301
|
Molecular crosstalk between tumour and brain parenchyma instructs histopathological features in glioblastoma. Oncotarget 2017; 7:31955-71. [PMID: 27049916 PMCID: PMC5077988 DOI: 10.18632/oncotarget.7454] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/29/2016] [Indexed: 12/11/2022] Open
Abstract
The histopathological and molecular heterogeneity of glioblastomas represents a major obstacle for effective therapies. Glioblastomas do not develop autonomously, but evolve in a unique environment that adapts to the growing tumour mass and contributes to the malignancy of these neoplasms. Here, we show that patient-derived glioblastoma xenografts generated in the mouse brain from organotypic spheroids reproducibly give rise to three different histological phenotypes: (i) a highly invasive phenotype with an apparent normal brain vasculature, (ii) a highly angiogenic phenotype displaying microvascular proliferation and necrosis and (iii) an intermediate phenotype combining features of invasion and vessel abnormalities. These phenotypic differences were visible during early phases of tumour development suggesting an early instructive role of tumour cells on the brain parenchyma. Conversely, we found that tumour-instructed stromal cells differentially influenced tumour cell proliferation and migration in vitro, indicating a reciprocal crosstalk between neoplastic and non-neoplastic cells. We did not detect any transdifferentiation of tumour cells into endothelial cells. Cell type-specific transcriptomic analysis of tumour and endothelial cells revealed a strong phenotype-specific molecular conversion between the two cell types, suggesting co-evolution of tumour and endothelial cells. Integrative bioinformatic analysis confirmed the reciprocal crosstalk between tumour and microenvironment and suggested a key role for TGFβ1 and extracellular matrix proteins as major interaction modules that shape glioblastoma progression. These data provide novel insight into tumour-host interactions and identify novel stroma-specific targets that may play a role in combinatorial treatment strategies against glioblastoma.
Collapse
|
302
|
Focus on Mesenchymal Stem Cell-Derived Exosomes: Opportunities and Challenges in Cell-Free Therapy. Stem Cells Int 2017; 2017:6305295. [PMID: 29410682 PMCID: PMC5749272 DOI: 10.1155/2017/6305295] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/05/2017] [Accepted: 11/22/2017] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells have been at the forefront of regenerative medicine for many years. Exosomes, which are nanovesicles involved in intercellular communication and the transportation of genetic material transportation that can be released by mesenchymal stem cells, have been recently reported to play a role in cell-free therapy of many diseases, including myocardial infarction, drug addiction, and status epilepticus. They are also thought to help ameliorate inflammation-induced preterm brain injury, liver injury, and various types of cancer. This review highlights recent advances in the exploration of mesenchymal stem cell-derived exosomes in therapeutic applications. The natural contents, drug delivery potency, modification methods, and drug loading methods of exosomes are also discussed.
Collapse
|
303
|
Li Y, Wan S, Liu G, Cai W, Huo D, Li G, Yang M, Wang Y, Guan G, Ding N, Liu F, Zeng W, Zhu C. Netrin-1 Promotes Inflammation Resolution to Achieve Endothelialization of Small-Diameter Tissue Engineering Blood Vessels by Improving Endothelial Progenitor Cells Function In Situ. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2017; 4:1700278. [PMID: 29270340 PMCID: PMC5738088 DOI: 10.1002/advs.201700278] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/07/2017] [Indexed: 05/05/2023]
Abstract
The transplant of small-diameter tissue engineering blood vessels (small-diameter TEBVs) (<6 mm) in vascular replacement therapy often fails because of early onset thrombosis and long-standing chronic inflammation. The specific inflammation state involved in small-diameter TEBVs transplants remains unclear, and whether promoting inflammation resolution would be useful for small-diameter TEBVs therapy need study. The neural protuberant orientation factor 1 (Netrin-1) is found present in endothelial cells of natural blood vessels and has anti-inflammatory effects. This work generates netrin-1-modified small-diameter TEBVs by using layer-by-layer self-assembly to resolve the inflammation. The results show that netrin-1 reprograms macrophages (MΦ) to assume an anti-inflammatory phenotype and promotes the infiltration and subsequent efflux of MΦ from inflamed sites over time, which improves the local microenvironment and the function of early homing endothelial progenitor cells (EPCs). Small-diameter TEBVs modified by netrin-1 achieve endothelialization after 30 d and retain patency at 14 months. These findings suggest that promoting the resolution of inflammation in time is necessary to induce endothelialization of small-diameter TEBVs and prevent early thrombosis and problems associated with chronic inflammation. Furthermore, this work finds that the MΦ-derived exosomes can target and regulate EPCs, which may serve as a useful treatment for other inflammatory diseases.
Collapse
Affiliation(s)
- Yanzhao Li
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Simin Wan
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Ge Liu
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Wang Cai
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Da Huo
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Gang Li
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Mingcan Yang
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Yuxin Wang
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Ge Guan
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Ning Ding
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Feila Liu
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Wen Zeng
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| | - Chuhong Zhu
- Department of AnatomyNational and Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory for Vascular ImplantsKey Lab for Biomechanics and Tissue Engineering of ChongqingState Key Laboratory of Trauma, burn and Combined injuryThird Military Medical UniversityChongqing400038China
| |
Collapse
|
304
|
Neo SH, Chung KY, Quek JM, Too HP. Trehalose significantly enhances the recovery of serum and serum exosomal miRNA from a paper-based matrix. Sci Rep 2017; 7:16686. [PMID: 29192155 PMCID: PMC5709463 DOI: 10.1038/s41598-017-16960-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/19/2017] [Indexed: 12/27/2022] Open
Abstract
The preservation of nucleic acids from clinical samples is critical to facilitate accurate molecular diagnosis. The use of a paper matrix, Flinders Technology Associates (FTA) Elute cards, to archive DNA and viral RNA is well-documented. However, the feasibility of FTA Elute cards for archiving serum and serum exosomal microRNAs (miRNAs) remains unclear. Here, we performed a comprehensive evaluation of FTA Elute cards for miRNA storage and recovery in different pre-analytical conditions. The recovery of serum miRNA dry-spotted on FTA Elute cards by direct elution with water at high temperature was poor. However, serum miRNAs dry-spotted on the cards were isolated with about 40% yield when using QIAzol lysis reagent and recovery was improved remarkably (>80%) upon extraction from cards pre-treated with trehalose. miRNAs stored on the cards remained stable at room temperature and can be kept for prolonged periods. Furthermore, miRNAs could be similarly recovered from serum exosomes dry-spotted on the cards. Importantly, when using sera from gastric cancer (GC) patients, the miRNAs were efficiently recovered from trehalose pre-treated cards without affecting their representation. Collectively, we have demonstrated the potential of FTA Elute cards to archive serum and serum exosomal miRNAs, making it useful for biomarker discovery and diagnostics.
Collapse
Affiliation(s)
- Shu Hui Neo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
| | - Ka Yan Chung
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Jia Min Quek
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Heng-Phon Too
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Block MD7, 8 Medical Drive, Singapore, 117597, Singapore.
| |
Collapse
|
305
|
Multiple Myeloma-Derived Exosomes Regulate the Functions of Mesenchymal Stem Cells Partially via Modulating miR-21 and miR-146a. Stem Cells Int 2017; 2017:9012152. [PMID: 29333170 PMCID: PMC5733127 DOI: 10.1155/2017/9012152] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/05/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Exosomes derived from cancer cells can affect various functions of mesenchymal stem cells (MSCs) via conveying microRNAs (miRs). miR-21 and miR-146a have been demonstrated to regulate MSC proliferation and transformation. Interleukin-6 (IL-6) secreted from transformed MSCs in turn favors the survival of multiple myeloma (MM) cells. However, the effects of MM exosomes on MSC functions remain largely unclear. In this study, we investigated the effects of OPM2 (a MM cell line) exosomes (OPM2-exo) on regulating the proliferation, cancer-associated fibroblast (CAF) transformation, and IL-6 secretion of MSCs and determined the role of miR-21 and miR-146a in these effects. We found that OPM2-exo harbored high levels of miR-21 and miR-146a and that OPM2-exo coculture significantly increased MSC proliferation with upregulation of miR-21 and miR-146a. Moreover, OPM2-exo induced CAF transformation of MSCs, which was evidenced by increased fibroblast-activated protein (FAP), α-smooth muscle actin (α-SMA), and stromal-derived factor 1 (SDF-1) expressions and IL-6 secretion. Inhibition of miR-21 or miR-146a reduced these effects of OPM2-exo on MSCs. In conclusion, MM could promote the proliferation, CAF transformation, and IL-6 secretion of MSCs partially through regulating miR21 and miR146a.
Collapse
|
306
|
Pando A, Reagan JL, Quesenberry P, Fast LD. Extracellular vesicles in leukemia. Leuk Res 2017; 64:52-60. [PMID: 29190514 DOI: 10.1016/j.leukres.2017.11.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/12/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EV) are nano-sized membrane enclosed vehicles that are involved in cell-to-cell communication and carry cargo that is representative of the parent cell. Recent studies have highlighted the significant roles leukemia EVs play in tumor progression, and ways in which they can lead to treatment evasion, thus meriting further investigation. Leukemia EVs are involved in crosstalk between the leukemia cell and its surroundings, transforming it into a cancer favorable microenvironment. Due to the diverse biological content found in leukemia EVs, they have an assortment of effects on the cells they interact with and can be harnessed as candidates for diagnostic and therapeutic treatments. This review focuses on EVs in the context of leukemia and the means by which they modulate their microenvironment, hematopoiesis, and the immune system to facilitate malignancy. We will also address current and prospective EV-based therapeutics.
Collapse
Affiliation(s)
- Alejandro Pando
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - John L Reagan
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter Quesenberry
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Loren D Fast
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
307
|
Griessinger E, Moschoi R, Biondani G, Peyron JF. Mitochondrial Transfer in the Leukemia Microenvironment. Trends Cancer 2017; 3:828-839. [PMID: 29198439 DOI: 10.1016/j.trecan.2017.10.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/18/2022]
Abstract
The bone marrow microenvironment (BMME) is a complex ecosystem that instructs and protects hematopoietic stem cells (HSCs) and their malignant counterparts, the leukemia-initiating cells (LICs). Within the physical and functional crosstalk that takes place between HSCs, LICs, and the BMME, the transfer of organelles and of mitochondria in particular is an important new intercellular communication mode in addition to adhesion molecules, tunneling nanotubes (TNTs), and the paracrine secretion of cytokines, (onco)metabolites, and extracellular vesicles (EVs). In this review we discuss the functional roles of mitochondrial transfer between BMME and leukemic cells, and give insights into this new mechanism of drug resistance whose understanding will open the way to innovative anticancer adjuvant treatments.
Collapse
Affiliation(s)
- Emmanuel Griessinger
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France.
| | - Ruxanda Moschoi
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France
| | - Giulia Biondani
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France
| | - Jean-François Peyron
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France.
| |
Collapse
|
308
|
Aubatin A, Sako N, Decrouy X, Donnadieu E, Molinier-Frenkel V, Castellano F. IL4-induced gene 1 is secreted at the immune synapse and modulates TCR activation independently of its enzymatic activity. Eur J Immunol 2017; 48:106-119. [PMID: 28891065 DOI: 10.1002/eji.201646769] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 01/11/2023]
Abstract
Amino-acid catabolizing enzymes produced by mononuclear phagocytes play a central role in regulating the immune response. The mammalian phenylalanine-catabolizing enzyme IL4-induced gene 1 (IL4I1) inhibits effector T lymphocyte proliferation and facilitates regulatory T-cell development. IL4I1 expression by macrophages of various human tumors may affect patient prognosis as it facilitates tumor escape from the T-cell response in murine models. Its enzymatic activity appears to participate in its effects, but some actions of IL4I1 remain unclear. Here, we show that the presence of IL4I1 during T-cell activation decreases early signaling events downstream of TCR stimulation, resulting in global T-cell inhibition which is more pronounced when there is CD28 costimulation. Surprisingly, the enzymatic activity of IL4I1 is not involved. Focal secretion of IL4I1 into the immune synaptic cleft and its binding to CD3+ lymphocytes could be important in IL4I1 immunosuppressive mechanism of action.
Collapse
Affiliation(s)
- Aude Aubatin
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Nouhoum Sako
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Xavier Decrouy
- INSERM, U955, Plateforme d'imagerie, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Emmanuel Donnadieu
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| | - Flavia Castellano
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Plateforme de Ressources Biologiques, Créteil, France
| |
Collapse
|
309
|
Alonso S, Jones RJ, Ghiaur G. Retinoic acid, CYP26, and drug resistance in the stem cell niche. Exp Hematol 2017; 54:17-25. [PMID: 28754309 PMCID: PMC5603425 DOI: 10.1016/j.exphem.2017.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/25/2022]
Abstract
The bone marrow niche is essential for hematopoietic stem cells to maintain lifelong blood production by balancing their self-renewal and differentiation. Hematologic malignancies have a similar hierarchical organization to their normal counterparts, with rare populations of cancer stem cells that rely on the microenvironment to survive and propagate their differentiated malignant progenitor cells. Cancer cells alter their microenvironment to create a supportive niche, where they endure chemotherapy, survive as minimal residual disease (MRD), and eventually prevail at relapse. Powerful morphogens, such as retinoids, Wnt/βcatenin, Notch, and Hedgehog, control stem cell fates across tissues, including normal and malignant hematopoiesis. The molecular conversations between these pathways and the mechanisms that control their activity and create gradients at cellular scale remain a mystery. Here, we discuss accumulating evidence suggesting that cytochrome P450 (CYP26), the primary retinoid-inactivating enzyme, plays a critical role in the integration of two of these molecular programs: the retinoid and Hedgehog pathways. Induction of stromal CYP26 by either one of these pathways limits retinoic acid concentration in the stem cell niche, with profound effects on tissue homeostasis and drug resistance. Bypassing this gatekeeping mechanism holds promise for overcoming drug resistance and improving clinical outcomes in hematological malignancies and cancer in general.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Cytochrome P450 Family 26/genetics
- Cytochrome P450 Family 26/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Hematologic Neoplasms/drug therapy
- Hematologic Neoplasms/genetics
- Hematologic Neoplasms/metabolism
- Hematologic Neoplasms/pathology
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplasm, Residual
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Stem Cell Niche/drug effects
- Stem Cell Niche/genetics
- Tretinoin/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Salvador Alonso
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Richard J Jones
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Gabriel Ghiaur
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
310
|
Alamoudi AA, Alnoury A, Gad H. miRNA in tumour metabolism and why could it be the preferred pathway for energy reprograming. Brief Funct Genomics 2017; 17:157-169. [DOI: 10.1093/bfgp/elx023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
311
|
Yang F, Ning Z, Ma L, Liu W, Shao C, Shu Y, Shen H. Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Mol Cancer 2017; 16:148. [PMID: 28851377 PMCID: PMC5576273 DOI: 10.1186/s12943-017-0718-4] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 12/21/2022] Open
Abstract
Purpose The present review aimed to assess the role of exosomal miRNAs in cancer-associated fibroblasts (CAFs), normal fibroblasts (NFs), and cancer cells. The roles of exosomal miRNAs and miRNA dysregulation in CAF formation and activation were summarized. Methods All relevant publications were retrieved from the PubMed database, with key words such as CAFs, CAF, stromal fibroblasts, cancer-associated fibroblasts, miRNA, exosomal, exosome, and similar terms. Results Recent studies have revealed that CAFs, NFs, and cancer cells can secrete exosomal miRNAs to affect each other. Dysregulation of miRNAs and exosomal miRNAs influence the formation and activation of CAFs. Furthermore, miRNA dysregulation in CAFs is considered to be associated with a secretory phenotype change, tumor invasion, tumor migration and metastasis, drug resistance, and poor prognosis. Conclusions Finding of exosomal miRNA secretion provides novel insights into communication among CAFs, NFs, and cancer cells. MicroRNA dysregulation is also involved in the whole processes of CAF formation and function. Dysregulation of miRNAs in CAFs can affect the secretory phenotype of the latter cells.
Collapse
Affiliation(s)
- Fengming Yang
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Shanghai, China
| | - Zhiqiang Ning
- Department of Oncology, The first People's Hospital of Wujiang district, Suzhou, 215200, China
| | - Ling Ma
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Shanghai, China
| | - Weitao Liu
- Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Chuchu Shao
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Shanghai, China
| | - Yongqian Shu
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China. .,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Shanghai, China.
| | - Hua Shen
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China. .,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Shanghai, China.
| |
Collapse
|
312
|
Mesenchymal Stem Cells in Myeloid Malignancies: A Focus on Immune Escaping and Therapeutic Implications. Stem Cells Int 2017; 2017:6720594. [PMID: 28947904 PMCID: PMC5602646 DOI: 10.1155/2017/6720594] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/06/2017] [Accepted: 07/20/2017] [Indexed: 01/07/2023] Open
Abstract
The importance of the bone marrow microenvironment forming the so-called niche in physiologic hemopoiesis is largely known, and recent evidences support the presence of stromal alterations from the molecular to the cytoarchitectural level in hematologic malignancies. Various alterations in cell adhesion, metabolism, cytokine signaling, autophagy, and methylation patterns of tumor-derived mesenchymal stem cells have been demonstrated, contributing to the genesis of a leukemic permissive niche. This niche allows both the ineffective haematopoiesis typical of myelodysplastic syndromes and the differentiation arrest, proliferation advantage, and clone selection which is the hallmark of acute myeloid leukemia. Furthermore, the immune system, both adaptive and innate, encompassing mesenchymal-derived cells, has been shown to take part to the leukemic niche. Here, we critically review the state of art about mesenchymal stem cell role in myelodysplastic syndromes and acute myeloid leukemia, focusing on immune escaping mechanisms as a target for available and future anticancer therapies.
Collapse
|
313
|
Isolation of Fibroblast-Activation Protein-Specific Cancer-Associated Fibroblasts. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4825108. [PMID: 28890895 PMCID: PMC5584363 DOI: 10.1155/2017/4825108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 01/27/2023]
Abstract
The current study is to develop a gentle and efficient method for purification of fibroblast-activation protein positive (FAP+) cancer-associated fibroblasts (CAFs) from tumor tissues. Fresh tissues were isolated from BALB/c-Nude mice bearing human liver cancer cell line (HepG2), fully minced and separated into three parts, and digested with trypsin digestion and then treated with collagenase type IV once, twice, or thrice, respectively. Finally, the cells were purified by using FAP magnetic beads. The isolated CAFs were grown in culture medium and detected for the surface expression of fibroblast-activation protein (FAP). The number of adherent cells which were obtained by digestion process with twice collagenase type IV digestion was (5.99 ± 0.18) × 104, much more than that with the only once collagenase type IV digestion (2.58 ± 0.41) × 104 (P < 0.0001) and similar to thrice collagenase type IV digestion. The percentage of FAP+ CAFs with twice collagenase type IV digestion (38.5%) was higher than that with the only once collagenase type IV digestion (20.0%) and little higher than thrice collagenase type IV digestion (37.5%). The FAP expression of CAFs was quite different from normal fibroblasts (NFs). The fibroblasts isolated by the innovation are with high purity and being in wonderful condition and display the features of CAFs.
Collapse
|
314
|
S100-A9 protein in exosomes from chronic lymphocytic leukemia cells promotes NF-κB activity during disease progression. Blood 2017; 130:777-788. [DOI: 10.1182/blood-2017-02-769851] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
Key Points
Plasma-derived exosomes from patients with CLL exhibit different protein cargo compositions depending on disease status and progression. S100-A9 protein is overexpressed and S100-A9 cargo in exosomes activates NF-κB pathway in patients with CLL during disease progression.
Collapse
|
315
|
van Attekum MH, Eldering E, Kater AP. Chronic lymphocytic leukemia cells are active participants in microenvironmental cross-talk. Haematologica 2017; 102:1469-1476. [PMID: 28775118 PMCID: PMC5685246 DOI: 10.3324/haematol.2016.142679] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
The importance of the tumor microenvironment in chronic lymphocytic leukemia is
widely accepted. Nevertheless, the understanding of the complex interplay
between the various types of bystander cells and chronic lymphocytic leukemia
cells is incomplete. Numerous studies have indicated that bystander cells
provide chronic lymphocytic leukemia-supportive functions, but it has also
become clear that chronic lymphocytic leukemia cells actively engage in the
formation of a supportive tumor microenvironment through several cross-talk
mechanisms. In this review, we describe how chronic lymphocytic leukemia cells
participate in this interplay by inducing migration and tumor-supportive
differentiation of bystander cells. Furthermore, chronic lymphocytic
leukemia-mediated alterations in the interactions between bystander cells are
discussed. Upon bystander cell interaction, chronic lymphocytic leukemia cells
secrete cytokines and chemokines such as migratory factors [chemokine
(C-C motif) ligand 22 and chemokine (CC motif) ligand 2], which result
in further recruitment of T cells but also of monocyte-derived cells. Within the
tumor microenvironment, chronic lymphocytic leukemia cells induce
differentiation towards a tumor-supportive M2 phenotype of monocyte-derived
cells and suppress phagocytosis, but also induce increased numbers of supportive
regulatory T cells. Like other tumor types, the differentiation of stromal cells
towards supportive cancer-associated fibroblasts is critically dependent on
chronic lymphocytic leukemia-derived factors such as exosomes and
platelet-derived growth factor. Lastly, both chronic lymphocytic leukemia and
bystander cells induce a tolerogenic tumor microenvironment; chronic lymphocytic
leukemia-secreted cytokines, such as interleukin-10, suppress cytotoxic T-cell
functions, while chronic lymphocytic leukemia-associated monocyte-derived cells
contribute to suppression of T-cell function by producing the immune checkpoint
factor, programmed cell death-ligand 1. Deeper understanding of the active
involvement and cross-talk of chronic lymphocytic leukemia cells in shaping the
tumor microenvironment may offer novel clues for designing therapeutic
strategies.
Collapse
Affiliation(s)
- Martijn Ha van Attekum
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| | - Arnon P Kater
- Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands .,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
316
|
Shimada Y, Minna JD. Exosome mediated phenotypic changes in lung cancer pathophysiology. Transl Cancer Res 2017; 6:S1040-S1042. [PMID: 30613485 DOI: 10.21037/tcr.2017.07.09] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yoshihisa Shimada
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
317
|
Haderk F, Schulz R, Iskar M, Cid LL, Worst T, Willmund KV, Schulz A, Warnken U, Seiler J, Benner A, Nessling M, Zenz T, Göbel M, Dürig J, Diederichs S, Paggetti J, Moussay E, Stilgenbauer S, Zapatka M, Lichter P, Seiffert M. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol 2017; 2:2/13/eaah5509. [PMID: 28754746 DOI: 10.1126/sciimmunol.aah5509] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 03/23/2017] [Accepted: 06/12/2017] [Indexed: 12/14/2022]
Abstract
In chronic lymphocytic leukemia (CLL), monocytes and macrophages are skewed toward protumorigenic phenotypes, including the release of tumor-supportive cytokines and the expression of immunosuppressive molecules such as programmed cell death 1 ligand 1 (PD-L1). To understand the mechanism driving protumorigenic skewing in CLL, we evaluated the role of tumor cell-derived exosomes in the cross-talk with monocytes. We carried out RNA sequencing and proteome analyses of CLL-derived exosomes and identified noncoding Y RNA hY4 as a highly abundant RNA species that is enriched in exosomes from plasma of CLL patients compared with healthy donor samples. Transfer of CLL-derived exosomes or hY4 alone to monocytes resulted in key CLL-associated phenotypes, including the release of cytokines, such as C-C motif chemokine ligand 2 (CCL2), CCL4, and interleukin-6, and the expression of PD-L1. These responses were abolished in Toll-like receptor 7 (TLR7)-deficient monocytes, suggesting exosomal hY4 as a driver of TLR7 signaling. Pharmacologic inhibition of endosomal TLR signaling resulted in a substantially reduced activation of monocytes in vitro and attenuated CLL development in vivo. Our results indicate that exosome-mediated transfer of noncoding RNAs to monocytes contributes to cancer-related inflammation and concurrent immune escape via PD-L1 expression.
Collapse
Affiliation(s)
- Franziska Haderk
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralph Schulz
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Murat Iskar
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Llaó Cid
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Worst
- Division of Signaling and Functional Genomics, DKFZ, Heidelberg, Germany
| | - Karolin V Willmund
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angela Schulz
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Genomics and Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Uwe Warnken
- Genomics and Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Jana Seiler
- Division of RNA Biology and Cancer (B150), DKFZ, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, DKFZ, Heidelberg, Germany
| | | | - Thorsten Zenz
- Department of Molecular Therapy in Hematology and Oncology and Department of Translational Oncology, National Center for Tumor Diseases (NCT); DKFZ; and Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Maria Göbel
- Department of Hematology, Essen University Hospital, Essen, Germany
| | - Jan Dürig
- Department of Hematology, Essen University Hospital, Essen, Germany
| | - Sven Diederichs
- Division of RNA Biology and Cancer (B150), DKFZ, Heidelberg, Germany.,Division of Cancer Research, Department of Thoracic Surgery, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany
| | - Jérôme Paggetti
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Etienne Moussay
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | | | - Marc Zapatka
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Seiffert
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
318
|
Abramowicz A, Widlak P, Pietrowska M. Proteomic analysis of exosomal cargo: the challenge of high purity vesicle isolation. MOLECULAR BIOSYSTEMS 2017; 12:1407-19. [PMID: 27030573 DOI: 10.1039/c6mb00082g] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The re-discovery of exosomes as intercellular messengers with high potential for diagnostic and therapeutic utility has led to them becoming a popular topic of research in recent years. One of the essential research areas in this field is the characterization of exosomal cargo, which includes numerous non-randomly packed proteins and nucleic acids. Unexpectedly, a very challenging aspect of exploration of extracellular vesicles has turned out to be their effective and selective isolation. The plurality of developed protocols leads to qualitative and quantitative variability in terms of the obtained exosomes, which significantly affects the results of downstream analyses and makes them difficult to compare, reproduce and interpret between research groups. Currently, there is a general consensus among the exosome-oriented community concerning the urgent need for the optimization and standardization of methods employed for the purification of these vesicles. Hence, we review here several strategies for exosome preparation including ultracentrifugation, chemical precipitation, affinity capturing and filtration techniques. The advantages and disadvantages of different approaches are discussed with special emphasis being placed on their adequacy for proteomics applications, which are particularly sensitive to sample quality. We conclude that certain methods, exemplified by ultracentrifugation combined with iodixanol density gradient centrifugation or gel filtration, although labor-intensive, provide superior quality exosome preparations suitable for reliable analysis by mass spectrometry.
Collapse
Affiliation(s)
- Agata Abramowicz
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland.
| | - Piotr Widlak
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland.
| | - Monika Pietrowska
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland.
| |
Collapse
|
319
|
Edlinger L, Berger-Becvar A, Menzl I, Hoermann G, Greiner G, Grundschober E, Bago-Horvath Z, Al-Zoughbi W, Hoefler G, Brostjan C, Gille L, Moriggl R, Spittler A, Sexl V, Hoelbl-Kovacic A. Expansion of BCR/ABL1 + cells requires PAK2 but not PAK1. Br J Haematol 2017; 179:229-241. [PMID: 28707321 PMCID: PMC5655792 DOI: 10.1111/bjh.14833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/30/2017] [Indexed: 01/13/2023]
Abstract
The p21‐activated kinases (PAKs) are key nodes in oncogenic signalling pathways controlling growth, survival, and motility of cancer cells. Their activity is increased in many human cancers and is associated with poor prognosis. To date, PAK deregulation has mainly been studied in solid tumours, where PAK1 and PAK4 are the main isoforms deregulated. We show that PAK1 and PAK2 are the critical isoforms in a BCR/ABL1+ haematopoietic malignancy. In suspension, leukaemic cells deficient for PAK1 and PAK2 undergo apoptosis, while the loss of either protein is well tolerated. Transfer of medium conditioned by shPAK2‐ but not shPAK1‐expressing leukaemic cells interferes with endothelial cell growth. We found that leukaemic cells produce exosomes containing PAK2. Transfer of isolated exosomes supports endothelial cell proliferation. In parallel, we found that leukaemic cells explicitly require PAK2 to grow towards an extracellular matrix. PAK2‐deficient cells fail to form colonies in methylcellulose and to induce lymphomas in vivo. PAK2 might therefore be the critical isoform in leukaemic cells by controlling tumour growth in a dual manner: vascularization via exosome‐mediated transfer to endothelial cells and remodelling of the extracellular matrix. This finding suggests that the PAK2 isoform represents a promising target for the treatment of haematological diseases.
Collapse
Affiliation(s)
- Leo Edlinger
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Angelika Berger-Becvar
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria.,Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Ingeborg Menzl
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Eva Grundschober
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria.,Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Wael Al-Zoughbi
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Christine Brostjan
- Department of Surgery, Research Laboratories, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Lars Gille
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andreas Spittler
- Core Facility Flow Cytometry & Department of Surgery, Research Laboratories, Medical University of Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
320
|
Lobb RJ, Lima LG, Möller A. Exosomes: Key mediators of metastasis and pre-metastatic niche formation. Semin Cell Dev Biol 2017; 67:3-10. [DOI: 10.1016/j.semcdb.2017.01.004] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/22/2016] [Accepted: 01/06/2017] [Indexed: 12/21/2022]
|
321
|
Vargas A, Roux-Dalvai F, Droit A, Lavoie JP. Neutrophil-Derived Exosomes: A New Mechanism Contributing to Airway Smooth Muscle Remodeling. Am J Respir Cell Mol Biol 2017; 55:450-61. [PMID: 27105177 DOI: 10.1165/rcmb.2016-0033oc] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neutrophils infiltrate the airways of patients with asthma of all severities, yet their role in the pathogenesis of asthma and their contribution to airway remodeling is largely unknown. We hypothesized that neutrophils modulate airway smooth muscle (ASM) proliferation in asthma by releasing bioactive exosomes. These newly discovered nano-sized vesicles have the capacity to modulate immune responses, cell migration, cell differentiation, and other aspects of cell-to-cell communication. The aim of the study is to determine whether bioactive exosomes are released by neutrophils, and, if so, characterize their proteomic profile and evaluate their capacity to modulate ASM cell proliferation. Exosomes were isolated from equine neutrophil supernatants by differential centrifugation and filtration methods, followed by size-exclusion chromatography. Nanovesicles were characterized using electron microscopy, particle size determination, and proteomic analyses. Exosomes were cocultured with ASM cells and analyzed for exosome internalization by confocal microscopy. ASM proliferation was measured using an impedance-based system. Neutrophils release exosomes that have characteristic size, morphology, and exosomal markers. We identified 271 proteins in exosomes from both LPS and unstimulated neutrophils, and 16 proteins that were differentially expressed, which carried proteins associated with immune response and positive regulation of cell communication. Furthermore, neutrophil-derived exosomes were rapidly internalized by ASM cells and altered their proliferative properties. Upon stimulation of LPS, neutrophil-derived exosomes can enhance the proliferation of ASM cells and could therefore play an important role in the progression of asthma and promoting airway remodeling in severe and corticosteroid-insensitive patients with asthma.
Collapse
Affiliation(s)
- Amandine Vargas
- 1 Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada; and
| | - Florence Roux-Dalvai
- 2 Proteomics Platform, Centre Hospitalier Universitaire de Québec, Research Center and Faculty of Medicine, Laval University, Sainte-Foy, Quebec, Canada
| | - Arnaud Droit
- 2 Proteomics Platform, Centre Hospitalier Universitaire de Québec, Research Center and Faculty of Medicine, Laval University, Sainte-Foy, Quebec, Canada
| | - Jean-Pierre Lavoie
- 1 Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada; and
| |
Collapse
|
322
|
Nakata R, Shimada H, Fernandez GE, Fanter R, Fabbri M, Malvar J, Zimmermann P, DeClerck YA. Contribution of neuroblastoma-derived exosomes to the production of pro-tumorigenic signals by bone marrow mesenchymal stromal cells. J Extracell Vesicles 2017; 6:1332941. [PMID: 28717423 PMCID: PMC5505006 DOI: 10.1080/20013078.2017.1332941] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Indexed: 12/20/2022] Open
Abstract
The bone marrow (BM) niche is a microenvironment promoting survival, dormancy and therapeutic resistance in tumor cells. Central to this function are mesenchymal stromal cells (MSCs). Here, using neuroblastoma (NB) as a model, we demonstrate that NB cells release an extracellular vesicle (EVs) whose protein cargo is enriched in exosomal proteins but lacks cytokines and chemokines. Using three different purification methods, we then demonstrate that NB-derived exosomes were captured by MSCs and induced the production of pro-tumorigenic cytokines and chemokines, including interleukin-6 (IL-6), IL-8/CXCL8, vascular endothelial cell growth factor and monocyte-chemotactic protein-1, with exosomes prepared by size exclusion chromatography having the highest activity. We found no correlation between the IL-6 and IL-8/CXCL8 stimulatory activity of exosomes from eight NB cell lines and their origin, degree of MYCN amplification, drug resistance and disease status. We then demonstrate that the uptake of NB exosomes by MSCs was associated with a rapid increase in ERK1/2 and AKT activation, and that blocking ERK1/2 but not AKT activation inhibited the IL-6 and IL-8/CXCL8 production by MSCs without affecting exosome uptake. Thus, we describe a new mechanism by which NB cells induce in MSCs an inflammatory reaction that contributes to a favorable microenvironment in the BM.
Collapse
Affiliation(s)
- Rie Nakata
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA.,Department of Pediatrics, University of Southern California, Los Angeles, CA, USA
| | - Hiroyuki Shimada
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, University of Southern California, Los Angeles, CA, USA
| | - G Esteban Fernandez
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Rob Fanter
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Muller Fabbri
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA.,Department of Pediatrics, University of Southern California, Los Angeles, CA, USA.,Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, USA
| | - Jemily Malvar
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068-CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Yves A DeClerck
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA.,Department of Pediatrics, University of Southern California, Los Angeles, CA, USA.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
323
|
Mirzaei H, Fathullahzadeh S, Khanmohammadi R, Darijani M, Momeni F, Masoudifar A, Goodarzi M, Mardanshah O, Stenvang J, Jaafari MR, Mirzaei HR. State of the art in microRNA as diagnostic and therapeutic biomarkers in chronic lymphocytic leukemia. J Cell Physiol 2017; 233:888-900. [PMID: 28084621 DOI: 10.1002/jcp.25799] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022]
Abstract
Early diagnostic is one of the most important steps in cancer therapy which helps to design and choose a better therapeutic approach. The finding of biomarkers in various levels including genomics, transcriptomics, and proteomics levels could provide better treatment for various cancers such as chronic lymphocytic leukemia (CLL). The CLL is the one of main lymphoid malignancies which is specified by aggregation of mature B lymphocytes. Among different biomarkers (e.g., CD38, chromosomes abnormalities, ZAP-70, TP53, and microRNA [miRNA]), miRNAs have appeared as new diagnostic and therapeutic biomarkers in patients with the CLL disease. Multiple lines of evidence indicated that deregulation of miRNAs could be associated with pathological events which are present in the CLL. These molecules have an effect on a variety of targets such as Bcl2, c-fos, c-Myc, TP53, TCL1, and STAT3 which play critical roles in the CLL pathogenesis. It has been shown that expression of miRNAs could lead to the activation of B cells and B cell antigen receptor (BCR). Moreover, exosomes containing miRNAs are one of the other molecules which could contribute to BCR stimulation and progression of CLL cells. Hence, miRNAs and exosomes released from CLL cells could be used as potential diagnostic and therapeutic biomarkers for CLL. This critical review focuses on a very important aspect of CLL based on biomarker discovery covers the pros and cons of using miRNAs as important diagnostics and therapeutics biomarkers for this deadly disease.
Collapse
Affiliation(s)
- Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sima Fathullahzadeh
- Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran
| | - Razieh Khanmohammadi
- Department of pediatric dentistry, School of dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansoreh Darijani
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Momeni
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Goodarzi
- Faculty of Bioscience Engineering, Department of Biosystems, Katholieke Universiteit Leuven - KULeuven, Heverlee, Belgium
| | - Omid Mardanshah
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jan Stenvang
- Faculty of Health and Medical Sciences, Section for Molecular Disease Biology, Department of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Mirzaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
324
|
Crompot E, Van Damme M, Pieters K, Vermeersch M, Perez-Morga D, Mineur P, Maerevoet M, Meuleman N, Bron D, Lagneaux L, Stamatopoulos B. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica 2017; 102:1594-1604. [PMID: 28596280 PMCID: PMC5685228 DOI: 10.3324/haematol.2016.163337] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 06/05/2017] [Indexed: 12/13/2022] Open
Abstract
Interactions between chronic lymphocytic leukemia (CLL) B cells and the bone marrow (BM) microenvironment play a major function in the physiopathology of CLL. Extracellular vesicles (EVs), which are composed of exosomes and microparticles, play an important role in cell communication. However, little is known about their role in CLL / microenvironment interactions. In the present study, EVs purified by ultracentrifugation from BM mesenchymal stromal cell (BM-MSC) cultures were added to CLL B cells. After their integration into CLL B cells, we observed a decrease of leukemic cell spontaneous apoptosis and an increase in their chemoresistance to several drugs, including fludarabine, ibrutinib, idelalisib and venetoclax after 24 hours. Spontaneous (P=0.0078) and stromal cell-derived factor 1α -induced migration capacities of CLL B cells were also enhanced (P=0.0020). A microarray study highlighted 805 differentially expressed genes between leukemic cells cultured with or without EVs. Of these, genes involved in the B-cell receptor pathway such as CCL3/4, EGR1/2/3, and MYC were increased. Interestingly, this signature presents important overlaps with other microenvironment stimuli such as B-cell receptor stimulation, CLL/nurse-like cells co-culture or those provided by a lymph node microenvironment. Finally, we showed that EVs from MSCs of leukemic patients also rescue leukemic cells from spontaneous or drug-induced apoptosis. However, they induce a higher migration and also a stronger gene modification compared to EVs of healthy MSCs. In conclusion, we show that EVs play a crucial role in CLL B cells/BM microenvironment communication.
Collapse
Affiliation(s)
- Emerence Crompot
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Michael Van Damme
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Karlien Pieters
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Perez-Morga
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Philippe Mineur
- Department of Hemato-Oncology, Grand Hôpital de Charleroi, Gilly, Belgium
| | - Marie Maerevoet
- Hematology Department, Jules Bordet Institute, Brussels, Belgium
| | | | - Dominique Bron
- Hematology Department, Jules Bordet Institute, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| |
Collapse
|
325
|
Han L, Xu J, Xu Q, Zhang B, Lam EWF, Sun Y. Extracellular vesicles in the tumor microenvironment: Therapeutic resistance, clinical biomarkers, and targeting strategies. Med Res Rev 2017; 37:1318-1349. [PMID: 28586517 DOI: 10.1002/med.21453] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 12/16/2022]
Abstract
Numerous studies have proved that cell-nonautonomous regulation of neoplastic cells is a distinctive and essential characteristic of tumorigenesis. Two way communications between the tumor and the stroma, or within the tumor significantly influence disease progression and modify treatment responses. In the tumor microenvironment (TME), malignant cells utilize paracrine signaling initiated by adjacent stromal cells to acquire resistance against multiple types of anticancer therapies, wherein extracellular vesicles (EVs) substantially promote such events. EVs are nanoscaled particles enclosed by phospholipid bilayers, and can mediate intercellular communications between cancerous cells and the adjacent microenvironment to accelerate pathological proceeding. Here we review the most recent studies of EV biology and focus on key cell lineages of the TME and their EV cargoes that are biologically active and responsible for cancer resistance, including proteins, RNAs, and other potentially essential components. Since EVs are emerging as novel but critical elements in establishing and maintaining hallmarks of human cancer, timely and insightful understanding of their molecular properties and functional mechanisms would pave the road for clinical diagnosis, prognosis, and effective targeting in the global landscape of precision medicine. Further, we address the potential of EVs as promising biomarkers in cancer clinics and summarize the technical improvements in EV preparation, analysis, and imaging. We highlight the practical issues that should be exercised with caution to guide the development of targeting agents and therapeutic methodologies to minimize cancer resistance driven by EVs, thereby allowing to effectively control the early steps of disease exacerbation.
Collapse
Affiliation(s)
- L Han
- Key Lab of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, China
| | - J Xu
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine & Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Q Xu
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine & Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - B Zhang
- Key Lab of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, China
| | - E W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Y Sun
- Key Lab of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, China.,Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, USA
| |
Collapse
|
326
|
Caivano A, La Rocca F, Laurenzana I, Trino S, De Luca L, Lamorte D, Del Vecchio L, Musto P. Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy. Int J Mol Sci 2017; 18:E1183. [PMID: 28574430 PMCID: PMC5486006 DOI: 10.3390/ijms18061183] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of particles, between 15 nanometers and 10 microns in diameter, released by almost all cell types in physiological and pathological conditions, including tumors. EVs have recently emerged as particularly interesting informative vehicles, so that they could be considered a true "cell biopsy". Indeed, EV cargo, including proteins, lipids, and nucleic acids, generally reflects the nature and status of the origin cells. In some cases, EVs are enriched of peculiar molecular cargo, thus suggesting at least a degree of specific cellular packaging. EVs are identified as important and critical players in intercellular communications in short and long distance interplays. Here, we examine the physiological role of EVs and their activity in cross-talk between bone marrow microenvironment and neoplastic cells in hematological malignancies (HMs). In these diseases, HM EVs can modify tumor and bone marrow microenvironment, making the latter "stronger" in supporting malignancy, inducing drug resistance, and suppressing the immune system. Moreover, EVs are abundant in biologic fluids and protect their molecular cargo against degradation. For these and other "natural" characteristics, EVs could be potential biomarkers in a context of HM liquid biopsy and therapeutic tools. These aspects will be also analyzed in this review.
Collapse
Affiliation(s)
- Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Francesco La Rocca
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Luigi Del Vecchio
- CEINGE-Biotecnologie Avanzate scarl, Federico II University, 80138 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
327
|
de Couto G, Gallet R, Cambier L, Jaghatspanyan E, Makkar N, Dawkins JF, Berman BP, Marbán E. Exosomal MicroRNA Transfer Into Macrophages Mediates Cellular Postconditioning. Circulation 2017; 136:200-214. [PMID: 28411247 DOI: 10.1161/circulationaha.116.024590] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 03/30/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiosphere-derived cells (CDCs) confer cardioprotection in acute myocardial infarction by distinctive macrophage (Mϕ) polarization. Here we demonstrate that CDC-secreted exosomes (CDCexo) recapitulate the cardioprotective effects of CDC therapy known as cellular postconditioning. METHODS Rats and pigs underwent myocardial infarction induced by ischemia/reperfusion before intracoronary infusion of CDCexo, inert fibroblast exosomes (Fbexo; control), or vehicle. Two days later, infarct size was quantified. Macrophages were isolated from cardiac tissue or bone marrow for downstream analyses. RNA sequencing was used to determine exosome content and alterations in gene expression profiles in Mϕ. RESULTS Administration of CDCexo but not Fbexo after reperfusion reduces infarct size in rat and pig models of myocardial infarction. Furthermore, CDCexo reduce the number of CD68+ Mϕ within infarcted tissue and modify the polarization state of Mϕ so as to mimic that induced by CDCs. CDCexo are enriched in several miRNAs (including miR-146a, miR-181b, and miR-126) relative to Fbexo. Reverse pathway analysis of whole-transcriptome data from CDCexo-primed Mϕ implicated miR-181b as a significant (P=1.3x10-21) candidate mediator of CDC-induced Mϕ polarization, and PKCδ (protein kinase C δ) as a downstream target. Otherwise inert Fbexo loaded selectively with miR-181b alter Mϕ phenotype and confer cardioprotective efficacy in a rat model of myocardial infarction. Adoptive transfer of PKCδ-suppressed Mϕ recapitulates cardioprotection. CONCLUSIONS Our data support the hypothesis that exosomal transfer of miR-181b from CDCs into Mϕ reduces PKCδ transcript levels and underlies the cardioprotective effects of CDCs administered after reperfusion.
Collapse
Affiliation(s)
- Geoffrey de Couto
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.)
| | - Romain Gallet
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.)
| | - Linda Cambier
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.)
| | - Ervin Jaghatspanyan
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.)
| | - Nupur Makkar
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.)
| | - James Frederick Dawkins
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.)
| | - Benjamin P Berman
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.).
| | - Eduardo Marbán
- From Cedars-Sinai Heart Institute, Los Angeles, CA (G.d.C., R.G., L.C., E.J., N.M., J.F.D., B.P.B., E.M.); and Cedars-Sinai Center for Bioinformatics and Functional Genomics, Los Angeles, CA (B.P.B.).
| |
Collapse
|
328
|
Rashed MH, Kanlikilicer P, Rodriguez-Aguayo C, Pichler M, Bayraktar R, Bayraktar E, Ivan C, Filant J, Silva A, Aslan B, Denizli M, Mitra R, Ozpolat B, Calin GA, Sood AK, Abd-Ellah MF, Helal GK, Berestein GL. Exosomal miR-940 maintains SRC-mediated oncogenic activity in cancer cells: a possible role for exosomal disposal of tumor suppressor miRNAs. Oncotarget 2017; 8:20145-20164. [PMID: 28423620 PMCID: PMC5386751 DOI: 10.18632/oncotarget.15525] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022] Open
Abstract
Exosomes have emerged as important mediators of diverse biological functions including tumor suppression, tumor progression, invasion, immune escape and cell-to-cell communication, through the release of molecules such as mRNAs, miRNAs, and proteins. Here, we identified differentially expressed exosomal miRNAs between normal epithelial ovarian cell line and both resistant and sensitive ovarian cancer (OC) cell lines. We found miR-940 as abundant in exosomes from SKOV3-IP1, HeyA8, and HeyA8-MDR cells. The high expression of miR-940 is associated with better survival in patients with ovarian serous cystadenocarcinoma. Ectopic expression of miR-940 inhibited proliferation, colony formation, invasion, and migration and triggered G0/G1 cell cycle arrest and apoptosis in OC cells. Overexpression of miR-940 also inhibited tumor cell growth in vivo. We showed that proto-oncogene tyrosine-protein kinase (SRC) is directly targeted by miR-940 and that miR-940 inhibited SRC expression at mRNA and protein levels. Following this inhibition, the expression of proteins downstream of SRC, such as FAK, paxillin and Akt was also reduced. Collectively, our results suggest that OC cells secrete the tumor-suppressive miR-940 into the extracellular environment via exosomes, to maintain their invasiveness and tumorigenic phenotype.
Collapse
Affiliation(s)
- Mohammed H Rashed
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Pinar Kanlikilicer
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martin Pichler
- Division of Clinical Oncology, Department of Medicine, Medical University of Graz, Graz, Austria
| | - Recep Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emine Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Justyna Filant
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andreia Silva
- Instituto de Investigação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Institute of Biomedical Engineering, Universidade do Porto, Porto, Portugal
| | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Merve Denizli
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rahul Mitra
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A. Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K. Sood
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed F. Abd-Ellah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Gouda K. Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Gabriel Lopez Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
329
|
Boyiadzis M, Whiteside TL. The emerging roles of tumor-derived exosomes in hematological malignancies. Leukemia 2017; 31:1259-1268. [PMID: 28321122 DOI: 10.1038/leu.2017.91] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/14/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
Exosomes are small (30-150 nm) membranous vesicles of endocytic origin produced by all cells under physiological and pathological conditions. They have recently emerged as vehicles for intercellular transfer of molecular and genetic contents from parent to recipient cells. Exosome-mediated transfer of proteins or genes (RNA, miRNA, DNA) results in reprogramming of recipient cell functions. Exosomes carry and deliver information that is essential for health, and they participate in pathological events, including malignant transformation. Within the hematopoietic system, exosomes maintain crosstalk between cells located in the bone marrow compartment and at distant tissue sites. In hematological malignancies, tumor-derived exosomes (TEX) reprogram the bone marrow environment, suppress anti-leukemia immunity, mediate drug resistance and interfere with immunotherapies. TEX are also viewed as promising biomarkers of malignant progression and as potential therapeutic targets. The involvement of TEX in nearly all aspects of malignant transformation has generated much interest in their biology, mechanisms responsible for information transfer and the role they play in cancer escape from the host immune system.
Collapse
Affiliation(s)
- M Boyiadzis
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Cancer Institute and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - T L Whiteside
- Departments of Pathology, Immunology and Otolaryngology, University of Pittsburgh Cancer Institute and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
330
|
Phinney DG, Pittenger MF. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017; 35:851-858. [PMID: 28294454 DOI: 10.1002/stem.2575] [Citation(s) in RCA: 1182] [Impact Index Per Article: 147.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/13/2016] [Accepted: 01/02/2017] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cell transplantation is undergoing extensive evaluation as a cellular therapy in human clinical trials. Because MSCs are easily isolated and amenable to culture expansion in vitro there is a natural desire to test MSCs in many diverse clinical indications. This is exemplified by the rapidly expanding literature base that includes many in vivo animal models. More recently, MSC-derived extracellular vesicles (EVs), which include exosomes and microvesicles (MV), are being examined for their role in MSC-based cellular therapy. These vesicles are involved in cell-to-cell communication, cell signaling, and altering cell or tissue metabolism at short or long distances in the body. The exosomes and MVs can influence tissue responses to injury, infection, and disease. MSC-derived exosomes have a content that includes cytokines and growth factors, signaling lipids, mRNAs, and regulatory miRNAs. To the extent that MSC exosomes can be used for cell-free regenerative medicine, much will depend on the quality, reproducibility, and potency of their production, in the same manner that these parameters dictate the development of cell-based MSC therapies. However, the MSC exosome's contents are not static, but rather a product of the MSC tissue origin, its activities and the immediate intercellular neighbors of the MSCs. As such, the exosome content produced by MSCs appears to be altered when MSCs are cultured with tumor cells or in the in vivo tumor microenvironment. Therefore, careful attention to detail in producing MSC exosomes may provide a new therapeutic paradigm for cell-free MSC-based therapies with decreased risk. Stem Cells 2017;35:851-858.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida, USA
| | | |
Collapse
|
331
|
Ferguson Bennit HR, Gonda A, Oppegard LJ, Chi DP, Khan S, Wall NR. Uptake of lymphoma-derived exosomes by peripheral blood leukocytes. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2017; 7:9-23. [PMID: 31360082 PMCID: PMC6467345 DOI: 10.2147/blctt.s130826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exosomes are nanosized lipid vesicles secreted into blood and other body fluids and serve as vehicles for intercellular communication. Despite being an important component of the tumor microenvironment (TME), exosomal targeting and uptake into recipient cells are still not fully understood. Few studies have looked at lymphoma exosomes and their interactions with circulating blood cells. In this study, we examine the exosomal uptake distribution among peripheral blood leukocytes (PBLs) using vesicles derived from a diffuse large B cell lymphoma cell line, WSU-DLCL2. Lymphoma cells survive, proliferate, and are protected from the cytotoxic effects of chemotherapeutic agents by soluble factors or by direct contact with inflammatory and stromal cells within the TME. In an attempt to close the gap in knowledge concerning lymphoma TME immunosuppression, we have treated normal human PBLs with PKH67-labeled lymphoma exosomes and monitored the uptake by measuring fluorescence at different time points using flow cytometry and fluorescent microscopy. Our results show that of the four populations examined, B cells and monocytes demonstrated uptake of PKH67-labeled exosomes, while T cells and NK cells displayed significantly less uptake.
Collapse
Affiliation(s)
- Heather R Ferguson Bennit
- Center for Health Disparities & Molecular Medicine, .,Division of Biochemistry, Department of Basic Sciences,
| | - Amber Gonda
- Center for Health Disparities & Molecular Medicine, .,Department of Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | | | - David P Chi
- Division of Biochemistry, Department of Basic Sciences,
| | - Salma Khan
- Center for Health Disparities & Molecular Medicine, .,Division of Biochemistry, Department of Basic Sciences,
| | - Nathan R Wall
- Center for Health Disparities & Molecular Medicine, .,Division of Biochemistry, Department of Basic Sciences,
| |
Collapse
|
332
|
Wendler F, Favicchio R, Simon T, Alifrangis C, Stebbing J, Giamas G. Extracellular vesicles swarm the cancer microenvironment: from tumor-stroma communication to drug intervention. Oncogene 2017; 36:877-884. [PMID: 27546617 DOI: 10.1038/onc.2016.253] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023]
Abstract
Intercellular communication sets the pace for transformed cells to survive and to thrive. Extracellular vesicles (EVs), such as exosomes, microvesicles and large oncosomes, are involved in this process shuttling reciprocal signals and other molecules between transformed and stromal cells, including fibroblasts, endothelial and immune cells. As a result, these cells are adapted or recruited to a constantly evolving cancer microenvironment. Moreover, EVs take part in the response to anticancer therapeutics not least by promoting drug resistance throughout the targeted tumor. Finally, circulating EVs can also transport important molecules to remote destinations in order to prime metastatic niches in an otherwise healthy tissue. Although the understanding of EV biology remains a major challenge in the field, their characteristics create new opportunities for advances in cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- F Wendler
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, UK
| | - R Favicchio
- Imperial College London, Department of Surgery and Cancer, Division of Cancer, London, UK
| | - T Simon
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, UK
| | - C Alifrangis
- Imperial College London, Department of Medical Oncology, NHS Trust, Hammersmith Hospital, London, UK
| | - J Stebbing
- Imperial College London, Department of Surgery and Cancer, Division of Cancer, London, UK
| | - G Giamas
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, UK
| |
Collapse
|
333
|
Dostert G, Mesure B, Menu P, Velot É. How Do Mesenchymal Stem Cells Influence or Are Influenced by Microenvironment through Extracellular Vesicles Communication? Front Cell Dev Biol 2017; 5:6. [PMID: 28224125 PMCID: PMC5293793 DOI: 10.3389/fcell.2017.00006] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/20/2017] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are widely used in cell therapy and tissue engineering thanks to their self-renewal, their multipotency, and their immunomodulatory properties that make them an attractive tool for regenerative medicine. A large part of MSCs positive effects is due to their secretion products which participate in creating a favorable microenvironment and closely relate these cells to other cell types. Extracellular vesicles (EVs) belong to cellular secretions. They are produced by cells continuously or after stimulation (e.g., calcium flux, cellular stress) and act in tissue homeostasis and intercellular communication. The understanding of the role of EVs is growing, more particularly their impact on cell migration, differentiation, or immunomodulation. EVs derived from MSCs show these interesting properties that may be considered in therapeutics, although they can have adverse effects by facilitating cancer propagation. Moreover, MSC behavior may also be influenced (proliferation, differentiation) by EVs derived from other donor cells. The aim of this mini review is to summarize the two-way communication between MSCs and other cell types, and how they can affect each other with their microenvironment through EVs. On the one hand, the manuscript presents the influence of MSC-derived EVs on diverse recipient cells and on the other hand, the effects of EVs derived from various donor cells on MSCs. The discrepancies between cancer cells and MSCs communication according to the sources of MSCs but also the tumor origins are also mentioned.
Collapse
Affiliation(s)
- Gabriel Dostert
- Laboratoire d'Ingénierie Moléculaire et Physiopathologie Articulaire, UMR 7365 Centre National de la Recherche Scientifique - Université de Lorraine, Biopôle de l'Université de Lorraine Vandœuvre-lès-Nancy, France
| | - Benjamin Mesure
- Laboratoire d'Ingénierie Moléculaire et Physiopathologie Articulaire, UMR 7365 Centre National de la Recherche Scientifique - Université de Lorraine, Biopôle de l'Université de Lorraine Vandœuvre-lès-Nancy, France
| | - Patrick Menu
- Laboratoire d'Ingénierie Moléculaire et Physiopathologie Articulaire, UMR 7365 Centre National de la Recherche Scientifique - Université de Lorraine, Biopôle de l'Université de LorraineVandœuvre-lès-Nancy, France; Faculté de Pharmacie, Université de LorraineVandœuvre-lès-Nancy, France
| | - Émilie Velot
- Laboratoire d'Ingénierie Moléculaire et Physiopathologie Articulaire, UMR 7365 Centre National de la Recherche Scientifique - Université de Lorraine, Biopôle de l'Université de LorraineVandœuvre-lès-Nancy, France; Faculté de Pharmacie, Université de LorraineVandœuvre-lès-Nancy, France
| |
Collapse
|
334
|
Abstract
Tumor extracellular vesicles (EVs), including exosomes, emerged as key drivers of the pro-tumorigenic dialog between the tumor mass and its microenvironment by mediating long and short distance communication. In vitro studies defined the capacity of tumor EVs to modify the phenotypes of stromal and tumor cells. These studies are now supported by a growing number of functional in vivo experiments. Remarkably, they allowed the identification of a new role for tumor EVs in priming the pre-metastatic niches (PMN). Several molecules transported in tumor EVs (RNAs and proteins) have recently been found to be essential for tumor progression and metastasis in vivo. In parallel, novel EV labeling and tracking strategies have very recently allowed the first descriptions of tumor EVs in vivo and pave the way for a better understanding of their function in realistic pathophysiological contexts. Here, we review the functional approaches and the recent progress in in vivo imaging of EVs, which have refined our understanding of the role played by tumor EVs. Finally, we emphasize the remaining challenges and open questions related to the biology of tumor EVs.
Collapse
Affiliation(s)
- Vincent Hyenne
- a Inserm U1109, MN3T , Strasbourg , France.,b Université de Strasbourg , Strasbourg , France.,c LabEx Medalis, Université de Strasbourg , Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) , Strasbourg , France.,e CNRS SNC5055 , Strasbourg , France
| | - Olivier Lefebvre
- a Inserm U1109, MN3T , Strasbourg , France.,b Université de Strasbourg , Strasbourg , France.,c LabEx Medalis, Université de Strasbourg , Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) , Strasbourg , France
| | - Jacky G Goetz
- a Inserm U1109, MN3T , Strasbourg , France.,b Université de Strasbourg , Strasbourg , France.,c LabEx Medalis, Université de Strasbourg , Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) , Strasbourg , France
| |
Collapse
|
335
|
Park Y. MicroRNA Exocytosis by Vesicle Fusion in Neuroendocrine Cells. Front Endocrinol (Lausanne) 2017; 8:355. [PMID: 29312145 PMCID: PMC5743741 DOI: 10.3389/fendo.2017.00355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that posttranscriptionally regulate gene expression inside the cell. Extracellular circulating miRNAs are also observed outside the cell, but their origin is poorly understood. Recently, miRNA has been shown to be exocytosed by vesicle fusion; this observation demonstrates that vesicle-free miRNAs are secreted from neuroendocrine cells, in a manner similar to hormone secretion. miRNAs are stored in large dense-core vesicles together with catecholamines, then released by vesicle fusion in response to stimulation; in this way, vesicle-free miRNA may regulate cell-to-cell communication including the regulation of gene expression and cellular signaling. Therefore, miRNA has been suggested to function as a hormone; i.e., a ribomone (ribonucleotide + hormone). This review focuses on the mechanisms by which vesicle-free miRNAs are secreted from neuroendocrine cells and will discuss potential functions of vesicle-free miRNAs and how vesicle-free miRNAs regulate cell-to-cell communication.
Collapse
Affiliation(s)
- Yongsoo Park
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
- *Correspondence: Yongsoo Park,
| |
Collapse
|
336
|
Bruns H, Böttcher M, Qorraj M, Fabri M, Jitschin S, Dindorf J, Busch L, Jitschin R, Mackensen A, Mougiakakos D. CLL-cell-mediated MDSC induction by exosomal miR-155 transfer is disrupted by vitamin D. Leukemia 2016; 31:985-988. [PMID: 28008175 DOI: 10.1038/leu.2016.378] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- H Bruns
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - M Böttcher
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - M Qorraj
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - M Fabri
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - S Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - J Dindorf
- Department of Dermatology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - L Busch
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - R Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - A Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - D Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
337
|
Exosomes promote bone marrow angiogenesis in hematologic neoplasia: the role of hypoxia. Curr Opin Hematol 2016; 23:268-73. [PMID: 26866730 DOI: 10.1097/moh.0000000000000235] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW To review the data on angiogenesis related to exosomes secreted by tumor cells in hematologic neoplasia and to elucidate the role of exosomes and exosomal miRNA in the bone marrow microenvironment, especially under hypoxic conditions. RECENT FINDINGS Cross-talk between bone marrow tumor cells and surrounding cells, including endothelial cells, is important for tumor growth in hematologic neoplasia. In addition to conventional signaling pathways, exosomes, which are small endosome-derived vesicles containing miRNAs, can help to modulate the microenvironment without directly contacting nontumorous cells. The human myeloid leukemia cell line K562 secretes exosomes containing a large amount of miR-92a that enhances angiogenesis under normoxic and hypoxic conditions. With chronic hypoxia, exosomes secreted by multiple myeloma cells also enhance angiogenesis by targeting factor-inhibiting hypoxia-inducible factor-1 via miR-135b. SUMMARY Intercellular communication between tumor cells and a heterogeneous population of bone marrow stromal cells is mediated by exosomes containing various functional proteins, mRNA, and miRNA. Hypoxia is a major regulator of exosomal content and affects angiogenesis in various types of hematologic neoplasia. Functional analysis of exosomes and exosome-mediated cell-cell interactions not only clarifies molecular pathogenesis but also suggests new treatment strategies for hematologic neoplasia through targeting exosomes.
Collapse
|
338
|
Alonso S, Hernandez D, Chang YT, Gocke CB, McCray M, Varadhan R, Matsui WH, Jones RJ, Ghiaur G. Hedgehog and retinoid signaling alters multiple myeloma microenvironment and generates bortezomib resistance. J Clin Invest 2016; 126:4460-4468. [PMID: 27775549 PMCID: PMC5127663 DOI: 10.1172/jci88152] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/15/2016] [Indexed: 01/08/2023] Open
Abstract
Interactions between multiple myeloma (MM) cells and the BM microenvironment play a critical role in bortezomib (BTZ) resistance. However, the mechanisms involved in these interactions are not completely understood. We previously showed that expression of CYP26 in BM stromal cells maintains a retinoic acid-low (RA-low) microenvironment that prevents the differentiation of normal and malignant hematopoietic cells. Since a low secretory B cell phenotype is associated with BTZ resistance in MM and retinoid signaling promotes plasma cell differentiation and Ig production, we investigated whether stromal expression of the cytochrome P450 monooxygenase CYP26 modulates BTZ sensitivity in the BM niche. CYP26-mediated inactivation of RA within the BM microenvironment prevented plasma cell differentiation and promoted a B cell-like, BTZ-resistant phenotype in human MM cells that were cocultured on BM stroma. Moreover, paracrine Hedgehog secretion by MM cells upregulated stromal CYP26 and further reinforced a protective microenvironment. These results suggest that crosstalk between Hedgehog and retinoid signaling modulates BTZ sensitivity in the BM niche. Targeting these pathological interactions holds promise for eliminating minimal residual disease in MM.
Collapse
|
339
|
Song YH, Warncke C, Choi SJ, Choi S, Chiou AE, Ling L, Liu HY, Daniel S, Antonyak MA, Cerione RA, Fischbach C. Breast cancer-derived extracellular vesicles stimulate myofibroblast differentiation and pro-angiogenic behavior of adipose stem cells. Matrix Biol 2016; 60-61:190-205. [PMID: 27913195 DOI: 10.1016/j.matbio.2016.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/12/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
Adipose-derived stem cells (ASCs) are abundantly present in the mammary microenvironment and can promote breast cancer malignancy by differentiating into myofibroblasts. However, it remains largely unclear which role tumor-derived extracellular vesicles (TEVs) play in this process. Here, we used microfabricated, type I collagen-based 3-D tissue culture platforms to investigate the effect of breast cancer cell-derived TEVs on ASCs myofibroblast differentiation and consequential changes in extracellular matrix remodeling and vascular sprouting. TEVs collected from MDA MB-231 human metastatic breast cancer cells (MDAs) promoted ASC myofibroblast differentiation in both 2-D and 3-D cultures as indicated by increased alpha smooth muscle actin (α-SMA) and fibronectin (Fn) levels. Correspondingly, TEV-treated ASCs were more contractile, secreted more vascular endothelial growth factor (VEGF), and promoted angiogenic sprouting of human umbilical vein endothelial cells (HUVECs). These changes were dependent on transforming growth factor beta (TGF-β)-related signaling and tumor cell glutaminase activity as their inhibition decreased TEV-related myofibroblastic differentiation of ASCs and related functional consequences. In summary, our data suggest that TEVs are important signaling factors that contribute to ASC desmoplastic reprogramming in the tumor microenvironment, and suggest that tumor cell glutamine metabolism may be used as a therapeutic target to interfere with this process.
Collapse
Affiliation(s)
- Young Hye Song
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Christine Warncke
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Sung Jin Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Aaron E Chiou
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Han-Yuan Liu
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| | - Marc A Antonyak
- Department of Molecular Medicine, Cornell University, Ithaca, NY, United States
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY, United States; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, United States
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
340
|
Reiners KS, Shatnyeva O, Vasyutina E, Bösl T, Hansen HP, Hallek M, Herling M, von Strandmann EP. Extracellular vesicles released from chronic lymphocytic leukemia cells exhibit a disease relevant mRNA signature and transfer mRNA to bystander cells. Haematologica 2016; 102:e100-e103. [PMID: 27856511 DOI: 10.3324/haematol.2016.153197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Katrin S Reiners
- Department I of Internal Medicine, University Hospital of Cologne, Germany
| | - Olga Shatnyeva
- Department I of Internal Medicine, University Hospital of Cologne, Germany
| | - Elena Vasyutina
- Department I of Internal Medicine, University Hospital of Cologne, Germany.,Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases, CECAD, Germany
| | - Teresa Bösl
- Department I of Internal Medicine, University Hospital of Cologne, Germany
| | - Hinrich P Hansen
- Department I of Internal Medicine, University Hospital of Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University Hospital of Cologne, Germany.,Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases, CECAD, Germany.,Center of Integrated Oncology Köln Bonn, CIO, Germany
| | - Marco Herling
- Department I of Internal Medicine, University Hospital of Cologne, Germany.,Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases, CECAD, Germany.,Center of Integrated Oncology Köln Bonn, CIO, Germany
| | - Elke Pogge von Strandmann
- Department I of Internal Medicine, University Hospital of Cologne, Germany .,Clinic for Hematology, Oncology and Immunology, Experimental Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
341
|
Poggi A, Giuliani M. Mesenchymal Stromal Cells Can Regulate the Immune Response in the Tumor Microenvironment. Vaccines (Basel) 2016; 4:E41. [PMID: 27834810 PMCID: PMC5192361 DOI: 10.3390/vaccines4040041] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/01/2016] [Accepted: 10/31/2016] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment is a good target for therapy in solid tumors and hematological malignancies. Indeed, solid tumor cells' growth and expansion can influence neighboring cells' behavior, leading to a modulation of mesenchymal stromal cell (MSC) activities and remodeling of extracellular matrix components. This leads to an altered microenvironment, where reparative mechanisms, in the presence of sub-acute inflammation, are not able to reconstitute healthy tissue. Carcinoma cells can undergo epithelial mesenchymal transition (EMT), a key step to generate metastasis; these mesenchymal-like cells display the functional behavior of MSC. Furthermore, MSC can support the survival and growth of leukemic cells within bone marrow participating in the leukemic cell niche. Notably, MSC can inhibit the anti-tumor immune response through either carcinoma-associated fibroblasts or bone marrow stromal cells. Experimental data have indicated their relevance in regulating cytolytic effector lymphocytes of the innate and adaptive arms of the immune system. Herein, we will discuss some of the evidence in hematological malignancies and solid tumors. In particular, we will focus our attention on the means by which it is conceivable to inhibit MSC-mediated immune suppression and trigger anti-tumor innate immunity.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy.
| | - Massimo Giuliani
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City L-1526, Luxembourg.
| |
Collapse
|
342
|
Bell E, Taylor MA. Functional Roles for Exosomal MicroRNAs in the Tumour Microenvironment. Comput Struct Biotechnol J 2016; 15:8-13. [PMID: 27872688 PMCID: PMC5109280 DOI: 10.1016/j.csbj.2016.10.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022] Open
Abstract
Extracellular microRNAs are released from cells both passively and actively. The presence of these microRNAs in the tumour microenvironment (TME) can significantly impact on the plasticity of cancer cells leading to the promotion of metastatic and angiogenic processes. These extracellular microRNAs can act not only on other cancer cells, but also cells present in the TME, such as immune cells, endothelial cells, fibroblasts, and others acting to subvert the host immune system and drive tumour progression. In this review we highlight the current understanding of both the mechanisms by which microRNAs are released from tumour cells and the downstream functional effects that extracellular microRNAs have on recipient cells.
Collapse
Affiliation(s)
- Emma Bell
- AstraZeneca, Personalised Healthcare and Biomarkers, iMed, Cambridge Science Park, Cambridge CB4 0FZ, United Kingdom
| | - Molly A. Taylor
- AstraZeneca, Oncology iMed Bioscience, Cambridge CB2 0RE, United Kingdom
- Corresponding author at: AstraZeneca, Oncology iMed Bioscience, Lab 140, CRUK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, United Kingdom.AstraZenecaOncology iMed BioscienceCRUK Cambridge InstituteLab 140Robinson WayCambridgeCB2 0REUnited Kingdom
| |
Collapse
|
343
|
Caspase-3-dependent cleavage of Bcl-xL in the stroma exosomes is required for their uptake by hematological malignant cells. Blood 2016; 128:2655-2665. [PMID: 27742710 DOI: 10.1182/blood-2016-05-715961] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/05/2016] [Indexed: 12/31/2022] Open
Abstract
The intercellular crosstalk between hematological malignancies and the tumor microenvironment is mediated by cell-to-cell interactions and soluble factors. One component of the secretome that is gaining increasing attention is the extracellular vesicles and, in particular, the exosomes. Apart from the role as vectors of molecular information, exosomes have been shown to possess intrinsic biological activity. In this study, we found that caspase-3 is activated in L88 bone marrow stroma cell-derived exosomes and identified 1 of the substrates to be the antiapoptotic protein Bcl-xL. The cleaved Bcl-xL is found in a panel of normal and cancer cell-derived exosomes and is localized on the outer leaflet of the exosomal membrane. Incubation of the exosomes with a caspase-3 inhibitor or the pan-caspase inhibitor prevents the cleavage of Bcl-xL. Importantly, MCF-7 cell-derived exosomes that are caspase-3-deficient are enriched in full-length Bcl-xL, whereas ectopic expression of caspase-3 restores the cleavage of Bcl-xL. Chemical inhibition of Bcl-xL with ABT737 or molecular inhibition by using the D61A and D76A Bcl-xL mutant leads to a significant decrease in the uptake of exosomes by hematopoietic malignant cells. These data indicate that the cleaved Bcl-xL is required for the uptake of exosomes by myeloma and lymphoma cells, leading to their increased proliferation. In summary, we demonstrate for the first time that Bcl-xL is an exosomal caspase-3 substrate and that this processing is required for the uptake of exosomes by recipient cells.
Collapse
|
344
|
De Luca L, D'Arena G, Simeon V, Trino S, Laurenzana I, Caivano A, La Rocca F, Villani O, Mansueto G, Deaglio S, Innocenti I, Laurenti L, Molica S, Pietrantuono G, De Stradis A, Del Vecchio L, Musto P. Characterization and prognostic relevance of circulating microvesicles in chronic lymphocytic leukemia. Leuk Lymphoma 2016; 58:1424-1432. [PMID: 27739922 DOI: 10.1080/10428194.2016.1243790] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Microvescicles (MV) are shedding particles released by normal and neoplastic cells, whose levels in biological fluids highlight their potential role as disease biomarkers and therapeutic targets. By analyzing 131 newly diagnosed chronic lymphocytic leukemia (CLL), we found that the absolute number of serum CLL MV was significantly higher than in controls, in particular in advanced stages of disease. In addition, CD19 + and CD37+, B-cell derived MV, significantly correlated with high tumor burden. Absolute MV number cutoff selected by ROC analysis distinguished Rai stage 0 patients with shorter time to treatment (TTT) from those with more stable disease. Likewise, in the entire cohort, two groups of patients with different overall survival (OS) and different TTT were identified. At multivariate analysis, serum MV independently predicted for OS (along with Rai stage) and TTT (along with Rai stage, lymphocytes and CD38). In conclusion, circulating MV represent a new potential prognostic biomarker in CLL.
Collapse
Affiliation(s)
- Luciana De Luca
- a Laboratory of Preclinical and Translational Research , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Giovanni D'Arena
- b Department of Onco-Hematology , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Vittorio Simeon
- a Laboratory of Preclinical and Translational Research , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Stefania Trino
- a Laboratory of Preclinical and Translational Research , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Ilaria Laurenzana
- a Laboratory of Preclinical and Translational Research , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Antonella Caivano
- a Laboratory of Preclinical and Translational Research , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Francesco La Rocca
- a Laboratory of Preclinical and Translational Research , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Oreste Villani
- b Department of Onco-Hematology , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Giovanna Mansueto
- b Department of Onco-Hematology , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Silvia Deaglio
- c Department of Medical Sciences , University of Turin and Human Genetics Foundation (HuGeF) , Turin , Italy
| | - Idanna Innocenti
- d Department of Hematology , Catholic University of Sacred Heart , Rome , Italy
| | - Luca Laurenti
- d Department of Hematology , Catholic University of Sacred Heart , Rome , Italy
| | - Stefano Molica
- e Department of Hematology , Ciaccio-Pugliese Hospital , Catanzaro , Italy
| | - Giuseppe Pietrantuono
- b Department of Onco-Hematology , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| | - Angelo De Stradis
- f Institute for Sustainable Plant Protection, National Research Council of Italy, University of Bari , Bari , Italy
| | - Luigi Del Vecchio
- g Department of Molecular Medicine and Medical Biotechnologies , Federico II University , Naples , Italy.,h CEINGE-Biotecnologie Avanzate S.C.a R.L , Naples , Italy
| | - Pellegrino Musto
- i Scientific Direction , IRCCS Referral Cancer Center of Basilicata (CROB) , Rionero in Vulture , PZ , Italy
| |
Collapse
|
345
|
Marks DL, Olson RL, Fernandez-Zapico ME. Epigenetic control of the tumor microenvironment. Epigenomics 2016; 8:1671-1687. [PMID: 27700179 DOI: 10.2217/epi-2016-0110] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stromal cells of the tumor microenvironment have been shown to play important roles in both supporting and limiting cancer growth. The altered phenotype of tumor-associated stromal cells (fibroblasts, immune cells, endothelial cells etc.) is proposed to be mainly due to epigenetic dysregulation of gene expression; however, only limited studies have probed the roles of epigenetic mechanisms in the regulation of stromal cell function. We review recent studies demonstrating how specific epigenetic mechanisms (DNA methylation and histone post-translational modification-based gene expression regulation, and miRNA-mediated translational regulation) drive aspects of stromal cell phenotype, and discuss the implications of these findings for treatment of malignancies. We also summarize the effects of epigenetic mechanism-targeted drugs on stromal cells and discuss the consideration of the microenvironment response in attempts to use these drugs for cancer treatment.
Collapse
Affiliation(s)
- David L Marks
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Rachel Lo Olson
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.,University of Minnesota Rochester, Rochester, MN 55904, USA
| | | |
Collapse
|
346
|
Matsuda A, Yan IK, Foye C, Parasramka M, Patel T. MicroRNAs as paracrine signaling mediators in cancers and metabolic diseases. Best Pract Res Clin Endocrinol Metab 2016; 30:577-590. [PMID: 27923452 PMCID: PMC5147504 DOI: 10.1016/j.beem.2016.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The contribution of microRNAs to the regulation of mRNA expression during physiological and developmental processes are well-recognized. These roles are being expanded by recent observations that emphasize the capability of miRNA to participate in inter-cellular signaling and communication. Several factors support a functional role for miRNA as mediators of cell-to-cell signaling. miRNA are able to exist within the extracellular milieu or circulation, and their stability and integrity maintained through association with binding proteins or lipoproteins, or through encapsulation within cell-derived membrane vesicles. Furthermore, miRNA can retain functionality and regulate target gene expression following their uptake by recipient cells. In this overview, we review specific examples that will highlight the potential of miRNA to serve as paracrine signaling mediators in metabolic diseases and cancers. Elucidating the mechanisms involved in inter-cellular communication involving miRNA will provide new insights into disease pathogenesis and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Akiko Matsuda
- Departments of Transplantation and Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Irene K Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Catherine Foye
- Departments of Transplantation and Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Mansi Parasramka
- Departments of Transplantation and Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA.
| |
Collapse
|
347
|
Fu H, Yang H, Zhang X, Xu W. The emerging roles of exosomes in tumor-stroma interaction. J Cancer Res Clin Oncol 2016; 142:1897-907. [PMID: 26987524 DOI: 10.1007/s00432-016-2145-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/09/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE The tumor-stroma interaction is critical for the development and progression of cancer. Cancer-associated fibroblasts (CAFs), one of the major components of the tumor stroma, can promote tumor growth and metastasis. Exosomes are secreted microvesicles that mediate cell-to-cell communication. Exosomal contents, including proteins, nucleic acids, and lipids, can be shuttled from donor cells to target cells. Recent studies suggest that exosomes play important roles in the tumor-stroma interaction. Herein, we review the multifaceted roles of exosomes in the tumor-stroma interaction and the underlying molecular mechanisms. METHODS Literature search for all relevant publications was performed on PubMed databases. The keywords of exosomes, tumor, stroma, CAFs, mesenchymal stem cells (MSCs) and other closely related terms were used for searching. RESULTS Tumor cell-derived exosomes induce the differentiation of fibroblasts and MSCs into CAFs. In turn, exosomes secreted by CAFs promote tumor growth, metastasis, and drug resistance through distinct mechanisms. Moreover, exosomes from stromal cells can be used as therapeutic vehicles for the delivery of anticancer drugs. CONCLUSIONS Tumor cells communicate with CAFs through exosomes, which establishes a bidirectional cross talk to promote tumor growth, metastasis, and drug resistance. Targeting exosomes in tumor-stroma interaction may have important implications for anticancer therapy.
Collapse
Affiliation(s)
- Hailong Fu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Huan Yang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
- The Affiliated Hospital, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
348
|
Wu L, Zhang X, Zhang B, Shi H, Yuan X, Sun Y, Pan Z, Qian H, Xu W. Exosomes derived from gastric cancer cells activate NF-κB pathway in macrophages to promote cancer progression. Tumour Biol 2016; 37:12169-12180. [PMID: 27220495 DOI: 10.1007/s13277-016-5071-5] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/05/2016] [Indexed: 01/25/2023] Open
Abstract
Exosomes are nano-sized membrane vesicles secreted by both normal and cancer cells. Emerging evidence indicates that cancer cells derived exosomes contribute to cancer progression through the modulation of tumor microenvironment. However, the effects of exosomes derived from gastric cancer cells on macrophages are not well understood. In this study, we investigated the biological role of gastric cancer cells derived exosomes in the activation of macrophages. We demonstrated that gastric cancer cells derived exosomes activated macrophages to express increased levels of proinflammatory factors, which in turn promoted tumor cell proliferation and migration. In addition, gastric cancer cells derived exosomes remarkably upregulated the phosphorylation of NF-κB in macrophages. Inhibiting the activation of NF-κB reversed the upregulation of proinflammatory factors in macrophages and blocked their promoting effects on gastric cancer cells. Moreover, we found that gastric cancer cells derived exosomes could also activate macrophages from human peripheral blood monocytes through the activation of NF-κB. In conclusion, our results suggest that gastric cancer cells derived exosomes stimulate the activation of NF-κB pathway in macrophages to promote cancer progression, which provides a potential therapeutic approach for gastric cancer by interfering with the interaction between exosomes and macrophages in tumor microenvironment.
Collapse
Affiliation(s)
- Lijun Wu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Bin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Xiao Yuan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Yaoxiang Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Zhaoji Pan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
349
|
Geyer MB, Brentjens RJ. Review: Current clinical applications of chimeric antigen receptor (CAR) modified T cells. Cytotherapy 2016; 18:1393-1409. [PMID: 27592405 DOI: 10.1016/j.jcyt.2016.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/27/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
The past several years have been marked by extraordinary advances in clinical applications of immunotherapy. In particular, adoptive cellular therapy utilizing chimeric antigen receptor (CAR)-modified T cells targeted to CD19 has demonstrated substantial clinical efficacy in children and adults with relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL) and durable clinical benefit in a smaller subset of patients with relapsed or refractory chronic lymphocytic leukemia (CLL) or B-cell non-Hodgkin lymphoma (B-NHL). Early-phase clinical trials are currently assessing CAR T-cell safety and efficacy in additional malignancies. Here, we discuss clinical results from the largest series to date investigating CD19-targeted CAR T cells in B-ALL, CLL, and B-NHL, including discussion of differences in CAR T-cell design and production and treatment approach, as well as clinical efficacy, nature of severe cytokine release syndrome and neurologic toxicities, and CAR T-cell expansion and persistence. We additionally review the current and forthcoming use of CAR T cells in multiple myeloma and several solid tumors and highlight challenges and opportunities afforded by the current state of CAR T-cell therapies, including strategies to overcome inhibitory aspects of the tumor microenvironment and enhance antitumor efficacy.
Collapse
Affiliation(s)
- Mark B Geyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
350
|
Naito Y, Yoshioka Y, Yamamoto Y, Ochiya T. How cancer cells dictate their microenvironment: present roles of extracellular vesicles. Cell Mol Life Sci 2016; 74:697-713. [PMID: 27582126 PMCID: PMC5272899 DOI: 10.1007/s00018-016-2346-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/16/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022]
Abstract
Intercellular communication plays an important role in cancer initiation and progression through secretory molecules, including growth factors and cytokines. Recent advances have revealed that small membrane vesicles, termed extracellular vesicles (EVs), served as a regulatory agent in the intercellular communication of cancer. EVs enable the transfer of functional molecules, including proteins, mRNA and microRNAs (miRNAs), into recipient cells. Cancer cells utilize EVs to dictate the unique phenotype of surrounding cells, thereby promoting cancer progression. Against such "education" by cancer cells, non-tumoral cells suppress cancer initiation and progression via EVs. Therefore, researchers consider EVs to be important cues to clarify the molecular mechanisms of cancer biology. Understanding the functions of EVs in cancer progression is an important aspect of cancer biology that has not been previously elucidated. In this review, we summarize experimental data that indicate the pivotal roles of EVs in cancer progression.
Collapse
Affiliation(s)
- Yutaka Naito
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yusuke Yoshioka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yusuke Yamamoto
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|