as promoting coagulation, they are active in fibrinolysis; wound healing, angiogenesis and bone formation as well as in maternal tissue and foetal vascular remodelling. Activated platelets and MPs intervene in the propagation of major diseases. They are major players in atherosclerosis and related diseases, pathologies of the central nervous system (Alzheimers disease, multiple sclerosis), cancer and tumour growth. They participate in other tissue-related acquired pathologies such as skin diseases and allergy, rheumatoid arthritis, liver disease; while, paradoxically, autologous platelet-rich plasma and platelet releasate are being used as an aid to promote tissue repair and cellular growth. The above mentioned roles of platelets are now discussed.
Collapse
|
241
|
Liu Q, Zhang H, Jiang X, Qian C, Liu Z, Luo D. Factors involved in cancer metastasis: a better understanding to "seed and soil" hypothesis. Mol Cancer 2017; 16:176. [PMID: 29197379 PMCID: PMC5712107 DOI: 10.1186/s12943-017-0742-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Metastasis has intrigued researchers for more than 100 years. Despite the development of technologies and therapeutic strategies, metastasis is still the major cause of cancer-related death until today. The famous "seed and soil" hypothesis is widely cited and accepted, and it still provides significant instructions in cancer research until today. To our knowledge, there are few reviews that comprehensively and correlatively focus on both the seed and soil factors involved in cancer metastasis; moreover, despite the fact that increasingly underlying mechanisms and concepts have been defined recently, previous perspectives are appealing but may be limited. Hence, we reviewed factors involved in cancer metastasis, including both seed and soil factors. By integrating new concepts with the classic hypothesis, we aim to provide a comprehensive understanding of the "seed and soil" hypothesis and to conceptualize the framework for understanding factors involved in cancer metastasis. Based on a dynamic overview of this field, we also discuss potential implications for future research and clinical therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Liu
- First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Hongfei Zhang
- Queen Mary School, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Xiaoli Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China
| | - Caiyun Qian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
| |
Collapse
|
242
|
Seizer P, May AE. Platelets and matrix metalloproteinases. Thromb Haemost 2017; 110:903-9. [DOI: 10.1160/th13-02-0113] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 06/18/2013] [Indexed: 11/05/2022]
Abstract
SummaryMatrix metalloproteinases (MMPs) and their inhibitors essentially contribute to a variety of pathophysiologies by modulating cell migration, tissue degradation and inflammation. Platelet-associated MMP activity appears to play a major role in these processes. First, platelets can concentrate leukocyte-derived MMP activity to sites of vascular injury by leukocyte recruitment. Second, platelets stimulate MMP production in e.g. leukocytes, endothelial cells, or tumour cells by direct receptor interaction or/and by paracrine pathways. Third, platelets synthesise and secrete a variety of MMPs including MMP-1, MMP-2, MMP-3, and MMP-14 (MT1-MMP), and potentially MMP-9 as well as the tissue inhibitors of metalloproteinase (TIMPs). This review focuses on platelet-derived and platelet-induced MMPs and their inhibitors.
Collapse
|
243
|
Lima L, Oliveira A, Campos L, Bonamino M, Chammas R, Werneck C, Vicente C, Barcinski M, Petersen L, Monteiro R. Malignant transformation in melanocytes is associated with increased production of procoagulant microvesicles. Thromb Haemost 2017; 106:712-23. [DOI: 10.1160/th11-03-0143] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 06/30/2011] [Indexed: 01/08/2023]
Abstract
SummaryShedding of microvesicles (MVs) by cancer cells is implicated in a variety of biological effects, including the establishment of cancer-associated hypercoagulable states. However, the mechanisms underlying malignant transformation and the acquisition of procoagulant properties by tumour-derived MVs are poorly understood. Here we investigated the procoagulant and prothrombotic properties of MVs produced by a melanocyte-derived cell line (melan-a) as compared to its tumourigenic melanoma counterpart Tm1. Tumour cells exhibit a two-fold higher rate of MVs production as compared to melan-a. Melanoma MVs display greater procoagulant activity and elevated levels of the clotting initiator protein tissue factor (TF). On the other hand, tumour- and melanocyte- derived MVs expose similar levels of the procoagulant lipid phosphatidylserine, displaying identical abilities to support thrombin generation by the prothrombinase complex. By using an arterial thrombosis model, we observed that melanoma- but not melanocyte-derived MVs strongly accelerate thrombus formation in a TF-dependent manner, and accumulate at the site of vascular injury. Analysis of plasma obtained from melanoma-bearing mice showed the presence of MVs with a similar procoagulant pattern as compared to Tm1 MVs produced in vitro. Remarkably, flow-cytometric analysis demonstrated that 60% of ex vivo MVs are TF-positive and carry the melanoma-associated antigen, demonstrating its tumour origin. Altogether our data suggest that malignant transformation in melanocytes increases the production of procoagulant MVs, which may contribute for a variety of coagulation- related protumoural responses.
Collapse
|
244
|
Ball S, Nugent K. Microparticles in Hematological Malignancies: Role in Coagulopathy and Tumor Pathogenesis. Am J Med Sci 2017; 355:207-214. [PMID: 29549921 DOI: 10.1016/j.amjms.2017.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/09/2017] [Accepted: 11/25/2017] [Indexed: 12/12/2022]
Abstract
Microparticles (MP) are submicron vesicles released from various cells in response to activation, injury or apoptosis. They contain different structural and functional proteins and RNAs, which contribute to physiological intercellular "crosstalk" and to the pathogenesis of various diseases including cancer. In hematological malignancies, these MPs participate in the initiation and propagation of thrombosis through different pathways. They have a role in the angiogenesis, malignant cell survival and metastasis. MPs act as a mediator of resistance of leukemic cells to chemotherapy. The number of MPs is one of the prognostic factors following stem cell transplant, and studies have also found they contribute to the pathogenesis of graft versus host disease. MPs are being tested as therapeutic options in leukemias and graft versus host disease. Future studies should help us understand the interactions between MPs and cancer cells better, thereby opening new approaches for treatment of hematological malignancies.
Collapse
Affiliation(s)
- Somedeb Ball
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas.
| | - Kenneth Nugent
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
245
|
Pando A, Reagan JL, Quesenberry P, Fast LD. Extracellular vesicles in leukemia. Leuk Res 2017; 64:52-60. [PMID: 29190514 DOI: 10.1016/j.leukres.2017.11.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/12/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EV) are nano-sized membrane enclosed vehicles that are involved in cell-to-cell communication and carry cargo that is representative of the parent cell. Recent studies have highlighted the significant roles leukemia EVs play in tumor progression, and ways in which they can lead to treatment evasion, thus meriting further investigation. Leukemia EVs are involved in crosstalk between the leukemia cell and its surroundings, transforming it into a cancer favorable microenvironment. Due to the diverse biological content found in leukemia EVs, they have an assortment of effects on the cells they interact with and can be harnessed as candidates for diagnostic and therapeutic treatments. This review focuses on EVs in the context of leukemia and the means by which they modulate their microenvironment, hematopoiesis, and the immune system to facilitate malignancy. We will also address current and prospective EV-based therapeutics.
Collapse
Affiliation(s)
- Alejandro Pando
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - John L Reagan
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter Quesenberry
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Loren D Fast
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
246
|
Zaldivia MTK, McFadyen JD, Lim B, Wang X, Peter K. Platelet-Derived Microvesicles in Cardiovascular Diseases. Front Cardiovasc Med 2017; 4:74. [PMID: 29209618 PMCID: PMC5702324 DOI: 10.3389/fcvm.2017.00074] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/07/2017] [Indexed: 12/15/2022] Open
Abstract
Microvesicles (MVs) circulating in the blood are small vesicles (100–1,000 nm in diameter) derived from membrane blebs of cells such as activated platelets, endothelial cells, and leukocytes. A growing body of evidence now supports the concept that platelet-derived microvesicles (PMVs), the most abundant MVs in the circulation, are important regulators of hemostasis, inflammation, and angiogenesis. Compared with healthy individuals, a large increase of circulating PMVs has been observed, particularly in patients with cardiovascular diseases. As observed in MVs from other parent cells, PMVs exert their biological effects in multiple ways, such as triggering various intercellular signaling cascades and by participating in transcellular communication by the transfer of their “cargo” of cytoplasmic components and surface receptors to other cell types. This review describes our current understanding of the potential role of PMVs in mediating hemostasis, inflammation, and angiogenesis and their consequences on the pathogenesis of cardiovascular diseases, such as atherosclerosis, myocardial infarction, and venous thrombosis. Furthermore, new developments of the therapeutic potential of PMVs for the treatment of cardiovascular diseases will be discussed.
Collapse
Affiliation(s)
- Maria T K Zaldivia
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - James D McFadyen
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,Department of Haematology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Bock Lim
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,Heart Centre, The Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
247
|
Renaud-Picard B, Toussaint J, Leclercq A, Reeb J, Kessler L, Toti F, Kessler R. [Membranous microparticles and respiratory disease]. Rev Mal Respir 2017; 34:1058-1071. [PMID: 29132745 DOI: 10.1016/j.rmr.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/05/2017] [Indexed: 01/30/2023]
Abstract
Microparticles (MP) are plasmic membrane fragments released from cells after physiological stimulation or stress conditions like inflammation or infection. Their production is correlated to the rate of cell apoptosis. All types of cells can produce MP but they are produced mainly by platelets, endothelial cells, and leukocytes. They carry many bio-active molecules on their surface, specific to the parental cell, giving them the ability to be biomarkers and bio-effectors. MP are present in circulating blood, tissues and many biological fluids. Circulating MP levels can change during the course of many diseases. They have been the subject of many studies in the fields of cardiovascular disease and oncology. In the lungs, they are present in circulating blood and in the airways. They seem to have a role in pulmonary homeostasis in physiological situations and also in the expression of several disease processes. In this review of the literature, we were interested in the quantitative and qualitative variations in MP and their impact in airway diseases like chronic obstructive pulmonary disease (COPD) and asthma, pulmonary fibrosis and pulmonary hypertension.
Collapse
Affiliation(s)
- B Renaud-Picard
- Service de pneumologie, nouvel hôpital Civil, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67000 Strasbourg, France; EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France.
| | - J Toussaint
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - A Leclercq
- Service de pneumologie, nouvel hôpital Civil, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67000 Strasbourg, France; EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - J Reeb
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - L Kessler
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - F Toti
- EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| | - R Kessler
- Service de pneumologie, nouvel hôpital Civil, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67000 Strasbourg, France; EA 7293, fédération de médecine translationnelle, université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
248
|
You B, Shan Y, Bao L, Chen J, Yang L, Zhang Q, Zhang W, Zhang Z, Zhang J, Shi S, You Y. The biology and function of extracellular vesicles in nasopharyngeal carcinoma (Review). Int J Oncol 2017; 52:38-46. [PMID: 29138808 DOI: 10.3892/ijo.2017.4202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/02/2017] [Indexed: 11/06/2022] Open
Abstract
Extracellular vesicles are a heterogeneous group of membrane-enclosed vesicles, which play an important role in intercellular communication. Increasing number of studies have shown that tumor-derived extracellular vesicles might be involved in the transfer of oncogenic cargo (proteins, lipids, messenger RNA, microRNA, non-coding RNAs and DNA) through which cancer cells could shape the tumor microenvironment and influence tumor progression. Nasopharyngeal carcinoma-derived extracellular vesicles have also reported to facilitate tumor proliferation, metastasis and immune escape. Moreover, nasopharyngeal carcinoma-derived extracellular vesicles might serve as biomarkers for early diagnosis and therapeutic targets. The present review provides information on the biological and clinical significance of extracellular vesicles in tumors, especially in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Bo You
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ying Shan
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lili Bao
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jing Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Liu Yang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qicheng Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wei Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhenxin Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jie Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Si Shi
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yiwen You
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
249
|
Chen L, Chen R, Kemper S, Brigstock DR. Pathways of production and delivery of hepatocyte exosomes. J Cell Commun Signal 2017; 12:343-357. [PMID: 29063370 DOI: 10.1007/s12079-017-0421-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022] Open
Abstract
Hepatocyte exosomes (ExoHep) are proposed to mediate physiological or pathophysiological signaling in a variety of hepatic target cells. ExoHep were purified from the medium of primary mouse hepatocytes or AML12 cells and characterized as ~100 nm nanovesicles that were positive for proteins commonly found in exosomes (CD9, CD81, flotillin) or hepatocytes (asialoglycoprotein receptor). Ethanol treatment of hepatocytes caused increased ExoHep release and increased cellular mRNA expression of components involved in intracellular vesicle trafficking (Rab 5a,b,c, Rab 7a, Rab 27a,b) or exosome biogenesis via the ESCRT (HGS, Alix, STAM1, TSG101, VTA1, YKT6) or ceramide (nSmase2) pathways. RNA interference of HGS, Alix, TSG101 or nSmase 2 caused exosome production by normal or ethanol-treated hepatocytes to be reduced. In mice, in vivo administration of fluorescently-labeled ExoHep resulted in their accumulation in the liver and preferential localization to hepatic stellate cells (HSC) or hepatocytes, the latter of which showed enhanced ExoHep binding when isolated from fibrotic mice. In cell co-cultures, the intercellular transfer of RNA from hepatocytes to hepatocytes or HSC was blocked by the exosome inhibitor GW4869. ExoHep binding to HSC or hepatocytes occurred via mechanisms that involved heparin-like molecules and cellular integrin αv or β1 subunits , and resulted in a reversal of fibrosis-associated gene expression in HSC and of ethanol-induced damage in hepatocytes. These studies provide insight regarding the regulation and/or participation of exosome biogenesis or trafficking components in hepatocytes and show that ExoHep can mediate therapeutic changes in activated HSC or injured hepatocytes that occur downstream of heparin- or integrin-dependent binding interactions.
Collapse
Affiliation(s)
- Li Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Ruju Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Sherri Kemper
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - David R Brigstock
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA. .,Department of Surgery, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
250
|
Ethanol Induces Enhanced Vascularization Bioactivity of Endothelial Cell-Derived Extracellular Vesicles via Regulation of MicroRNAs and Long Non-Coding RNAs. Sci Rep 2017; 7:13794. [PMID: 29062004 PMCID: PMC5653762 DOI: 10.1038/s41598-017-14356-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/10/2017] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs), such as exosomes, have been identified as regulators of vascular remodeling and have promise as therapeutics for vascularization applications. Towards development of EVs as therapeutics, it has been demonstrated that physiological stimuli of angiogenic phenotypes in EV-producing cells can enhance the potency of EVs for vascularization. The goal of this study was to assess whether ethanol, which induces angiogenic phenotypes in endothelial cells, could be employed to enhance endothelial-derived EV vascularization bioactivity. The results indicate that ethanol conditioning of endothelial cells increases the ability of endothelial EVs to induce a pro-vascularization response. This response is due in part to increased CD34 expression in recipient endothelial cells that may result from downregulation of microRNA-106b in EVs isolated from ethanol-conditioned producer endothelial cells. Further, ethanol-induced upregulation of long non-coding RNAs (lncRNAs) HOTAIR and MALAT1 in endothelial EVs was observed to play a significant role in mediating pro-angiogenic effects of these vesicles. Overall, these studies validate ethanol conditioning as a method to enhance the bioactivity of endothelial EVs via regulation of EV-associated microRNAs (miRNAs) and, especially, lncRNAs. Further, the results suggest that alcohol consumption may activate endothelial EVs towards a pro-vascularization phenotype, which could have implications for alcohol-induced tumor angiogenesis.
Collapse
|