1
|
Katana Z, Sianidou K, Kaiopoulos G, Deligianni F, Tsetsakos S, Kouvatsi A, Sakellari I, Kritis A, Touraki M, Sotiropoulos D, Xagorari A. Molecular and biochemical evaluation of oxidative effects of cord blood CD34+ MPs on hematopoietic cells. Blood Cells Mol Dis 2024; 108:102871. [PMID: 39013336 DOI: 10.1016/j.bcmd.2024.102871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
A graft source for allogeneic hematopoietic stem cell transplantation is umbilical cord blood, which contains umbilical cord blood mononuclear cells (MNCs and mesenchymal stem cells, both an excellent source of extracellular microparticles (MPs). MPs act as cell communication mediators, which are implicated in reactive oxygen species formation or detoxification depending on their origin. Oxidative stress plays a crucial role in both the development of cancer and its treatment by triggering apoptotic mechanisms, in which CD34+ cells are implicated. The aim of this work is to investigate the oxidative stress status and the apoptosis of HL-60 and mononuclear cells isolated from umbilical cord blood (UCB) following a 24- and 48-hour exposure to CD34 + microparticles (CD34 + MPs). The activity of superoxide dismutase, glutathione reductase, and glutathione S-transferase, as well as lipid peroxidation in the cells, were employed as oxidative stress markers. A 24- and 48-hour exposure of leukemic and mononuclear cells to CD34 + -MPs resulted in a statistically significant increase in the antioxidant activity and lipid peroxidation in both cells types. Moreover, CD34 + MPs affect the expression of BCL2 and FAS and related proteins and downregulate the hematopoietic differentiation program in both HL-60 and mononuclear cells. Our results indicate that MPs through activation of antioxidant enzymes in both homozygous and nonhomozygous cells might serve as a means for graft optimization and enhancement.
Collapse
Affiliation(s)
- Zoi Katana
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriaki Sianidou
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gregory Kaiopoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fani Deligianni
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sarantis Tsetsakos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Anastasia Kouvatsi
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Sakellari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Aristeidis Kritis
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Touraki
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Damianos Sotiropoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Angeliki Xagorari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece.
| |
Collapse
|
2
|
Santiago VF, Rosa-Fernandes L, Macedo-da-Silva J, Angeli CB, Mule SN, Marinho CRF, Torrecilhas AC, Marie SNK, Palmisano G. Isolation of Extracellular Vesicles Using Titanium Dioxide Microspheres. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:1-22. [PMID: 38409413 DOI: 10.1007/978-3-031-50624-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Extracellular vesicles (EVs) are bilayer membrane particles released from several cell types to the extracellular environment. EVs have a crucial role in cell-cell communication, involving different biological processes in health and diseases. Due to the potential of biomarkers for several diseases as diagnostic and therapeutic tools, it is relevant to understand the biology of the EVs and their content. One of the current challenges involving EVs is regarding the purification method, which is a critical step for EV's functional and characterization studies. Ultracentrifugation is the most used method for EV isolation, where the nanoparticles are separated in sequential centrifugation to isolate the EVs based on their size. However, for viscous biofluids such as plasma, there is a co-isolation of the most abundant proteins, which can impair the EV's protein identification due to the low abundance of these proteins and signal suppression by the most abundant plasma proteins. Emerging techniques have gained attention in recent years. Titanium dioxide (TiO2) is one of the most promising techniques due to its property for selective isolation based on the interaction with phospholipids in the EV membrane. Using a small amount of TiO2 beads and a low volume of plasma, it is possible to isolate EVs with reduced plasma protein co-isolation. This study describes a comprehensive workflow for the isolation and characterization of plasma extracellular vesicles (EVs) using mass spectrometry-based proteomics techniques. The aim of this chapter is describe the EV isolation using TiO2 beads enrichment and high-throughput mass spectrometry techniques to efficiently identify the protein composition of EVs in a fast and straightforward manner.
Collapse
Affiliation(s)
- Veronica Feijoli Santiago
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Livia Rosa-Fernandes
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Janaina Macedo-da-Silva
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Claudia B Angeli
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Simon Ngao Mule
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Claudio R F Marinho
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Torrecilhas
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas; Departamento de Ciências Farmacêuticas; Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários. Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Suely N K Marie
- Laboratory of Molecular and Cellular Biology (LIM15), Department of Neurology, Fac-uldade de Medicina FMUSP, University of São Paulo, São Paulo, Brazil
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, Australia.
| |
Collapse
|
3
|
Borsellino B, Bravo-Perez C, Visconte V, Guarnera L. Thrombosis in Myeloid Malignancies: From CHIP to AML. Cardiovasc Hematol Disord Drug Targets 2024; 24:2-12. [PMID: 38879768 DOI: 10.2174/011871529x307253240530060107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 09/04/2024]
Abstract
The development of myeloid malignancies is a multi-step process starting from pre-malignant stages. Large-scale studies on clonal hematopoiesis of indeterminate potential (CHIP) identified this condition as a risk factor for developing hematologic malignancies, in particular myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). In parallel, CHIP was found to confer an enhanced thrombotic risk, in particular for cardiovascular diseases. In a similar fashion, in recent years, alongside their life-threatening features, increasing attention has been drawn toward thrombotic complications in myeloid malignancies. Thus, the purpose of this review is to gather a growing body of evidence on incidence, pathogenesis and clinical impact of thrombosis in myeloid malignancies at every step of malignant progression, from CHIP to AML.
Collapse
Affiliation(s)
- Beatrice Borsellino
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Carlos Bravo-Perez
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH44195, USA
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER-Instituto de Salud Carlos III, 30005, Murcia, Spain
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH44195, USA
| | - Luca Guarnera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH44195, USA
| |
Collapse
|
4
|
Ge Y, Ye T, Fu S, Jiang X, Song H, Liu B, Wang G, Wang J. Research progress of extracellular vesicles as biomarkers in immunotherapy for non-small cell lung cancer. Front Immunol 2023; 14:1114041. [PMID: 37153619 PMCID: PMC10162406 DOI: 10.3389/fimmu.2023.1114041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/07/2023] [Indexed: 05/10/2023] Open
Abstract
Lung cancer is one of the most severe forms of malignancy and a leading cause of cancer-related death worldwide, of which non-small cell lung cancer (NSCLC) is the most primary type observed in the clinic. NSCLC is mainly treated with surgery, radiotherapy, and chemotherapy. Additionally, targeted therapy and immunotherapy have also shown promising results. Several immunotherapies, including immune checkpoint inhibitors, have been developed for clinical use and have benefited patients with NSCLC. However, immunotherapy faces several challenges like poor response and unknown effective population. It is essential to identify novel predictive markers to further advance precision immunotherapy for NSCLC. Extracellular vesicles (EVs) present an important research direction. In this review, we focus on the role of EVs as a biomarker in NSCLC immunotherapy considering various perspectives, including the definition and properties of EVs, their role as biomarkers in current NSCLC immunotherapy, and different EV components as biomarkers in NSCLC immunotherapy research. We describe the cross-talk between the role of EVs as biomarkers and novel technical approaches or research concepts in NSCLC immunotherapy, such as neoadjuvants, multi-omics analysis, and the tumour microenvironment. This review will provide a reference for future research to improve the benefits of immunotherapy for patients with NSCLC.
Collapse
Affiliation(s)
- Yang Ge
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Ting Ye
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Siyun Fu
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoying Jiang
- Department of Science and Technology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Bin Liu
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Bin Liu, ; Guoquan Wang, ; Jinghui Wang,
| | - Guoquan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- *Correspondence: Bin Liu, ; Guoquan Wang, ; Jinghui Wang,
| | - Jinghui Wang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Bin Liu, ; Guoquan Wang, ; Jinghui Wang,
| |
Collapse
|
5
|
Krayem B, Brenner B, Horowitz NA. Thrombosis in Pregnant Women with Hematological Malignancies: A Case-Based Review. Semin Thromb Hemost 2022; 49:348-354. [PMID: 36535649 DOI: 10.1055/s-0042-1759683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AbstractCancer and pregnancy induce a procoagulant environment which may lead to maternal and fetal complications, such as venous thromboembolism, fetal growth restriction, and fetal loss. The incidence of hematological malignancies diagnosed during pregnancy is rising, and thrombotic events in such malignancies are not rare. Management of thrombosis during pregnancy poses a therapeutic challenge, that is further exacerbated by the impact of cancer. The available data on managing pregnant women with hematological malignancies are limited to those with myeloproliferative neoplasms, mainly essential thrombocythemia, and, to a lesser extent, polycythemia vera. Low-dose aspirin is recommended throughout pregnancy, and considering treatment with low-molecular-weight heparin and interferon formulations is advised for high-risk patients. Currently, guidelines for handling thrombotic events in pregnant women with lymphoma or leukemia are lacking, and their management is based on data extrapolated from retrospective studies, and guidelines for prevention and treatment of cancer-associated thrombosis. The present case-based review will focus on the complex issue of thrombotic risk in pregnant women with hematological malignancies, specifically myeloproliferative neoplasms, lymphomas, and leukemias.
Collapse
Affiliation(s)
- Baher Krayem
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Netanel A. Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
6
|
Tawil N, Rak J. Blood coagulation and cancer genes. Best Pract Res Clin Haematol 2022; 35:101349. [DOI: 10.1016/j.beha.2022.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
|
7
|
Tian Z, Liang G, Cui K, Liang Y, Wang Q, Lv S, Cheng X, Zhang L. Insight Into the Prospects for RNAi Therapy of Cancer. Front Pharmacol 2021; 12:644718. [PMID: 33796026 PMCID: PMC8007863 DOI: 10.3389/fphar.2021.644718] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
RNA interference (RNAi), also known as gene silencing, is a biological process that prevents gene expression in certain diseases such as cancer. It can be used to improve the accuracy, efficiency, and stability of treatments, particularly genetic therapies. However, challenges such as delivery of oligonucleotide drug to less accessible parts of the body and the high incidence of toxic side effects are encountered. It is therefore imperative to improve their delivery to target sites and reduce their harmful effects on noncancerous cells to harness their full potential. In this study, the role of RNAi in the treatment of COVID-19, the novel coronavirus disease plaguing many countries, has been discussed. This review aims to ascertain the mechanism and application of RNAi and explore the current challenges of RNAi therapy by identifying some of the cancer delivery systems and providing drug information for their improvement. It is worth mentioning that delivery systems such as lipid-based delivery systems and exosomes have revolutionized RNAi therapy by reducing their immunogenicity and improving their cellular affinity. A deeper understanding of the mechanism and challenges associated with RNAi in cancer therapy can provide new insights into RNAi drug development.
Collapse
Affiliation(s)
- Zhili Tian
- Institute of Molecular Medicine, Henan University, Kaifeng, China.,School of Clinical Medical Sciences, Henan University, Kaifeng, China
| | - Guohui Liang
- Institute of Molecular Medicine, Henan University, Kaifeng, China.,School of Clinical Medical Sciences, Henan University, Kaifeng, China
| | - Kunli Cui
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yayu Liang
- Institute of Molecular Medicine, Henan University, Kaifeng, China.,School of Stomatology, Henan University, Kaifeng, China
| | - Qun Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuangyu Lv
- Institute of Molecular Medicine, Henan University, Kaifeng, China
| | - Xiaoxia Cheng
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
8
|
Nazari M, Javandoost E, Talebi M, Movassaghpour A, Soleimani M. Platelet Microparticle Controversial Role in Cancer. Adv Pharm Bull 2020; 11:39-55. [PMID: 33747851 PMCID: PMC7961228 DOI: 10.34172/apb.2021.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are a group of micrometer-scale extracellular vesicles released by platelets upon activation that are responsible for the majority of microvesicles found in plasma. PMPs’ physiological properties and functions have long been investigated by researchers. In this regard, a noticeable area of studies has been devoted to evaluating the potential roles and effects of PMPs on cancer progression. Clinical and experimental evidence conflictingly implicates supportive and suppressive functions for PMPs regarding cancer. Many of these functions could be deemed as a cornerstone for future considerations of PMPs usage in cancer targeted therapy. This review discusses what is currently known about PMPs and provides insights for new and possible research directions for further grasping the intricate interplay between PMPs and cancer.
Collapse
Affiliation(s)
- Mahnaz Nazari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Javandoost
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. Introduction
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
9
|
Bar-Sela G, Cohen I, Avisar A, Loven D, Aharon A. Circulating blood extracellular vesicles as a tool to assess endothelial injury and chemotherapy toxicity in adjuvant cancer patients. PLoS One 2020; 15:e0240994. [PMID: 33108394 PMCID: PMC7591065 DOI: 10.1371/journal.pone.0240994] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 10/06/2020] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) are subcellular membrane blebs that include exosomes and microparticles, which represent a potential source for cancer biomarker discovery. We assess EVs characteristics as a tool to evaluate the endothelial and anti-tumor treatment injury during adjuvant chemotherapy in breast (BC) and colon cancer (CC) patients. Blood samples were taken from 29 BC and 25 CC patients before and after chemotherapy, as well as from healthy control donors (HC). Circulating blood EVs were isolated and characterized by size/concentration, membrane antigens for cell origin, thrombogenicity, and protein content. We observed higher EVs concentration and particle size in CC patients after chemotherapy compared with HC. Higher levels of endothelial EVs (CD144-positive) and vascular endothelial growth factor receptor 1 (VEGFR1), apparently as an indication of endothelial dysfunction, were found in all cancer patients, regardless of a given treatment, compared to HC. Levels of EVs labeled CD62E, CD34+41-, the lymphocyte markers CD11+ and CD-14+, Annexin-V, and the coagulation proteins TF and TFPI, however, sometimes demonstrate significant differences between patients, although HC did not show significant differences between patients pre- and post-chemotherapy. Most importantly, increasing levels of EVs encapsulated Angiostatin were found in patients with CC, while chemotherapy treatment leads to its notable rise in circulating blood EVs. Our results demonstrate the potential of EVs encapsulated Angiostatin as a tool to evaluate endothelial damage during adjuvant chemotherapy in BC and CC patients.
Collapse
Affiliation(s)
- Gil Bar-Sela
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Cancer Center, Emek Medical Center, Afula, Israel
| | - Idan Cohen
- Cancer Center, Emek Medical Center, Afula, Israel
| | | | - David Loven
- Cancer Center, Emek Medical Center, Afula, Israel
| | - Anat Aharon
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Hematology and Bone Marrow Transplantation, Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
10
|
Abstract
Exosomes are membrane-bound cargo measuring 30–140 nm comprised of a lipid bilayer containing various proteins, RNAs, DNAs, and bioactive lipids that can be transferred between cells. They have been shown to be produced and released by many different types of healthy and diseased cells. Exosomes are secreted by all types of cells in culture, and are also found in various body fluids including blood, saliva, urine, and breast milk. Exosomes are essential for healthy physiological as well as pathological processes. In addition to their normal function, exosomes are involved in the development and progression of various diseases, potentiating cellular stress and damage. Pathogens take advantage of exosome release from infected host cells by manipulating host-derived exosomes to evade the immune system responses. Exosomes are involved in other pathological conditions such as neurodegenerative diseases, liver diseases, heart failure, cancer, diabetes, kidney diseases, osteoporosis and atherosclerotic cardiovascular disease. Hence, we can exploit exosomes as biomarkers and vaccines and modify them rationally for therapeutic interventions including tissue engineering. Further studies on exosomes will explore their potential and provide new methodology for effective clinical diagnostics and therapeutic strategies: such uses can be called exosome theragnostics. This chapter reviews the potential theragnostic (diagnostic and therapeutic) application of exosomes in major organ systems in clinical fields.
Collapse
|
11
|
Hata T, Yasui M, Ikeda M, Miyake M, Ide Y, Okuyama M, Ikenaga M, Kitani K, Morita S, Matsuda C, Mizushima T, Yamamoto H, Murata K, Sekimoto M, Nezu R, Mori M, Doki Y. Efficacy and safety of anticoagulant prophylaxis for prevention of postoperative venous thromboembolism in Japanese patients undergoing laparoscopic colorectal cancer surgery. Ann Gastroenterol Surg 2019; 3:568-575. [PMID: 31549017 PMCID: PMC6749951 DOI: 10.1002/ags3.12279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 01/24/2023] Open
Abstract
AIM To investigate the efficacy and safety of anticoagulant prophylaxis to prevent postoperative venous thromboembolism (VTE) during laparoscopic colorectal cancer (CRC) surgery, which is unknown in Japanese patients. METHODS We conducted this randomized controlled trial at nine institutions in Japan from 2011 to 2015. It included 302 eligible patients aged 20 years or older who underwent elective laparoscopic surgery for CRC. Patients were randomly assigned to an intermittent pneumatic compression (IPC) therapy group or to an IPC + anticoagulation therapy group. Anticoagulation therapy comprised fondaparinux or enoxaparin for postoperative VTE prophylaxis. Postoperative VTE was diagnosed based on enhanced multi-detector helical computed tomography. The primary endpoint was VTE incidence, including asymptomatic cases, the secondary endpoint was incidence of major bleeding, and we conducted an intention-to-treat analysis. This study is registered in UMINCTR (UMIN000008435). RESULTS Postoperative VTE incidence was 5.10% with IPC therapy (n = 157) and 2.76% with IPC + anticoagulant therapy (n = 145; P = .293). We identified no symptomatic VTE cases. The major bleeding rates were 1.27% with IPC alone and 1.38% with the combination (P = .936). The overall bleeding rates were 7.69% for enoxaparin and 13.6% for fondaparinux (P = .500), and there were no bleeding-related deaths. CONCLUSION Anticoagulant prophylaxis did not reduce the incidence of VTE and the incidence of major bleeding was comparable between the two groups. Usefulness of perioperative anticoagulation was not demonstrated in this study. Pharmacological prophylaxis must be restricted in Japanese patients with higher risk of VTE.
Collapse
Affiliation(s)
- Taishi Hata
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
| | - Masayoshi Yasui
- Department of Gastroenterological SurgeryOsaka International Cancer InstituteOsaka-city, OsakaJapan
| | - Masataka Ikeda
- Department of SurgeryHyogo College of MedicineNishinomiya-city, HyogoJapan
| | - Masakazu Miyake
- Department of SurgeryOsaka General Medical CenterOsaka-city, OsakaJapan
| | - Yoshihito Ide
- Department of SurgeryYao Municipal HospitalYao-city, OsakaJapan
| | - Masaki Okuyama
- Department of surgeryKaizuka City HospitalKaizuka-city, OsakaJapan
| | - Masakazu Ikenaga
- Department of Gastroenterological surgeryHigashiosaka City Medical CenterHigashiosaka-city, OsakaJapan
| | - Kotaro Kitani
- Department of Gastroenterological SurgeryKindai University Nara HospitalIkoma-city, NaraJapan
| | - Shunji Morita
- Department of Gastroenterological SurgeryToyonaka Municipal HospitalToyonaka-city,OsakaJapan
| | - Chu Matsuda
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
| | - Tsunekazu Mizushima
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
- Department of Therapeutics for Inflammatory Bowel DiseasesGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
| | - Hirofumi Yamamoto
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
- Department of Molecular PathologyDivision of Health SciencesGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
| | - Kohei Murata
- Department of SurgeryKansai Rosai HospitalAmagasaki-city, HyogoJapan
| | - Mitsugu Sekimoto
- Department of SurgeryOsaka General Medical CenterOsaka-city, OsakaJapan
| | - Riichiro Nezu
- Department of surgeryNishinomiya Municipal Central HospitalNishinomiya-city, HyogoJapan
| | - Masaki Mori
- Department of Surgery and ScienceGraduate School of Medical SciencesKyushu UniversityFukuoka-city, FukuokaJapan
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuita-city, OsakaJapan
| | | |
Collapse
|
12
|
Zannoni J, Mauz N, Seyve L, Meunier M, Pernet-Gallay K, Brault J, Jouzier C, Laurin D, Pezet M, Pernollet M, Cahn JY, Cognasse F, Polack B, Park S. Tumor microenvironment and clonal monocytes from chronic myelomonocytic leukemia induce a procoagulant climate. Blood Adv 2019; 3:1868-1880. [PMID: 31221660 PMCID: PMC6595258 DOI: 10.1182/bloodadvances.2018026955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/14/2019] [Indexed: 01/22/2023] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is a myeloid hematological malignancy with overlapping features of myelodysplastic syndromes (MDSs) and myeloproliferative neoplasms (MPNs). The knowledge of the role of the tumor microenvironment (TME), particularly mesenchymal stromal cells (MSCs), in MDS pathogenesis is increasing. Generally, cancer is associated with a procoagulant state participating in tumor development. Monocytes release procoagulant, tissue factor (TF)-bearing microparticles. We hypothesized that MSCs and clonal monocytes release procoagulant extracellular vesicles (EVs) within the CMML TME, inducing a procoagulant state that could modify hematopoietic stem cell (HSC) homeostasis. We isolated and cultured MSCs and monocytes from CMML patients and MSCs from healthy donors (HDs). Their medium EVs and small EVs (sEVs) were collected after iterative ultracentrifugations and characterized by nanoparticle tracking analysis. Their impact on hemostasis was studied with a thrombin generation assay and fibrinography. CMML or HD HSCs were exposed to sEVs from either CMML or HD MSCs. CMML MSC sEVs increased HD HSC procoagulant activity, suggesting a transfer of TF from the CMML TME to HD HSCs. The presence of TF on sEVs was shown by electron microscopy and western blot. Moreover, CMML monocyte EVs conferred a procoagulant activity to HD MSCs, which was reversed by an anti-TF antibody, suggesting the presence of TF on the EVs. Our findings revealed a procoagulant "climate" within the CMML environment related to TF-bearing sEVs secreted by CMML MSCs and monocytes.
Collapse
Affiliation(s)
- Johanna Zannoni
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
| | - Natacha Mauz
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Landry Seyve
- Techniques de l'Ingénierie Médicale et de la Complexité Informatique, Mathématiques et Applications-Thérapeutique Recombinante Expérimentale, UMR 5525 Centre National de la Recherche Scientifique, Grenoble Alpes University, Grenoble, France
- Laboratory of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Mathieu Meunier
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Karin Pernet-Gallay
- Grenoble Institute for Neurosciences, INSERM U1216, Plateforme de Microscopie Electronique, Grenoble, France
| | - Julie Brault
- Techniques de l'Ingénierie Médicale et de la Complexité Informatique, Mathématiques et Applications-Thérapeutique Recombinante Expérimentale, UMR 5525 Centre National de la Recherche Scientifique, Grenoble Alpes University, Grenoble, France
- Centre de Diagnostic de la Granulomatose Septique Diagnosis and Research Center, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Claire Jouzier
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - David Laurin
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Etablissement Français du Sang Rhône-Alpes-Auvergne, Grenoble, France
| | - Mylène Pezet
- Plateforme de Microscopie Photonique, Cytométrie en Flux, Institute for Advanced Biosciences, Grenoble, France
| | - Martine Pernollet
- Institut de Biologie et de Pathologie, Laboratoire d'Immunologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Jean-Yves Cahn
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Fabrice Cognasse
- Etablissement Français du Sang Rhône-Alpes-Auvergne, Saint-Etienne, France; and
- GIMAP-EA3064, Lyon University, Saint-Etienne, France
| | - Benoît Polack
- Techniques de l'Ingénierie Médicale et de la Complexité Informatique, Mathématiques et Applications-Thérapeutique Recombinante Expérimentale, UMR 5525 Centre National de la Recherche Scientifique, Grenoble Alpes University, Grenoble, France
- Laboratory of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Sophie Park
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| |
Collapse
|
13
|
Haen P, Mege D, Crescence L, Dignat-George F, Dubois C, Panicot-Dubois L. Thrombosis Risk Associated with Head and Neck Cancer: A Review. Int J Mol Sci 2019; 20:E2838. [PMID: 31212608 PMCID: PMC6600456 DOI: 10.3390/ijms20112838] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/30/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022] Open
Abstract
Venous thromboembolism (VTE) is a common complication for cancer patients. VTE-associated risk varies according to the type of tumor disease. Head and neck cancer is a common cancer worldwide, and most tumors are squamous cell carcinomas due to tobacco and alcohol abuse. The risk of VTE associated with head and neck (H&N) cancer is considered empirically low, but despite the high incidence of H&N cancer, few data are available on this cancer; thus, it is difficult to state the risk of VTE. Our review aims to clarify this situation and tries to assess the real VTE risk associated with H&N cancer. We report that most clinical studies have concluded that there is a very low thrombosis risk associated with H&N cancer. Even with the biases that often exist, this clinical review seems to confirm that the risk of VTE was empirically hypothesized. Furthermore, we highlight that H&N cancer has all the biological features of a cancer associated with a high thrombosis risk, including a strong expression of procoagulant proteins, modified thrombosis/fibrinolysis mechanisms, and secretions of procoagulant microparticles and procoagulant cytokines. Thus, this is a paradoxical situation, and some undiscovered mechanisms that could explain this clinical biological ambivalence might exist.
Collapse
Affiliation(s)
- Pierre Haen
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
- Department of Maxillo-Facial Surgery, Army Training Hospital, Laveran, 13013 Marseille, France.
| | - Diane Mege
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
- Department of Digestive Surgery, Timone University Hospital, AP-HM, 13005 Marseille, France.
| | - Lydie Crescence
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
- Laboratoire d'Hématologie, Centre Hospitalo-Universitaire Conception, 385 Boulevard Baille, 13385 Marseille, France.
| | - Christophe Dubois
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
| | - Laurence Panicot-Dubois
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
| |
Collapse
|
14
|
Sierko E, Sobierska M, Zabrocka E, Myśliwiec M, Kruszewska J, Lipska A, Radziwon P, Wojtukiewicz MZ. Endothelial Microparticles and Blood Coagulation Activation in Head and Neck Cancer Patients Undergoing Radiotherapy or Radiochemotherapy. In Vivo 2019; 33:627-632. [PMID: 30804151 DOI: 10.21873/invivo.11520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIM Endothelial microparticles (EMP) are small vesicles which are released from the endothelium and contribute to blood coagulation activation in various clinical settings. The aim of this study was to examine whether EMP influence blood coagulation activation in cancer patients during radiotherapy/radiochemotherapy (RT/RCT). MATERIALS AND METHODS Sixteen head and neck cancer (HNC) patients undergoing RT/RCT and 10 controls were examined. EMP and thrombin-antithrombin complex (TAT) were measured by flow cytometry and enzyme-linked immunosorbent assay (ELISA), respectively. Tissue factor-positive EMP (TF+EMP) were defined as CD31+/CD142+/CD42b- Results: TF+EMP were significantly elevated in HNC patients before RT/RCT (T0) (1299±1154/μl), one day after RT/RCT (T1d) (1257±603/μl) and 3 months after RT/RCT (T3m) (1289±372/μl) compared to controls (688±647/μl). TF+EMP levels at T0/T1d and T0, as well as at T1d and T3m were not significantly different. TAT levels at T0 and T1d did not differ significantly but at T3m were significantly lower compared to T0 and T1d TF+EMP and TAT concentrations were not significantly correlated at T0 (r=0.058; p=0.828), T1d (r=0.373, p=0.154) and T3m (r=-0.302, p=0.204). CONCLUSION TF+EMP may not contribute to hemostatic abnormalities in HNC patients.
Collapse
Affiliation(s)
- Ewa Sierko
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland .,Department of Radiotherapy, Comprehensive Cancer Center, Bialystok, Poland
| | - Monika Sobierska
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland.,Regional Centre for Transfusion Medicine, Bialystok, Poland
| | - Ewa Zabrocka
- Department of Medicine, Stony Brook University, Stony Brook, NY, U.S.A
| | - Marta Myśliwiec
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Kruszewska
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Alina Lipska
- Regional Centre for Transfusion Medicine, Bialystok, Poland
| | - Piotr Radziwon
- Regional Centre for Transfusion Medicine, Bialystok, Poland
| | | |
Collapse
|
15
|
Analysis of the thrombotic and fibrinolytic activities of tumor cell-derived extracellular vesicles. Blood Adv 2019; 2:1054-1065. [PMID: 29752284 DOI: 10.1182/bloodadvances.2017015479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/04/2018] [Indexed: 02/06/2023] Open
Abstract
Exosomes and microvesicles (MVs) are small extracellular vesicles secreted by tumor cells and are suggested to contribute to the thrombotic events that commonly occur in patients with advanced malignancies. Paradoxically, these vesicles have been reported to also possess fibrinolytic activity. To determine whether thrombotic or fibrinolytic activity is a predominant characteristic of these extracellular vesicles, we prepared exosomes and MVs from 2 breast cancer cell lines (MDA-MB-231 and MCF7), a lung cancer cell line (A549), and a leukemia cell line (NB4) and assayed their thrombotic and fibrinolytic activities. We observed that thrombotic activity was a common feature of MVs but not exosomes. Exosomes and/or MVs from several cell lines, with the exception of the A549 cell line, displayed fibrinolytic activity toward a pure fibrin clot, but only exosomes from MDA-MB-231 cells could degrade a fibrin clot formed in plasma. Increasing the malignant potential of MCF7 cells decreased the thrombotic activity of their MVs but did not alter their fibrinolytic activity. Decreasing the malignant potential of NB4 cells did not alter the thrombotic or fibrinolytic activity of their MVs or exosomes. Finally, the incubation of MDA-MB-231 cell-derived exosomes with A549 cells increased plasmin generation by these cells. Our data indicate that MVs generally have thrombotic activity, whereas thrombotic activity is not commonly observed for exosomes. Furthermore, exosomes and MVs generally do not display fibrinolytic activity under physiological conditions.
Collapse
|
16
|
Zhong WQ, Ren JG, Xiong XP, Man QW, Zhang W, Gao L, Li C, Liu B, Sun ZJ, Jia J, Zhang WF, Zhao YF, Chen G. Increased salivary microvesicles are associated with the prognosis of patients with oral squamous cell carcinoma. J Cell Mol Med 2019; 23:4054-4062. [PMID: 30907490 PMCID: PMC6533497 DOI: 10.1111/jcmm.14291] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 12/17/2018] [Accepted: 03/08/2019] [Indexed: 01/08/2023] Open
Abstract
Microvesicles (MVs), which are cell-derived membrane vesicles present in body fluids, are closely associated with the development of malignant tumours. Saliva, one of the most versatile body fluids, is an important source of MVs. However, the association between salivary MVs (SMVs) and oral squamous cell carcinoma (OSCC), which is directly immersed in the salivary milieu, remains unclear. SMVs from 65 patients with OSCC, 21 patients with oral ulcer (OU), and 42 healthy donors were purified, quantified and analysed for their correlations with the clinicopathologic features and prognosis of OSCC patients. The results showed that the level of SMVs was significantly elevated in patients with OSCC compared to healthy donors and OU patients. Meanwhile, the level of SMVs showed close correlations with the lymph node status, and the clinical stage of OSCC patients. Additionally, the ratio of apoptotic to non-apoptotic SMVs was significantly decreased in OSCC patients with higher pathological grade. Consistently, poorer overall survival was observed in patients with lower ratio of apoptotic to non-apoptotic SMVs. In conclusion, the elevated level of SMVs is associated with clinicopathologic features and decreased survival in patients with OSCC, suggesting that SMVs are a potential biomarker and/or regulator of the malignant progression of OSCC.
Collapse
Affiliation(s)
- Wen-Qun Zhong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jian-Gang Ren
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xue-Peng Xiong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qi-Wen Man
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chen Li
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wen-Feng Zhang
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Fang Zhao
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Sierko E, Sobierska M, Zabrocka E, Kruszewska J, Myśliwiec M, Lipska A, Radziwon P, Wojtukiewicz MZ. Endothelial Microparticles and Vascular Endothelial Growth Factor in Patients With Head and Neck Cancer Undergoing Radiotherapy or Radiochemotherapy. In Vivo 2019; 33:581-586. [PMID: 30804145 DOI: 10.21873/invivo.11514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/29/2018] [Accepted: 01/10/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Endothelial microparticles (EMPs) released from activated or apoptotic endothelial cells may play a role in coagulation and thrombus formation. However, there is insufficient evidence regarding the impact of EMPs on angiogenesis in patients with cancer. MATERIALS AND METHODS Sixteen patients with head and neck cancer (HNC) undergoing radiotherapy/radiochemotherapy (RT/RCT) and 10 healthy controls were studied. Serum EMPs were counted by flow cytometry, and vascular endothelial growth factor (VEGF) was measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The mean EMP level was significantly higher in patients with HNC before RT/RCT (1,601±1,479 EMP/μl) compared to the control group (782±698 EMP/μl). The number of EMPs was not notably increased after RT/RCT (1,629±769 EMP/μl). There was no significant correlation between the plasma EMP number and concentration of VEGF before (r=0.131; p=0.625), 1 day after (r=-0.042, p=0.874), nor 3 months after RT/RCT (r=0.454, p=0.076). CONCLUSION Released EMPs may not influence promotion of neovascularization in patients with HNC.
Collapse
Affiliation(s)
- Ewa Sierko
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland .,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Monika Sobierska
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland.,Regional Centre for Transfusion Medicine, Bialystok, Poland
| | - Ewa Zabrocka
- Department of Medicine, Stony Brook University, Stony Brook, NY, U.S.A
| | - Joanna Kruszewska
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Marta Myśliwiec
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Alina Lipska
- Regional Centre for Transfusion Medicine, Bialystok, Poland
| | - Piotr Radziwon
- Regional Centre for Transfusion Medicine, Bialystok, Poland
| | | |
Collapse
|
18
|
Tawil N, Chennakrishnaiah S, Bassawon R, Johnson R, D'Asti E, Rak J. Single cell coagulomes as constituents of the oncogene-driven coagulant phenotype in brain tumours. Thromb Res 2018; 164 Suppl 1:S136-S142. [PMID: 29703472 DOI: 10.1016/j.thromres.2018.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
Abstract
Molecular profiling of human cancers revealed a startling diversity in disease-causing mechanisms superseding histological and anatomical commonalities. The emerging molecular subtypes and disease entities are often driven by distinct oncogenic pathways and their effectors, including those acting extracellularly on the vascular and coagulation systems. Indeed, several oncogenic mutations such as those affecting protein-coding genes (RAS, EGFR, PTEN, TP53) and non-coding RNA (microRNA) regulate multiple effectors of the coagulation system (coagulome), including tissue factor, protease activated receptors, clotting factors, mediators of platelet function and fibrinolysis. This is exemplified by differential coagulome profiles in the molecular subtypes of glioblastoma, medulloblastoma and other human tumours. There is mounting clinical evidence that the mutational status of cancer driver genes such as KRAS or IDH1 may influence the risk of venous thromboembolism in patients with colorectal, lung or brain cancers. Notably, single cell sequencing in glioblastoma revealed a remarkable intra-tumoural heterogeneity of cancer cell populations with regard to their individual coagulomes, suggesting a combinatorial and dynamic nature of the global pro-thrombotic phenotype. We suggest that the cellular complexity of specific cancers may define their mechanisms of interactions with the coagulation system, and the risks of thrombosis. Thus, more biologically- based, disease-specific and personalized approaches may be needed to diagnose and manage cancer-related thrombosis.
Collapse
Affiliation(s)
- Nadim Tawil
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | | | - Rayhaan Bassawon
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Radia Johnson
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Esterina D'Asti
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Janusz Rak
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada.
| |
Collapse
|
19
|
Chennakrishnaiah S, Meehan B, D'Asti E, Montermini L, Lee TH, Karatzas N, Buchanan M, Tawil N, Choi D, Divangahi M, Basik M, Rak J. Leukocytes as a reservoir of circulating oncogenic DNA and regulatory targets of tumor-derived extracellular vesicles. J Thromb Haemost 2018; 16:1800-1813. [PMID: 29971917 DOI: 10.1111/jth.14222] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Indexed: 12/11/2022]
Abstract
Essentials Tumor-bearing mice were employed to follow oncogenic HRAS sequences in plasma, and blood cells. Cancer DNA accumulated in leukocytes above levels detected in exosomes, platelets and plasma. Extracellular vesicles and nucleosomes are required for uptake of tumor DNA by leukocytes. Uptake of tumor-derived extracellular vesicles by leukocytes triggers coagulant phenotype. SUMMARY Background Tumor-derived extracellular vesicles (EVs) and free nucleosomes (NSs) carry into the circulation a wealth of cancer-specific, bioactive and poorly understood molecular cargoes, including genomic DNA (gDNA). Objective Here we investigated the distribution of extracellular oncogenic gDNA sequences (HRAS and HER2) in the circulation of tumor-bearing mice. Methods and Results Surprisingly, circulating leukocytes (WBCs), especially neutrophils, contained the highest levels of mutant gDNA, which exceeded the amount of this material recovered from soluble fractions of plasma, circulating EVs, platelets, red blood cells (RBCs) and peripheral organs, as quantified by digital droplet PCR (ddPCR). Tumor excision resulted in disappearance of the WBC-associated gDNA signal within 2-9 days, which is in line with the expected half-life of these cells. EVs and nucleosomes were essential for the uptake of tumor-derived extracellular DNA by neutrophil-like cells and impacted their phenotype. Indeed, the exposure of granulocytic HL-60 cells to EVs from HRAS-driven cancer cells resulted in a selective increase in tissue factor (TF) procoagulant activity and interleukin 8 (IL-8) production. The levels of circulating thrombin-antithrombin complexes (TAT) were markedly elevated in mice harboring HRAS-driven xenografts. Conclusions Myeloid cells may represent a hitherto unrecognized reservoir of cancer-derived, EV/NS-associated oncogenic gDNA in the circulation, and a possible novel platform for liquid biopsy in cancer. In addition, uptake of this material alters the phenotype of myeloid cells, induces procoagulant and proinflammatory activity and may contribute to systemic effects associated with cancer.
Collapse
Affiliation(s)
- S Chennakrishnaiah
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - B Meehan
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - E D'Asti
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - L Montermini
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - T-H Lee
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - N Karatzas
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - M Buchanan
- Department of Oncology and Surgery, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - N Tawil
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - D Choi
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - M Divangahi
- Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Meakins-Christie Laboratories, Montreal, QC, Canada
| | - M Basik
- Department of Oncology and Surgery, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - J Rak
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| |
Collapse
|
20
|
Hosseini-Beheshti E, Choi W, Weiswald LB, Kharmate G, Ghaffari M, Roshan-Moniri M, Hassona MD, Chan L, Chin MY, Tai IT, Rennie PS, Fazli L, Tomlinson Guns ES. Exosomes confer pro-survival signals to alter the phenotype of prostate cells in their surrounding environment. Oncotarget 2018; 7:14639-58. [PMID: 26840259 PMCID: PMC4924741 DOI: 10.18632/oncotarget.7052] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 12/22/2015] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer in men. Current research on tumour-related extracellular vesicles (EVs) suggests that exosomes play a significant role in paracrine signaling pathways, thus potentially influencing cancer progression via multiple mechanisms. In fact, during the last decade numerous studies have revealed the role of EVs in the progression of various pathological conditions including cancer. Moreover, differences in the proteomic, lipidomic, and cholesterol content of exosomes derived from PCa cell lines versus benign prostate cell lines confirm that exosomes could be excellent biomarker candidates. As such, as part of an extensive proteomic analysis using LCMS we previously described a potential role of exosomes as biomarkers for PCa. Current evidence suggests that uptake of EV's into the local tumour microenvironment encouraging us to further examine the role of these vesicles in distinct mechanisms involved in the progression of PCa and castration resistant PCa. For the purpose of this study, we hypothesized that exosomes play a pivotal role in cell-cell communication in the local tumour microenvironment, conferring activation of numerous survival mechanisms during PCa progression and development of therapeutic resistance. Our in vitro results demonstrate that PCa derived exosomes significantly reduce apoptosis, increase cancer cell proliferation and induce cell migration in LNCaP and RWPE-1 cells. In conjunction with our in vitro findings, we have also demonstrated that exosomes increased tumor volume and serum PSA levels in vivo when xenograft bearing mice were administered DU145 cell derived exosomes intravenously. This research suggests that, regardless of androgen receptor phenotype, exosomes derived from PCa cells significantly enhance multiple mechanisms that contribute to PCa progression.
Collapse
Affiliation(s)
- Elham Hosseini-Beheshti
- Department of Experimental Medicine University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Wendy Choi
- The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Louis-Bastien Weiswald
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Geetanjali Kharmate
- The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Mazyar Ghaffari
- Department of Experimental Medicine University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Mani Roshan-Moniri
- Department of Experimental Medicine University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Mohamed D Hassona
- The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Leslie Chan
- The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Mei Yieng Chin
- The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Isabella T Tai
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Paul S Rennie
- Department of Urologic Sciences University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Ladan Fazli
- Department of Urologic Sciences University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- Department of Urologic Sciences University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,The Vancouver Prostate Centre University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| |
Collapse
|
21
|
Ay C, Pabinger I, Thaler J. Clinical significance of circulating microparticles for venous thrombo - embolism in cancer patients. Hamostaseologie 2017; 32:127-31. [DOI: 10.5482/ha-1164] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/20/2011] [Indexed: 12/13/2022] Open
Abstract
SummaryCancer patients have a four-to seven-fold increased risk to develop a venous thromboembolic event. Accumulating evidence from experimental and clinical studies indicates that microparticles (MPs), small procoagulant membrane vesicles that are defined by size and a negatively charged phosphatidylserine rich surface, play an important role in the pathogenesis of cancer-related venous thromboembolism (VTE). However, the clinical significance of MPs as a predictive biomarker for VTE in cancer patients has not been fully elucidated yet. This might be due to unresolved methodological problems and a lack of data from large prospective clinical studies that investigate the role of MPs in cancer-related VTE.It is the aim of this review to give an overview on the most important characteristics of MPs and studies dealing with the role of MPs in cancer-related VTE. Also recent progresses, unresolved problems and future perspectives in this research field will be discussed. In the conclusion we will assess the clinical significance of MPs in cancer-related VTE.
Collapse
|
22
|
Horowitz NA, Lavi N, Nadir Y, Brenner B. Haematological malignancies in pregnancy: An overview with an emphasis on thrombotic risks. Thromb Haemost 2017; 116:613-7. [DOI: 10.1160/th16-02-0099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
SummaryWith increase of maternal age, the incidence of haematological malignancies during pregnancy is rising and posing diagnostic and treatment challenges. Lymphoma is the fourth most common malignancy diagnosed in pregnancy; Hodgkin lymphoma is more frequent in pregnant women than non-Hodgkin lymphoma (NHL). The proportion of highly aggressive lymphomas in pregnant women is significantly higher than in non-pregnant women of reproductive age. Reproductive organ involvement is observed in almost half of pregnant women with NHL. The association of acute leukaemia and pregnancy is infrequent and it is assumed that pregnancy does not accelerate the disease course. Both cancer and pregnancy induce a procoagulant state which can lead to maternal venous thromboembolism (VTE) and placental occlusion. Pregnancy in woman with myeloproliferative neoplasms (MPN) promotes thrombotic environment, associating with an augmented risk of placental thrombosis, intrauterine growth retardation or loss and maternal thrombotic events.Haematological malignancies during pregnancy often require urgent diagnosis and management and are associated with potential adverse fetal outcomes. Most chemotherapeutic agents are teratogenic and should be avoided during the first trimester. Their use during the second and third trimesters may cause intrauterine growth restriction, premature birth and intrauterine fetal death. All chemotherapeutic drugs should be administered only after a detailed discussion with the patient and with close fetal monitoring. Chemotherapy and biological agents might also augment thrombotic risk. Guidelines for VTE prophylaxis in pregnant women with hematologic malignancies, apart from MPN, are currently unavailable, and therefore, clinical judgment should be made in each case.
Collapse
|
23
|
Yaftian M, Yari F, Ghasemzadeh M, Fallah Azad V, Haghighi M. Induction of Apoptosis in Cancer Cells of pre-B ALL Patients after Exposure to Platelets, Platelet-Derived Microparticles and Soluble CD40 Ligand. CELL JOURNAL 2017; 20:120-126. [PMID: 29308628 PMCID: PMC5759674 DOI: 10.22074/cellj.2018.5032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/12/2017] [Indexed: 11/13/2022]
Abstract
Objective The in vitro treatment of tumor cells with platelet (Plt) causes inhibition of tumor cell growth, although
mechanism of this effect is not clear yet. Induction of apoptosis has been proposed as a mechanism of Plt effects on
tumor cells. The purpose of this study was to clarify the role of Plts and Plt-derived components in the induction of
apoptosis in the blood mononuclear cells of patients with leukemia.
Materials and Methods In this experimental study, peripheral blood mononuclear cells (PBMCs) were isolated from
whole blood of five patients with childhood B-precursor acute lymphoblastic leukemia (pre-B ALL) and encountered with
Plts, Plt-derived microparticles (Plt-MPs) as well as purified soluble CD40L (sCD40L). After 48 hours of co-culture, the
anti-cancer activity of the aforementioned factors was surveyed using examination of apoptosis markers of the cells
including active caspase-3 and CD95 using ELISA and flow cytometer techniques, respectively. Additionally, staining of
the cells with 7-Aminoactinomycin D (7-AAD) was evaluated by flow cytometer technique. Trypan blue exclusion test
and WST-1 method were also used to compare the death/survival status of the cells.
Results Levels of CD95 and caspase-3 were significantly increased in the all treated groups (P<0.05). On the other
hand, trypan blue, 7-AAD and WST-1 methods showed significantly lower number of the live cells in the treated groups
(P<0.05).
Conclusion This study can show the ability of Plts, Plt-MPs and sCD40L for the induction of apoptosis in PBMCs of
pre-B-ALL patients. Further studies are necessary to elucidate the different effects of platelets on cancer cells in vitro
and in vivo.
Collapse
Affiliation(s)
- Morteza Yaftian
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Yari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran. Electronic address :
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | | | | |
Collapse
|
24
|
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
|
25
|
Breast-cancer extracellular vesicles induce platelet activation and aggregation by tissue factor-independent and -dependent mechanisms. Thromb Res 2017; 159:24-32. [DOI: 10.1016/j.thromres.2017.09.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 12/14/2022]
|
26
|
Rousseau A, Van Dreden P, Khaterchi A, Larsen AK, Elalamy I, Gerotziafas GT. Procoagulant microparticles derived from cancer cells have determinant role in the hypercoagulable state associated with cancer. Int J Oncol 2017; 51:1793-1800. [PMID: 29075792 DOI: 10.3892/ijo.2017.4170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/25/2017] [Indexed: 11/05/2022] Open
Abstract
Hypercoagulablity is a common alteration of blood coagulation in cancer patients. However, the procoagulant activity of cancer cells is not sufficient to induce hypercoagulability. The present study was aimed to identify the mechanism with which hypercoagulabilty is produced in the presence of cancer cells. We focused on the analysis of the procoagulant elements carried by cancer cell-derived microparticles (CaCe-dMP) and we evaluated the impact of microparticles associated with the cancer cells from which they stem on thrombin generation. CaCe-dMP from the cancer cells were isolated from the conditioned medium and analyzed for tissue factor (TF) and procoagulant phospholipid expression. Thrombin generation of normal plasma was assessed by the Thrombinoscope (CAT®) in the presence or absence of pancreas adeno-carcinoma cells (BXPC3) or breast cancer MCF7 cells supplemented with the respective CaCe-dMP. Both BXPC3 and MCF7 cells express abundant amounts of active TF. Phosphatidylserine was identified on the surface of CaCe-dMP, unlike the cancer cells themselves. The expression of TFa by the microparticles was significantly higher to that observed on the cancer cells. Culture of the cancer cells with their microparticles resulted in thrombin generation significantly higher as compared to the upper normal limit. In conclusion, cancer cells 'enrich' the microenvironment with procoagulant elements, especially procoagulant micro-particles which express TF and procoagulant phospholipids. The association of cancer cells with procoagulant microparticles is necessary for a state of hypercoagulability, at the level of the tumoral microenvironment. The intensity of the hypercoagulability depends on the histological type of the cancer cells.
Collapse
Affiliation(s)
- Aurélie Rousseau
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine, INSERM U938, Institut Universitaire de Cancérologie, Faculty of Medicine Pierre and Marie Curie (UPMC), Sorbonne Universities, Paris, France
| | | | - Amir Khaterchi
- Department of Biological Hematology, Tenon Hospital, University Hospitals of the East Paris, Paris, France
| | - Annette K Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine, INSERM U938, Institut Universitaire de Cancérologie, Faculty of Medicine Pierre and Marie Curie (UPMC), Sorbonne Universities, Paris, France
| | - Ismail Elalamy
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine, INSERM U938, Institut Universitaire de Cancérologie, Faculty of Medicine Pierre and Marie Curie (UPMC), Sorbonne Universities, Paris, France
| | - Grigoris T Gerotziafas
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine, INSERM U938, Institut Universitaire de Cancérologie, Faculty of Medicine Pierre and Marie Curie (UPMC), Sorbonne Universities, Paris, France
| |
Collapse
|
27
|
Savvateeva EN, Tikhonov AA, Butvilovskaya VI, Tsybulskaya MV, Rubina AY. Exosomal surface protein markers in diagnosis of colorectal cancer. Mol Biol 2017. [DOI: 10.1134/s0026893317050168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Aharon A, Sabbah A, Ben-Shaul S, Berkovich H, Loven D, Brenner B, Bar-Sela G. Chemotherapy administration to breast cancer patients affects extracellular vesicles thrombogenicity and function. Oncotarget 2017; 8:63265-63280. [PMID: 28968987 PMCID: PMC5609919 DOI: 10.18632/oncotarget.18792] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/23/2017] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BC) is the most prevalent type of malignancy in women. Extracellular vesicles (EVs) are subcellular membrane blebs that include exosomes and microparticles.
Collapse
Affiliation(s)
- Anat Aharon
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Anni Sabbah
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Shahar Ben-Shaul
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Hila Berkovich
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - David Loven
- Department of Oncology, Ha'emek Medical Center, Afula, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Gil Bar-Sela
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Oncology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
29
|
Whiteside TL. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin Exp Immunol 2017; 189:259-267. [PMID: 28369805 DOI: 10.1111/cei.12974] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2017] [Indexed: 12/17/2022] Open
Abstract
Recent emergence of exosomes as information carriers between cells has introduced us to a new previously unknown biological communication system. Multi-directional cross-talk mediated by exosomes carrying proteins, lipids and nucleic acids between normal cells, cells harbouring a pathogen or cancer and immune cells has been instrumental in determining outcomes of physiological as well as pathological conditions. Exosomes play a key role in the broad spectrum of human diseases. In cancer, tumour-derived exosomes carry multiple immunoinhibitory signals, disable anti-tumour immune effector cells and promote tumour escape from immune control. Exosomes delivering negative signals to immune cells in cancer, viral infections, autoimmune or other diseases may interfere with therapy and influence outcome. Exosomes can activate tissue cells to produce inhibitory factors and thus can suppress the host immune responses indirectly. Exosomes also promise to be non-invasive disease biomarkers with a dual capability to provide insights into immune dysfunction as well as disease progression and outcome.
Collapse
Affiliation(s)
- T L Whiteside
- Departments of Pathology, Immunology and Otolaryngology and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Procoagulant effects of lung cancer chemotherapy: impact on microparticles and cell-free DNA. Blood Coagul Fibrinolysis 2017; 28:72-82. [PMID: 26919453 DOI: 10.1097/mbc.0000000000000546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lung cancer is the second leading type of cancer, with venous thromboembolism being the second leading cause of death. Studies have shown increased levels of microparticles and cell-free DNA (CFDNA) in cancer patients, which can activate coagulation through extrinsic and intrinsic pathways, respectively. However, the impact of lung cancer chemotherapy on microparticle and/or CFDNA generation is not completely understood. The aim of the study was to study the effects of platinum-based chemotherapeutic agents on generation of procoagulant microparticles and CFDNA in vitro and in vivo. Microparticles were isolated from chemotherapy-treated monocytes, human umbilical vein endothelial cells, or cancer cells. Tissue factor (TF) and phosphatidylserine levels were characterized and thrombin/factor Xa generation assays were used to determine microparticle procoagulant activity. CFDNA levels were isolated from cell supernatants and plasma. A murine xenograft model of human lung carcinoma was used to study the procoagulant effects of TF microparticles and CFDNA in vivo. In vitro, platinum-based chemotherapy induced TF/phosphatidylserine microparticle shedding from A549 and A427 lung cancers cells, which enhanced thrombin generation in plasma in a FVII-dependent manner. CFDNA levels were increased in supernatants of chemotherapy-treated neutrophils and plasma of chemotherapy-treated mice. TF microparticles were elevated in plasma of chemotherapy-treated tumour-bearing mice. Plasma CFDNA levels are increased in chemotherapy-treated tumour-free mice and correlate with increased thrombin generation. In tumour-bearing mice, chemotherapy increases plasma levels of CFDNA and TF/phosphatidylserine microparticles. Platinum-based chemotherapy induces the shedding of TF/phosphatidylserine microparticles from tumour cells and the release of CFDNA from host neutrophils.
Collapse
|
31
|
Extracellular vesicles and blood diseases. Int J Hematol 2017; 105:392-405. [PMID: 28130731 DOI: 10.1007/s12185-017-2180-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 01/19/2023]
Abstract
Extracellular vesicles (EVs) are small membrane vesicles released from many different cell types by the exocytic budding of the plasma membrane in response to cellular activation or apoptosis. EVs disseminate various bioactive effectors originating from the parent cells and transfer functional RNA and protein between cells, enabling them to alter vascular function and induce biological responses involved in vascular homeostasis. Although most EVs in human blood originate from platelets, EVs are also released from leukocytes, erythrocytes, endothelial cells, smooth muscle cells, and cancer cells. EVs were initially thought to be small particles with procoagulant activity; however, they can also evoke cellular responses in the immediate microenvironments and transport microRNAs (miRNA) into target cells. In this review, we summarize the recent literature relevant to EVs, including a growing list of clinical disorders that are associated with elevated EV levels. These studies suggest that EVs play roles in various blood diseases.
Collapse
|
32
|
Armstrong JPK, Holme MN, Stevens MM. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS NANO 2017; 11:69-83. [PMID: 28068069 PMCID: PMC5604727 DOI: 10.1021/acsnano.6b07607] [Citation(s) in RCA: 416] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
In the past decade, extracellular vesicles (EVs) have emerged as a key cell-free strategy for the treatment of a range of pathologies, including cancer, myocardial infarction, and inflammatory diseases. Indeed, the field is rapidly transitioning from promising in vitro reports toward in vivo animal models and early clinical studies. These investigations exploit the high physicochemical stability and biocompatibility of EVs as well as their innate capacity to communicate with cells via signal transduction and membrane fusion. This review focuses on methods in which EVs can be chemically or biologically modified to broaden, alter, or enhance their therapeutic capability. We examine two broad strategies, which have been used to introduce a wide range of nanoparticles, reporter systems, targeting peptides, pharmaceutics, and functional RNA molecules. First, we explore how EVs can be modified by manipulating their parent cells, either through genetic or metabolic engineering or by introducing exogenous material that is subsequently incorporated into secreted EVs. Second, we consider how EVs can be directly functionalized using strategies such as hydrophobic insertion, covalent surface chemistry, and membrane permeabilization. We discuss the historical context of each specific technology, present prominent examples, and evaluate the complexities, potential pitfalls, and opportunities presented by different re-engineering strategies.
Collapse
Affiliation(s)
- James PK Armstrong
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College, London, U.K. SW7 2AZ
| | - Margaret N Holme
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College, London, U.K. SW7 2AZ
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College, London, U.K. SW7 2AZ
| |
Collapse
|
33
|
Abstract
Cancer patients have a significantly higher risk of developing venous thromboembolism (VTE) compared to non-cancer patients and several studies suggest that VTE risk among ambulatory cancer patients varies widely. Recently, predictive models capable of risk-stratifying a broad range of ambulatory cancer outpatients have been developed and validated; using the Khorana model a score of 2 is associated with an intermediate-high risk for VTE. However, the use of VTE prophylaxis in ambulatory patients who have cancer remains controversial. Even if important randomized clinical trials showed decreased rates of VTE events among patients who were receiving chemotherapy, the effect of prophylaxis on morbidity, mortality, and costs has not been rigorously studied. Outpatients with active cancer should be assessed for thrombosis risk and although most do not routinely require thromboprophylaxis, it should be considered for high risk patients.
Collapse
Affiliation(s)
- Davide Imberti
- Haemostasis and Thrombosis Center, Department of Internal Medicine, Hospital of Piacenza, Piacenza, Italy.
| | - Raffaella Benedetti
- Haemostasis and Thrombosis Center, Department of Internal Medicine, Hospital of Piacenza, Piacenza, Italy
| |
Collapse
|
34
|
D'Asti E, Rak J. Biological basis of personalized anticoagulation in cancer: oncogene and oncomir networks as putative regulators of coagulopathy. Thromb Res 2017; 140 Suppl 1:S37-43. [PMID: 27067976 DOI: 10.1016/s0049-3848(16)30096-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Activation of stromal response pathways in cancer is increasingly viewed as both a local and systemic extension of molecular alterations driving malignant transformation. Rather than reflecting passive and unspecific responses to anatomical abnormalities, the coagulation system is a target of oncogenic deregulation, impacting the role of clotting and fibrinolytic proteins, and integrating hemostasis, inflammation, angiogenesis and cellular growth effects in cancer. These processes signify, but do not depend on, the clinically manifest coagulopathy and thrombosis. In this regard, the role of driver mutations affecting oncoprotein coding genes such as RAS, EGFR or MET and tumour suppressors (PTEN, TP53) are well described as regulators of tissue factor (TF), protease activated receptors (PAR-1/2) and ectopic coagulation factors (FVII). Indeed, in both adult and pediatric brain tumours the expression patterns of coagulation and angiogenesis regulators (coagulome and angiome, respectively) reflect the molecular subtypes of the underlying diseases (glioblastoma or medulloblastoma) as defined by their oncogenic classifiers and clinical course. This emerging understanding is still poorly established in relation to the transforming effects of non-coding genes, including those responsible for the expression of microRNA (miR). Indeed, several miRs have been recently found to regulate TF and other effectors. We recently documented that in the context of the aggressive embryonal tumour with multilayered rosettes (ETMR) the oncogenic driver miR (miR-520g) suppresses the expression of TF and correlates with hypocoagulant tumour characteristics. Unlike in adult cancers, the growth of pediatric embryonal brain tumour cells as spheres (to maintain stem cell properties) results in upregulation of miR-520g and downregulation of TF expression and activity. We postulate that oncogenic protein and miR coding genes form alternative pathways of coagulation system regulation in different tumour settings, a property necessitating more personalised and biologically-based approaches to anticoagulation.
Collapse
Affiliation(s)
- Esterina D'Asti
- McGill University, Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Janusz Rak
- McGill University, Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
Khan E, Ambrose NL, Ahnström J, Kiprianos AP, Stanford MR, Eleftheriou D, Brogan PA, Mason JC, Johns M, Laffan MA, Haskard DO. A low balance between microparticles expressing tissue factor pathway inhibitor and tissue factor is associated with thrombosis in Behçet's Syndrome. Sci Rep 2016; 6:38104. [PMID: 27924945 PMCID: PMC5141484 DOI: 10.1038/srep38104] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/26/2016] [Indexed: 12/14/2022] Open
Abstract
Thrombosis is common in Behçet's Syndrome (BS), and there is a need for better biomarkers for risk assessment. As microparticles expressing Tissue Factor (TF) can contribute to thrombosis in preclinical models, we investigated whether plasma microparticles expressing Tissue Factor (TF) are increased in BS. We compared blood plasma from 72 healthy controls with that from 88 BS patients (21 with a history of thrombosis (Th+) and 67 without (Th-). Using flow cytometry, we found that the total plasma MP numbers were increased in BS compared to HC, as were MPs expressing TF and Tissue Factor Pathway Inhibitor (TFPI) (all p < 0.0001). Amongst BS patients, the Th+ group had increased total and TF positive MP numbers (both p ≤ 0.0002) compared to the Th- group, but had a lower proportion of TFPI positive MPs (p < 0.05). Consequently, the ratio of TFPI positive to TF positive MP counts (TFPI/TF) was significantly lower in Th+ versus Th- BS patients (p = 0.0002), and no patient with a TFPI/TF MP ratio >0.7 had a history of clinical thrombosis. We conclude that TF-expressing MP are increased in BS and that an imbalance between microparticulate TF and TFPI may predispose to thrombosis.
Collapse
Affiliation(s)
- E Khan
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College, London, UK
| | - N L Ambrose
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College, London, UK
| | - J Ahnström
- Centre for Haematology, Department of Medicine, Imperial College, London, UK
| | - A P Kiprianos
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College, London, UK
| | - M R Stanford
- Department of Ophthalmology, King's College, London, UK
| | - D Eleftheriou
- Institute of Child Heath, University College, London, UK
| | - P A Brogan
- Institute of Child Heath, University College, London, UK
| | - J C Mason
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College, London, UK
| | - M Johns
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College, London, UK
| | - M A Laffan
- Centre for Haematology, Department of Medicine, Imperial College, London, UK
| | - D O Haskard
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
36
|
Kim OY, Lee J, Gho YS. Extracellular vesicle mimetics: Novel alternatives to extracellular vesicle-based theranostics, drug delivery, and vaccines. Semin Cell Dev Biol 2016; 67:74-82. [PMID: 27916566 DOI: 10.1016/j.semcdb.2016.12.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles are nano-sized spherical bilayered proteolipids encasing various components. Cells of all domains of life actively release these vesicles to the surroundings including various biological fluids. These extracellular vesicles are known to play pivotal roles in numerous pathophysiological functions. Extracellular vesicles have distinct characteristics, like high biocompatibility, safety, and nano-sized diameters that allow efficient drug loading capacity and long blood circulation half-life. These characteristics of extracellular vesicles have engrossed many scientists to harness them as new tools for novel delivery systems. This review will highlight the current state of the arts and problems of such extracellular vesicle-based theranostics, drug delivery and vaccines, and introduce "extracellular vesicle mimetics" as the novel alternative of extracellular vesicles. We hope to provide insights into the potential of extracellular vesicle mimetics as superior substitute to the natural extracellular vesicles that can be applied to theranostics, drug delivery, and vaccines against various diseases.
Collapse
Affiliation(s)
- Oh Youn Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jaewook Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
| |
Collapse
|
37
|
Noulsri E, Udomwinijsilp P, Lerdwana S, Chongkolwatana V, Permpikul P. Differences in levels of platelet-derived microparticles in platelet components prepared using the platelet rich plasma, buffy coat, and apheresis procedures. Transfus Apher Sci 2016; 56:135-140. [PMID: 28029568 DOI: 10.1016/j.transci.2016.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/07/2016] [Accepted: 10/21/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND There has been an increased interest in platelet-derived microparticles (PMPs) in transfusion medicine. Little is known about PMP status during the preparation of platelet concentrates for transfusion. AIM The aim of this study is to compare the PMP levels in platelet components prepared using the buffy coat (BC), platelet-rich plasma platelet concentrate (PRP-PC), and apheresis (AP) processes. METHODS Platelet components were prepared using the PRP-PC and BC processes. Apheresis platelets were prepared using the Trima Accel and Amicus instruments. The samples were incubated with annexin A5-FITC, CD41-PE, and CD62P-APC. At day 1 after processing, the PMPs and activated platelets were determined using flow cytometry. RESULTS Both the percentage and number of PMPs were higher in platelet components prepared using the Amicus instrument (2.6±1.8, 32802±19036 particles/μL) than in platelet components prepared using the Trima Accel instrument (0.5±0.4, 7568±5298 particles/μL), BC (1.2±0.6, 12,920±6426 particles/μL), and PRP-PC (0.9±0.6, 10731±5514 particles/μL). Both the percentage and number of activated platelets were higher in platelet components prepared using the Amicus instrument (33.2±13.9, 427553±196965 cells/μL) than in platelet components prepared using the Trima Accel instrument (16.2±6.1, 211209±87706 cells/μL), BC (12.9±3.2, 140624±41003 cells/μL), and PRP-PC (21.1±6.3, 265210±86257 cells/μL). CONCLUSIONS The study suggests high variability of PMPs and activated platelets in platelet components prepared using different processes. This result may be important in validating the instruments involved in platelet blood collection and processing.
Collapse
Affiliation(s)
- Egarit Noulsri
- Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Prapaporn Udomwinijsilp
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surada Lerdwana
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Viroje Chongkolwatana
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parichart Permpikul
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
38
|
Flamant S, Tamarat R. Extracellular Vesicles and Vascular Injury: New Insights for Radiation Exposure. Radiat Res 2016; 186:203-18. [PMID: 27459703 DOI: 10.1667/rr14482.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This article reviews our current knowledge about cell-derived extracellular vesicles (EVs), including microparticles and exosomes, and their emergence as mediators of a new important mechanism of cell-to-cell communication. Particular emphasis has been given to the increasing involvement of EVs in the field of radiation-induced vascular injury. Although EVs have been considered for a long time as cell "dust", they in fact precisely reflect the physiological state of the cells. The role of microparticles and exosomes in mediating vascular dysfunction suggests that they may represent novel pathways in short- or long-distance paracrine intercellular signaling in vascular environment. In this article, the mechanisms involved in the biogenesis of microparticles and exosomes, their composition and participation in the pathogenesis of vascular dysfunction are discussed. Furthermore, this article highlights the concept of EVs as potent vectors of biological information and protagonists of an intercellular communication network. Special emphasis is made on EV-mediated microRNA transfer and on the principal consequences of such signal exchange on vascular injury and radiation-induced nontargeted effect. The recent progress in elucidating the biology of EVs has provided new insights for the field of radiation, advancing their use as diagnostic biomarkers or in therapeutic interventions.
Collapse
Affiliation(s)
- Stéphane Flamant
- Institute for Radiological Protection and Nuclear Safety (IRSN) PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Radia Tamarat
- Institute for Radiological Protection and Nuclear Safety (IRSN) PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| |
Collapse
|
39
|
Heiler S, Wang Z, Zöller M. Pancreatic cancer stem cell markers and exosomes - the incentive push. World J Gastroenterol 2016; 22:5971-6007. [PMID: 27468191 PMCID: PMC4948278 DOI: 10.3748/wjg.v22.i26.5971] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/03/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PaCa) has the highest death rate and incidence is increasing. Poor prognosis is due to late diagnosis and early metastatic spread, which is ascribed to a minor population of so called cancer stem cells (CSC) within the mass of the primary tumor. CSC are defined by biological features, which they share with adult stem cells like longevity, rare cell division, the capacity for self renewal, differentiation, drug resistance and the requirement for a niche. CSC can also be identified by sets of markers, which for pancreatic CSC (Pa-CSC) include CD44v6, c-Met, Tspan8, alpha6beta4, CXCR4, CD133, EpCAM and claudin7. The functional relevance of CSC markers is still disputed. We hypothesize that Pa-CSC markers play a decisive role in tumor progression. This is fostered by the location in glycolipid-enriched membrane domains, which function as signaling platform and support connectivity of the individual Pa-CSC markers. Outside-in signaling supports apoptosis resistance, stem cell gene expression and tumor suppressor gene repression as well as miRNA transcription and silencing. Pa-CSC markers also contribute to motility and invasiveness. By ligand binding host cells are triggered towards creating a milieu supporting Pa-CSC maintenance. Furthermore, CSC markers contribute to the generation, loading and delivery of exosomes, whereby CSC gain the capacity for a cell-cell contact independent crosstalk with the host and neighboring non-CSC. This allows Pa-CSC exosomes (TEX) to reprogram neighboring non-CSC towards epithelial mesenchymal transition and to stimulate host cells towards preparing a niche for metastasizing tumor cells. Finally, TEX communicate with the matrix to support tumor cell motility, invasion and homing. We will discuss the possibility that CSC markers are the initial trigger for these processes and what is the special contribution of CSC-TEX.
Collapse
|
40
|
Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin B 2016; 6:287-96. [PMID: 27471669 PMCID: PMC4951582 DOI: 10.1016/j.apsb.2016.02.001] [Citation(s) in RCA: 890] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/19/2016] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Exosomes are small intracellular membrane-based vesicles with different compositions that are involved in several biological and pathological processes. The exploitation of exosomes as drug delivery vehicles offers important advantages compared to other nanoparticulate drug delivery systems such as liposomes and polymeric nanoparticles; exosomes are non-immunogenic in nature due to similar composition as body׳s own cells. In this article, the origin and structure of exosomes as well as their biological functions are outlined. We will then focus on specific applications of exosomes as drug delivery systems in pharmaceutical drug development. An overview of the advantages and challenges faced when using exosomes as a pharmaceutical drug delivery vehicles will also be discussed.
Collapse
Key Words
- ALIX, ALG-2 interacting protein X
- ATPase, adenosine triphosphatase
- BBB, blood–brain barrier
- CCK-8, cell counting kit-8
- CD, cluster of differentiation
- DIL, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate
- DNA, deoxyribonucleic acid
- Drug delivery systems
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- ESCRT, endosomal sorting complexes required for transport
- EV, extracellular vesicle
- EpCAM, epithelial cell adhesion molecule
- Exosomes
- Extracellular vesicles
- HEK293, human embryonic kidney cell line 293
- HIV, human immunodeficiency virus
- HMGA2, high-mobility group AT-hook protein
- HeLa, Henrietta Lacks cells
- Hsp, heat shock proteins
- IL-6, interleukin-6
- ILVs, intraluminal vesicles
- LPS, lipopolysaccharides
- MAPK-1, mitogen-activated protein kinase 1
- MHC, major histocompatibility complex
- MPS, mononuclear phagocyte system
- MVB, multi-vesicular body biogenesis
- Nanocarrier
- PBMC, peripheral blood mononuclear cells
- PD, Parkinson’s disease
- PEG, polyethylene glycol
- RNA, ribonucleic acid
- ROS, reactive oxygen species
- RPE1, retinal pigment epithelial cells 1
- TNF-α, tumor necrosis factor α
- TSG101, tumor susceptibility gene 101
- VPS4, vacuolar protein sorting-associated protein 4
- kRAS, Kirsten rat sarcoma
- mRNA, messenger RNA
- miRNA, micro RNA
- siRNA, small interference RNA
Collapse
Affiliation(s)
| | | | - Venkatareddy Nadithe
- Manchester University, College of Pharmacy, Natural & Health Sciences, Fort Wayne, IN 46845, USA
| |
Collapse
|
41
|
Morgan ER, Mason WP, Maurice C. A critical balance: managing coagulation in patients with glioma. Expert Rev Neurother 2016; 16:803-14. [PMID: 27101362 DOI: 10.1080/14737175.2016.1181542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer-associated thrombosis, including both arterial and venous thromboembolism (VTE), is a significant source of morbidity and mortality in patients with glioma. This risk is highest in the immediate postoperative period and is increased by chemotherapy, radiation, and corticosteroids. Systemic anticoagulation with low molecular weight heparin is the treatment of choice in both the therapeutic and prophylactic settings. However, these patients are also at risk of intracranial hemorrhage, a potentially catastrophic complication of anticoagulation, and this risk must be carefully balanced against the risk of VTE. In this review we outline the incidence, pathophysiology and management of thrombosis in patients with glioma, with a focus on clinical considerations including perioperative management, chemotherapy-induced thrombocytopenia, and end-of-life management.
Collapse
Affiliation(s)
- Erin R Morgan
- a Pencer Brain Tumor Centre , Princess Margaret Hospital Cancer Centre , Toronto , Canada.,b Department of Medical Oncology and Hematology , University of Toronto , Toronto , Canada
| | - Warren P Mason
- a Pencer Brain Tumor Centre , Princess Margaret Hospital Cancer Centre , Toronto , Canada.,b Department of Medical Oncology and Hematology , University of Toronto , Toronto , Canada
| | - Catherine Maurice
- a Pencer Brain Tumor Centre , Princess Margaret Hospital Cancer Centre , Toronto , Canada.,b Department of Medical Oncology and Hematology , University of Toronto , Toronto , Canada
| |
Collapse
|
42
|
An Advanced Orthotopic Ovarian Cancer Model in Mice for Therapeutic Trials. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2585787. [PMID: 27110559 PMCID: PMC4821970 DOI: 10.1155/2016/2585787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 12/27/2022]
Abstract
A nude mouse received subcutaneous injection of human ovarian cancer cells HO-8910PM to form a tumor, and then the tumor fragment was surgically transplanted to the ovary of a recipient mouse to establish an orthotopic cancer model. Tumors occurred in 100% of animals. A mouse displayed an ovarian mass, ascites, intraperitoneal spread, and lung metastasis at natural death. The mean survival time was 34.1 ± 17.2 days, with median survival time of 28.5 days. The findings indicated that the present mouse model can reflect the biological behavior of advanced human ovarian cancers. This in vivo model can be used to explore therapeutic means against chemoresistance and metastasis, and an effective treatment would prolong the survival time.
Collapse
|
43
|
Nomura S, Niki M, Nisizawa T, Tamaki T, Shimizu M. Microparticles as Biomarkers of Blood Coagulation in Cancer. BIOMARKERS IN CANCER 2015; 7:51-6. [PMID: 26462252 PMCID: PMC4592056 DOI: 10.4137/bic.s30347] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/20/2022]
Abstract
Cancer is associated with hypercoagulopathy and increased risk of thrombosis. This negatively influences patient morbidity and mortality. Cancer is also frequently complicated by the development of venous thromboembolism (VTE). Tumor-derived tissue factor (TF)-bearing microparticles (MPs) are associated with VTE events in malignancy. MPs are small membrane vesicles released from many different cell types by exocytic budding of the plasma membrane in response to cellular activation or apoptosis. MPs may also be involved in clinical diseases through expression of procoagulative phospholipids. The detection of TF-expressing MPs in cancer patients may be clinically useful. In lung and breast cancer patients, MPs induce metastasis and angiogenesis and may be indicators of vascular complications. Additionally, MPs in patients with various types of cancer possess adhesion proteins and bind target cells to promoting cancer progression or metastasis. Overexpression of TF by cancer cells is closely associated with tumor progression, and shedding of TF-expressing MPs by cancer cells correlates with the genetic status of cancer. Consequently, TF-expressing MPs represent important markers to consider in the prevention of and therapy for VTE complications in cancer patients.
Collapse
Affiliation(s)
- Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Maiko Niki
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Tohru Nisizawa
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Takeshi Tamaki
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Michiomi Shimizu
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
44
|
Microparticles That Form Immune Complexes as Modulatory Structures in Autoimmune Responses. Mediators Inflamm 2015; 2015:267590. [PMID: 26300590 PMCID: PMC4537755 DOI: 10.1155/2015/267590] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/10/2014] [Accepted: 12/13/2014] [Indexed: 12/29/2022] Open
Abstract
Microparticles (MPs) are induced during apoptosis, cell activation, and even “spontaneous” release. Initially MPs were considered to be inert cellular products with no biological function. However, an extensive research and functional characterization have shown that the molecular composition and the effects of MPs depend upon the cellular background and the mechanism inducing them. They possess a wide spectrum of biological effects on intercellular communication by transferring different molecules able to modulate other cells. MPs interact with their target cells through different mechanisms: membrane fusion, macropinocytosis, and receptor-mediated endocytosis. However, when MPs remain in the extracellular milieu, they undergo modifications such as citrullination, glycosylation, and partial proteolysis, among others, becoming a source of neoantigens. In rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), reports indicated elevated levels of MPs with different composition, content, and effects compared with those isolated from healthy individuals. MPs can also form immune complexes amplifying the proinflammatory response and tissue damage. Their early detection and characterization could facilitate an appropriate diagnosis optimizing the pharmacological strategies, in different diseases including cancer, infection, and autoimmunity. This review focuses on the current knowledge about MPs and their involvement in the immunopathogenesis of SLE and RA.
Collapse
|
45
|
Tzoran I, Rebibo-Sabbah A, Brenner B, Aharon A. Disease dynamics in patients with acute myeloid leukemia: new biomarkers. Exp Hematol 2015; 43:936-43. [PMID: 26232698 DOI: 10.1016/j.exphem.2015.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/15/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by rapid growth of leukemic blast cells. Extracellular vesicles (EVs), shedding from various cells, express antigens, reflecting their cellular origin. The current study was designed to explore the role of circulating EVs as potential biomarkers of AML activity and predictors of thrombogenicity in patients with this malignancy. Blood samples were collected from healthy controls and patients with newly diagnosed AML at three time points: diagnosis, nadir, and remission. EV concentration, cell origin, and expression of coagulation proteins were characterized using fluorescence-activated cell sorting. EV cytokine contents were evaluated by protein array. Procoagulant activity was assessed using Factor Xa chromogenic assay. Forty-two AML patients were enrolled in the study. Total EV numbers were higher in patients in first remission compared with controls, whereas blast EV counts were higher in patients at diagnosis compared with controls and patients in remission. Blast EV levels were significantly lower in patients who achieved remission and were alive at 3-year follow up compared with their succumbed counterparts. At all three time points, percentage of endothelial EVs was higher in patients compared with controls. EV procoagulant activity was elevated at diagnosis and in remission, and, unlike controls' EVs, patients' EVs increased endothelial cell thrombogenicity. EVs of AML patients express membrane proteins of blast cells and might serve as biomarkers of leukemia dynamics and presence of minimal residual disease. Increased levels of endothelial EVs and their procoagulant activity may indicate a vascular injury associated with a hypercoagulable state in AML.
Collapse
Affiliation(s)
- Inna Tzoran
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Internal Medicine C, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
| | - Annie Rebibo-Sabbah
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Anat Aharon
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
46
|
Chalasani P, Marron M, Roe D, Clarke K, Iannone M, Livingston RB, Shan JS, Stopeck AT. A phase I clinical trial of bavituximab and paclitaxel in patients with HER2 negative metastatic breast cancer. Cancer Med 2015; 4:1051-9. [PMID: 25826750 PMCID: PMC4529343 DOI: 10.1002/cam4.447] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 12/20/2022] Open
Abstract
Bavituximab is a chimeric monoclonal antibody that targets phosphatidylserine (PS). PS is externalized on cells in the tumor microenvironment when exposed to hypoxia and/or other physiological stressors. On attaching to PS, bavituximab is thought to promote antitumor immunity through its effects on PS receptors in monocytes, and myeloid-derived suppressor cells, as well as trigger antitumor effects by inducing an antibody-dependent cellular cytotoxicity on tumor-associated endothelial cells. We conducted a phase I clinical trial of bavituximab in combination with paclitaxel in patients with HER2-negative metastatic breast cancer. Patients were treated with weekly paclitaxel (80 mg/m2 for 3/4 weeks) and weekly bavituximab (3 mg/kg for 4/4 weeks). Correlative studies included the measurement of circulating microparticles, endothelial cells, and apoptotic tumor cells by flow cytometry. Fourteen patients with metastatic breast cancer were enrolled; all were evaluable for toxicity and 13 were evaluable for response. Treatment resulted in an overall response rate (RR) of 85% with a median progression-free survival (PFS) of 7.3 months. Bone pain, fatigue, headache, and neutropenia were the most common adverse effects. Infusion-related reactions were the most common adverse event related to bavituximab therapy. Correlative studies showed an increase in the PS-expressing apoptotic circulating tumor cells in response to bavituximab, but not with paclitaxel. No changes in the number of circulating endothelial cells or apoptotic endothelial cells were observed with therapy. Platelet and monocyte-derived microparticles decreased after initiation of bavituximab. Bavituximab in combination with paclitaxel is well tolerated for treatment of patients with metastatic breast cancer with promising results observed in terms of clinical RRs and PFS. The toxicity profile of bavituximab is notable for manageable infusion-related reactions with no evidence for increased thrombogenicity. Recent preclinical data suggest that bavituximab can also promote antitumor immune activity that should be explored in future clinical trials.
Collapse
Affiliation(s)
- Pavani Chalasani
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Marilyn Marron
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Denise Roe
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Kathryn Clarke
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Maria Iannone
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Robert B Livingston
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Joseph S Shan
- Peregrine Pharmaceuticals, Inc., 14282 Franklin Avenue, Tustin, California, 92780
| | - Alison T Stopeck
- Stonybrook Medicine University, PO Box 1554, Stonybrook, New York, 11790
| |
Collapse
|
47
|
Brunetta DM, De Santis GC, Silva-Pinto AC, Oliveira de Oliveira LC, Covas DT. Hydroxyurea increases plasma concentrations of microparticles and reduces coagulation activation and fibrinolysis in patients with sickle cell anemia. Acta Haematol 2014; 133:287-94. [PMID: 25472687 DOI: 10.1159/000362148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 03/10/2014] [Indexed: 12/17/2022]
Abstract
Microparticles (MPs) are present in healthy subjects and their concentration increases in patients at high risk of thrombosis. We evaluated 10 patients with sickle cell anemia (SCA) treated with hydroxyurea (HU) and 13 SCA patients without this treatment. MP concentrations were determined by flow cytometry. Coagulation was evaluated using the thrombin-antithrombin complex (TAT) and D-dimers. Total MP concentrations were increased in the HU-treated group (265 × 10(6)/ml vs. 67.45 × 10(6)/ml; p = 0.0026), as well as MPs derived from RBC (67.83 × 10(6)/ml vs. 26.31 × 10(6)/ml; p = 0.05), monocytes (51.31 × 10(6)/ml vs. 9.03 × 10(6)/ml; p = 0.0084), monocytes with tissue factor (TF) expression (2.27 × 10(6)/ml vs. 0.27 × 10(6)/ml; p = 0.0058), endothelium (49.42 × 10(6)/ml vs. 7.23 × 10(6)/ml; p = 0.007) and endothelium with TF (1.42 × 10(6)/ml vs. 0.26 × 10(6)/ml; p = 0.0043). Furthermore, the concentrations of TAT (7.56 vs. 10.98 µg/l; p = 0.014) and D-dimers (0.65 vs. 1.29 µg/ml; p = 0.007) were reduced with HU. The MP elevation may suggest a direct cytotoxic effect of HU. Another explanation is a cell surface increase secondary to a megaloblastic process, resulting in increased vesicle release. In our opinion, the known benefits of HU on SCA patients, along with the reduction in coagulation activation, surpass its potential detrimental effect on MPs. Future studies should elucidate the role of MPs and demonstrate their significance in different contexts.
Collapse
Affiliation(s)
- Denise Menezes Brunetta
- Center for Cell-Based Therapy, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | | | | |
Collapse
|
48
|
Biologicals, platelet apoptosis and human diseases: An outlook. Crit Rev Oncol Hematol 2014; 93:149-58. [PMID: 25439323 DOI: 10.1016/j.critrevonc.2014.11.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 10/06/2014] [Accepted: 11/10/2014] [Indexed: 01/07/2023] Open
Abstract
Platelets, once considered mediators of hemostasis and thrombosis, are now known to be involved in wound healing, inflammation, cardiovascular diseases, diabetes, arthritis, and cancer. Recent reports attest that platelets possess the cellular machinery to undergo apoptosis and that platelet apoptosis can be triggered by myriad stimuli including chemical and physical agonists, and pathophysiological conditions. Augmented rate of platelet apoptosis leads to thrombocytopenia, bleeding disorders and microparticle generation. Despite knowing the significant role of platelets in health and disease, and that any alterations in platelet functions can wreak havoc to the health, the offshoot reactions of therapeutic drugs on platelets and the far-reaching consequences are often neglected. The present review focuses on the impact of platelet apoptosis and the role of platelet-derived microparticles on different pathophysiological conditions. It also touches upon the effects of biologicals on platelets, and discusses the need to overcome the adverse effects of pro-apoptotic drugs through auxiliary therapy.
Collapse
|
49
|
Abstract
Extracellular vesicles (EVs), comprised of exosomes, microparticles, apoptotic bodies, and other microvesicles, are shed from a variety of cells upon cell activation or apoptosis. EVs promote clot formation, mediate pro-inflammatory processes, transfer proteins and miRNA to cells, and induce cell signaling that regulates cell differentiation, proliferation, migration, invasion, and apoptosis. This paper will review the contribution of EVs in hematological disorders, including hemoglobinopathies (sickle cell disease, thalassemia), paroxysmal nocturnal hemoglobinuria, and hematological malignancies (lymphomas, myelomas, and acute and chronic leukemias).
Collapse
Affiliation(s)
- Anat Aharon
- Microvesicles Research Laboratory, Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus; ; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Annie Rebibo-Sabbah
- Microvesicles Research Laboratory, Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus; ; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Inna Tzoran
- Microvesicles Research Laboratory, Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus; ; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel; ; Department of Internal Medicine C, Rambam Health Care Campus, Haifa, Israel
| | - Carina Levin
- Microvesicles Research Laboratory, Thrombosis and Hemostasis Unit, Department of Hematology, Rambam Health Care Campus; ; Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel; ; Pediatric Hematology Unit and Pediatric Department B, Emek Medical Center, Afula, Israel
| |
Collapse
|
50
|
Tan X, Gong YZ, Wu P, Liao DF, Zheng XL. Mesenchymal stem cell-derived microparticles: a promising therapeutic strategy. Int J Mol Sci 2014; 15:14348-63. [PMID: 25196436 PMCID: PMC4159854 DOI: 10.3390/ijms150814348] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/25/2014] [Accepted: 08/04/2014] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells that give rise to various cell types of the mesodermal germ layer. Because of their unique ability to home in on injured and cancerous tissues, MSCs are of great potential in regenerative medicine. MSCs also contribute to reparative processes in different pathological conditions, including cardiovascular diseases and cancer. However, many studies have shown that only a small proportion of transplanted MSCs can actually survive and be incorporated into host tissues. The effects of MSCs cannot be fully explained by their number. Recent discoveries suggest that microparticles (MPs) derived from MSCs may be important for the physiological functions of their parent. Though the physiological role of MSC-MPs is currently not well understood, inspiring results indicate that, in tissue repair and anti-cancer therapy, MSC-MPs have similar pro-regenerative and protective properties as their cellular counterparts. Thus, MSC-MPs represent a promising approach that may overcome the obstacles and risks associated with the use of native or engineered MSCs.
Collapse
Affiliation(s)
- Xi Tan
- Division of Stem Cell Regulation and Application, College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha 410208, China.
| | - Yong-Zhen Gong
- Division of Stem Cell Regulation and Application, College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha 410208, China.
| | - Ping Wu
- Division of Stem Cell Regulation and Application, College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha 410208, China.
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha 410208, China.
| | - Xi-Long Zheng
- Division of Stem Cell Regulation and Application, College of Medicine, Hunan University of Traditional Chinese Medicine, Changsha 410208, China.
| |
Collapse
|