51
|
Gopal SK, Greening DW, Hanssen EG, Zhu HJ, Simpson RJ, Mathias RA. Oncogenic epithelial cell-derived exosomes containing Rac1 and PAK2 induce angiogenesis in recipient endothelial cells. Oncotarget 2017; 7:19709-22. [PMID: 26919098 PMCID: PMC4991413 DOI: 10.18632/oncotarget.7573] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/06/2016] [Indexed: 01/06/2023] Open
Abstract
The metastatic cascade describes the escape of primary tumour cells to distant secondary sites. Cells at the leading tumour edge are thought to undergo epithelial-mesenchymal transition (EMT), to enhance their motility and invasion for spreading. Whether EMT cells directly promote tumour angiogenesis, and the role of exosomes (30-150 nm extracellular vesicles) remains largely unknown. We examined the functional effects of exosomes from MDCK cells, MDCK cells stably expressing YBX1 (MDCKYBX1, intermediate EMT), and Ras-transformed MDCK cells (21D1 cells, complete EMT). 2F-2B cell motility and tube formation (length and branching) was significantly increased following supplementation with MDCKYBX1 or 21D1 exosomes, but not MDCK exosomes. Next, Matrigel™ plugs containing exosome-supplemented 2F-2B cells were subcutaneously injected into mice. Systemic perfusion was only observed for plugs supplemented with MDCKYBX1 or 21D1 exosomes. Comparative proteomics revealed that 21D1 exosomes contained VEGF-associated proteins, while MDCKYBX1 exosomes were enriched with activated Rac1 and PAK2. To validate, 2F-2B cells and HUVECs were pre-treated with PAK inhibitors prior to exosome supplementation. PAK inhibition nullified the effects of MDCKYBX1 exosomes by reducing the tube length and branching to baseline levels. By contrast, the effects of 21D1 exosomes were not significantly decreased. Our results demonstrate for the first time that oncogenic cells undergoing EMT can communicate with endothelial cells via exosomes, and establish exosomal Rac1/PAK2 as angiogenic promoters that may function from early stages of the metastatic cascade.
Collapse
Affiliation(s)
- Shashi K Gopal
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Eric G Hanssen
- Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hong-Jian Zhu
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Rommel A Mathias
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
52
|
Sharma A. Role of stem cell derived exosomes in tumor biology. Int J Cancer 2017; 142:1086-1092. [PMID: 28983919 DOI: 10.1002/ijc.31089] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/09/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
Abstract
Exosomes are nano-scale messengers loaded with bio-molecular cargo of RNA, DNA, and Proteins. As a master regulator of cellular signaling, stem cell (both normal, and cancer stem cells) secreted exosome orchestrate various autocrine and paracrine functions which alter tumor micro-environment, growth and progression. Exosomes secreted by one of the two important stem cell phenotypes in cancers a) Mesenchymal stem cells, and b) Cancer stem cells not only promote cancerous growth but also impart therapy resistance in cancer cells. In tumors, normal or mesenchymal stem cell (MSCs) derived exosomes (MSC-exo) modulate tumor hallmarks by delivering unique miRNA species to neighboring cells and help in tumor progression. Apart from regulating tumor cell fate, MSC-exo are also capable of inducing physiological processes, for example, angiogenesis, metastasis and so forth. Similarly, cancer stem cells (CSCs) derived exosomes (CSC-exo) contain stemness-specific proteins, self-renewal promoting regulatory miRNAs, and survival factors. CSC-exo specific cargo maintains tumor heterogeneity and alters tumor progression. In this review we critically discuss the importance of stem cell specific exosomes in tumor cell signaling pathways with their role in tumor biology.
Collapse
Affiliation(s)
- Aman Sharma
- ExoCan Healthcare Technologies Pvt Ltd, L4, 400 NCL Innovation Park, Dr Homi Bhabha Road, Pune, 411008, India
| |
Collapse
|
53
|
Xuesong D, Wei X, Heng L, Xiao C, Shunan W, Yu G, Weiguo Z. Evaluation of neovascularization patterns in an orthotopic rat glioma model with dynamic contrast-enhanced MRI. Acta Radiol 2017; 58:1138-1146. [PMID: 27956462 DOI: 10.1177/0284185116681038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has been proved useful in evaluating glioma angiogenesis, but the utility in evaluating neovascularization patterns has not been reported. Purpose To evaluate in vivo real-time glioma neovascularization patterns by measuring glioma perfusion quantitatively using DCE-MRI. Material and Methods Thirty Sprague-Dawley rats were used to establish C6 orthotopic glioma model and underwent MRI and pathology detections. As MRI and pathology were performed at six time points (i.e. 4, 8, 12, 16, 20, and 24 days) post transplantation, neovascularization patterns were evaluated via DCE-MRI. Results Four neovascularization patterns were observed in glioma tissues. Sprout angiogenesis and intussusceptive microvascular growth located inside tumor, while vascular co-option and vascular mimicry were found in the tumor margin and necrotic area, respectively. Sprout angiogenesis and intussusceptive microvascular growth increased with Ktrans, Kep, and Vp inside tumor tissue. In addition, Kep and Vp were positively correlated with sprout angiogenesis and intussusceptive microvascular growth. Vascular co-option was decreased at 12 and 16 days post transplantation and correlated negatively with Ktrans and Kep detected in the glioma margin, respectively. Changes of vascular mimicry showed no significant statistical difference at the six time points. Conclusion Our results indicate that DCE-MRI can evaluate neovascularization patterns in a glioma model. Furthermore, DCE-MRI could be an imaging biomarker for guidance of antiangiogenic treatments in humans in the future.
Collapse
Affiliation(s)
- Du Xuesong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Xue Wei
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Liu Heng
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Chen Xiao
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Wang Shunan
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Guo Yu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Zhang Weiguo
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
54
|
Tian DY, Jin XR, Zeng X, Wang Y. Notch Signaling in Endothelial Cells: Is It the Therapeutic Target for Vascular Neointimal Hyperplasia? Int J Mol Sci 2017; 18:ijms18081615. [PMID: 28757591 PMCID: PMC5578007 DOI: 10.3390/ijms18081615] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 01/09/2023] Open
Abstract
Blood vessels respond to injury through a healing process that includes neointimal hyperplasia. The vascular endothelium is a monolayer of cells that separates the outer vascular wall from the inner circulating blood. The disruption and exposure of endothelial cells (ECs) to subintimal components initiate the neointimal formation. ECs not only act as a highly selective barrier to prevent early pathological changes of neointimal hyperplasia, but also synthesize and release molecules to maintain vascular homeostasis. After vascular injury, ECs exhibit varied responses, including proliferation, regeneration, apoptosis, phenotypic switching, interacting with other cells by direct contact or secreted molecules and the change of barrier function. This brief review presents the functional role of the evolutionarily-conserved Notch pathway in neointimal hyperplasia, notably by regulating endothelial cell functions (proliferation, regeneration, apoptosis, differentiation, cell-cell interaction). Understanding endothelial cell biology should help us define methods to prompt cell proliferation, prevent cell apoptosis and dysfunction, block neointimal hyperplasia and vessel narrowing.
Collapse
Affiliation(s)
- Ding-Yuan Tian
- Trainee Brigade, Third Military Medical University, Chongqing 400038, China.
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| | - Xu-Rui Jin
- Trainee Brigade, Third Military Medical University, Chongqing 400038, China.
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| | - Xi Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| | - Yun Wang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
55
|
Klingeborn M, Dismuke WM, Bowes Rickman C, Stamer WD. Roles of exosomes in the normal and diseased eye. Prog Retin Eye Res 2017; 59:158-177. [PMID: 28465248 PMCID: PMC5537591 DOI: 10.1016/j.preteyeres.2017.04.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 12/21/2022]
Abstract
Exosomes are nanometer-sized vesicles that are released by cells in a controlled fashion and mediate a plethora of extra- and intercellular activities. Some key functions of exosomes include cell-cell communication, immune modulation, extracellular matrix turnover, stem cell division/differentiation, neovascularization and cellular waste removal. While much is known about their role in cancer, exosome function in the many specialized tissues of the eye is just beginning to undergo rigorous study. Here we review current knowledge of exosome function in the visual system in the context of larger bodies of data from other fields, in both health and disease. Additionally, we discuss recent advances in the exosome field including use of exosomes as a therapeutic vehicle, exosomes as a source of biomarkers for disease, plus current standards for isolation and validation of exosome populations. Finally, we use this foundational information about exosomes in the eye as a platform to identify areas of opportunity for future research studies.
Collapse
Affiliation(s)
- Mikael Klingeborn
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27710, USA
| | - W Michael Dismuke
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27710, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27710, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
56
|
Extracellular vesicles: their role in cancer biology and epithelial-mesenchymal transition. Biochem J 2017; 474:21-45. [PMID: 28008089 DOI: 10.1042/bcj20160006] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 10/04/2016] [Accepted: 10/10/2016] [Indexed: 12/31/2022]
Abstract
Cell-cell communication is critical across an assortment of physiological and pathological processes. Extracellular vesicles (EVs) represent an integral facet of intercellular communication largely through the transfer of functional cargo such as proteins, messenger RNAs (mRNAs), microRNA (miRNAs), DNAs and lipids. EVs, especially exosomes and shed microvesicles, represent an important delivery medium in the tumour micro-environment through the reciprocal dissemination of signals between cancer and resident stromal cells to facilitate tumorigenesis and metastasis. An important step of the metastatic cascade is the reprogramming of cancer cells from an epithelial to mesenchymal phenotype (epithelial-mesenchymal transition, EMT), which is associated with increased aggressiveness, invasiveness and metastatic potential. There is now increasing evidence demonstrating that EVs released by cells undergoing EMT are reprogrammed (protein and RNA content) during this process. This review summarises current knowledge of EV-mediated functional transfer of proteins and RNA species (mRNA, miRNA, long non-coding RNA) between cells in cancer biology and the EMT process. An in-depth understanding of EVs associated with EMT, with emphasis on molecular composition (proteins and RNA species), will provide fundamental insights into cancer biology.
Collapse
|
57
|
Vu TQ, de Castro RMB, Qin L. Bridging the gap: microfluidic devices for short and long distance cell-cell communication. LAB ON A CHIP 2017; 17:1009-1023. [PMID: 28205652 PMCID: PMC5473339 DOI: 10.1039/c6lc01367h] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cell-cell communication is a crucial component of many biological functions. For example, understanding how immune cells and cancer cells interact, both at the immunological synapse and through cytokine secretion, can help us understand and improve cancer immunotherapy. The study of how cells communicate and form synaptic connections is important in neuroscience, ophthalmology, and cancer research. But in order to increase our understanding of these cellular phenomena, better tools need to be developed that allow us to study cell-cell communication in a highly controlled manner. Some technical requirements for better communication studies include manipulating cells spatiotemporally, high resolution imaging, and integrating sensors. Microfluidics is a powerful platform that has the ability to address these requirements and other current limitations. In this review, we describe some new advances in microfluidic technologies that have provided researchers with novel methods to study intercellular communication. The advantages of microfluidics have allowed for new capabilities in both single cell-cell communication and population-based communication. This review highlights microfluidic communication devices categorized as "short distance", or primarily at the single cell level, and "long distance", which mostly encompasses population level studies. Future directions and translation/commercialization will also be discussed.
Collapse
Affiliation(s)
- Timothy Quang Vu
- Department of Bioengineering, Rice University, Houston, TX 77030, USA and Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Ricardo Miguel Bessa de Castro
- College of Engineering, Swansea University Singleton Park, Swansea, UK and Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA. and Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
58
|
Wieland E, Rodriguez-Vita J, Liebler SS, Mogler C, Moll I, Herberich SE, Espinet E, Herpel E, Menuchin A, Chang-Claude J, Hoffmeister M, Gebhardt C, Brenner H, Trumpp A, Siebel CW, Hecker M, Utikal J, Sprinzak D, Fischer A. Endothelial Notch1 Activity Facilitates Metastasis. Cancer Cell 2017; 31:355-367. [PMID: 28238683 DOI: 10.1016/j.ccell.2017.01.007] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 09/28/2016] [Accepted: 01/25/2017] [Indexed: 12/11/2022]
Abstract
Endothelial cells (ECs) provide angiocrine factors orchestrating tumor progression. Here, we show that activated Notch1 receptors (N1ICD) are frequently observed in ECs of human carcinomas and melanoma, and in ECs of the pre-metastatic niche in mice. EC N1ICD expression in melanoma correlated with shorter progression-free survival. Sustained N1ICD activity induced EC senescence, expression of chemokines and the adhesion molecule VCAM1. This promoted neutrophil infiltration, tumor cell (TC) adhesion to the endothelium, intravasation, lung colonization, and postsurgical metastasis. Thus, sustained vascular Notch signaling facilitates metastasis by generating a senescent, pro-inflammatory endothelium. Consequently, treatment with Notch1 or VCAM1-blocking antibodies prevented Notch-driven metastasis, and genetic ablation of EC Notch signaling inhibited peritoneal neutrophil infiltration in an ovarian carcinoma mouse model.
Collapse
Affiliation(s)
- Elfriede Wieland
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Juan Rodriguez-Vita
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sven S Liebler
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Vascular Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Carolin Mogler
- Institute of Pathology, Heidelberg University Hospital, Vascular Oncology and Metastasis (A190), German Cancer Research Center, 69120 Heidelberg, Germany
| | - Iris Moll
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefanie E Herberich
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Vascular Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Elisa Espinet
- Division of Stem Cells and Cancer (A010), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance and the German Cancer Consortium (DKTK), Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Esther Herpel
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Amitai Menuchin
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, 69978 Tel Aviv, Israel
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology (C020), German Cancer Research Center, 69120 Heidelberg, Germany; Research Group Genetic Cancer Epidemiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research (C070), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Christoffer Gebhardt
- Clinical Cooperation Unit Dermato-Oncology (G300), German Cancer Research Center (DKFZ), Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research (C070), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Division of Preventive Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer (A010), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance and the German Cancer Consortium (DKTK), Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University and Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Jochen Utikal
- Clinical Cooperation Unit Dermato-Oncology (G300), German Cancer Research Center (DKFZ), Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 69120 Heidelberg, Germany
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, 69978 Tel Aviv, Israel
| | - Andreas Fischer
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Vascular Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| |
Collapse
|
59
|
Adriani G, Ma D, Pavesi A, Kamm RD, Goh ELK. A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood-brain barrier. LAB ON A CHIP 2017; 17:448-459. [PMID: 28001148 DOI: 10.1039/c6lc00638h] [Citation(s) in RCA: 302] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The neurovascular unit is a complex, interdependent system composed of neurons and neural supporting cells, such as astrocytes, as well as cells that comprise the vascular system including endothelial cells, pericytes, and smooth muscle cells. Each cell type in the neurovascular unit plays an essential role, either in transmitting and processing neural signals or in maintaining the appropriate microenvironmental conditions for healthy neural function. In vitro neurovascular models can be useful for understanding the different roles and functions of the cells composing the neurovascular unit, as well as for assessing the effects on neural function of therapeutic compounds after crossing the endothelial barrier. Here, we report a novel three-dimensional neurovascular microfluidic model consisting of primary rat astrocytes and neurons together with human cerebral microvascular endothelial cells. These three cell types in our neurovascular chip (NVC) show distinct cell type-specific morphological characteristics and functional properties. In particular, morphological and functional analysis of neurons enables quantitative assessment of neuronal responses, while human cerebral endothelial cells form monolayers with size-selective permeability similar to existing in vitro blood-brain barrier (BBB) models.
Collapse
Affiliation(s)
- Giulia Adriani
- Singapore-MIT Alliance for Research and Technology, 138602 Singapore
| | - Dongliang Ma
- Department of Research, National Neuroscience Institute, 20 College Road, 169856 Singapore and Neuroscience Academic Clinical Programme, Duke-NUS Medical School, 169857 Singapore.
| | - Andrea Pavesi
- Singapore-MIT Alliance for Research and Technology, 138602 Singapore
| | - Roger D Kamm
- Singapore-MIT Alliance for Research and Technology, 138602 Singapore and Massachusetts Institute of Technology, Cambridge, MA, 02139 USA.
| | - Eyleen L K Goh
- Department of Research, National Neuroscience Institute, 20 College Road, 169856 Singapore and Neuroscience Academic Clinical Programme, Duke-NUS Medical School, 169857 Singapore. and Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore and KK Research Center, KK Women's and Children's Hospital, Singapore 229899, Singapore
| |
Collapse
|
60
|
Jarad M, Kuczynski EA, Morrison J, Viloria-Petit AM, Coomber BL. Release of endothelial cell associated VEGFR2 during TGF-β modulated angiogenesis in vitro. BMC Cell Biol 2017; 18:10. [PMID: 28114883 PMCID: PMC5260130 DOI: 10.1186/s12860-017-0127-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/18/2017] [Indexed: 01/11/2023] Open
Abstract
Background Sprouting angiogenesis requires vascular endothelial proliferation, migration and morphogenesis. The process is regulated by soluble factors, principally vascular endothelial growth factor (VEGF), and via bidirectional signaling through the Jagged/Notch system, leading to assignment of tip cell and stalk cell identity. The cytokine transforming growth factor beta (TGF-β) can either stimulate or inhibit angiogenesis via its differential surface receptor signaling. Here we evaluate changes in expression of angiogenic signaling receptors when bovine aortic endothelial cells were exposed to TGF-β1 under low serum conditions. Results TGF-β1 induced a dose dependent inhibition of tip cell assignment and subsequent angiogenesis on Matrigel, maximal at 5.0 ng/ml. This occurred via ALK5-dependent pathways and was accompanied by significant upregulation of the TGF-β co-receptor endoglin, and SMAD2 phosphorylation, but no alteration in Smad1/5 activation. TGF-β1 also induced ALK5-dependent downregulation of Notch1 but not of its ligand delta-like ligand 4. Cell associated VEGFR2 (but not VEGFR1) was significantly downregulated and accompanied by reciprocal upregulation of VEGFR2 in conditioned medium. Quantitative polymerase chain reaction analysis revealed that this soluble VEGFR2 was not generated by a selective shift in mRNA isoform transcription. This VEGFR2 in conditioned medium was full-length protein and was associated with increased soluble HSP-90, consistent with a possible shedding of microvesicles/exosomes. Conclusions Taken together, our results suggest that endothelial cells exposed to TGF-β1 lose both tip and stalk cell identity, possibly mediated by loss of VEGFR2 signaling. The role of these events in physiological and pathological angiogenesis requires further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s12860-017-0127-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Jarad
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - E A Kuczynski
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - J Morrison
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - A M Viloria-Petit
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - B L Coomber
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada.
| |
Collapse
|
61
|
Polychlorinated biphenyls target Notch/Dll and VEGF R2 in the mouse placenta and human trophoblast cell lines for their anti-angiogenic effects. Sci Rep 2017; 7:39885. [PMID: 28071720 PMCID: PMC5223111 DOI: 10.1038/srep39885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 11/29/2016] [Indexed: 01/02/2023] Open
Abstract
The intrauterine environment is particularly vulnerable to environmental exposures. We previously established a mouse model that provided evidence for pregnancy complications and placental anti-angiogenesis in response to Aroclor 1254 (A-1254), a mixture of polychlorinated biphenyls (PCBs). Importantly, these effects were observed in IL-10-/-, but not wild type, mice, suggesting that IL-10 deficiency predisposes to pregnancy disruptive effects of environmental toxicants. However, the mechanisms by which PCBs cause anti-angiogenic effects are unclear. Here, we evaluated PCB-mediated anti-angiogenic effects by diverse but complementary approaches, including HUVEC-mediated trophoblast invasion in nude mice, in vitro three-dimensional capillary tube formation involving HUVEC and/or HTR8 trophoblasts, and aortic ring endothelial cell outgrowth/sprouting. Taken together, our data suggest that PCBs act as potent anti-angiogenic agents. Importantly, we show that treatment of pregnant IL-10-/- mice with A-1254 resulted in placental activation of the Notch/Delta-like ligand (Dll) pathway, a master regulator of cell-cell interaction and vascular patterning. Similar results were obtained with HUVEC and HTR8 trophoblasts. Rescue of A-1254-induced disruption of HUVEC-based tube formation by γ-secretase inhibitor L1790 confirmed the critical role of the Notch/Dll pathway. Our data suggest that PCBs impart pregnancy disruptive functions by activating the Notch/Dll pathway and by inducing anti-angiogenic effects at the maternal-fetal interface.
Collapse
|
62
|
Luhtala N, Aslanian A, Yates JR, Hunter T. Secreted Glioblastoma Nanovesicles Contain Intracellular Signaling Proteins and Active Ras Incorporated in a Farnesylation-dependent Manner. J Biol Chem 2016; 292:611-628. [PMID: 27909058 DOI: 10.1074/jbc.m116.747618] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 11/11/2016] [Indexed: 12/21/2022] Open
Abstract
Glioblastomas (GBMs) are malignant brain tumors with a median survival of less than 18 months. Redundancy of signaling pathways represented within GBMs contributes to their therapeutic resistance. Exosomes are extracellular nanovesicles released from cells and present in human biofluids that represent a possible biomarker of tumor signaling state that could aid in personalized treatment. Herein, we demonstrate that mouse GBM cell-derived extracellular nanovesicles resembling exosomes from an H-RasV12 myr-Akt mouse model for GBM are enriched for intracellular signaling cascade proteins (GO: 0007242) and Ras protein signal transduction (GO: 0007265), and contain active Ras. Active Ras isolated from human and mouse GBM extracellular nanovesicles lysates using the Ras-binding domain of Raf also coprecipitates with ESCRT (endosomal sorting complex required for transport)-associated exosome proteins Vps4a and Alix. Although we initially hypothesized a role for active Ras protein signaling in exosome biogenesis, we found that GTP binding of K-Ras was dispensable for its packaging within extracellular nanovesicles and for the release of Alix. By contrast, farnesylation of K-Ras was required for its packaging within extracellular nanovesicles, yet expressing a K-Ras farnesylation mutant did not decrease the number of nanovesicles or the amount of Alix protein released per cell. Overall, these results emphasize the primary importance of membrane association in packaging of extracellular nanovesicle factors and indicate that screening nanovesicles within human fluids could provide insight into tissue origin and the wiring of signaling proteins at membranes to predict onset and behavior of cancer and other diseases linked to deregulated membrane signaling states.
Collapse
Affiliation(s)
- Natalie Luhtala
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037 and
| | - Aaron Aslanian
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037 and.,the Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
| | - John R Yates
- the Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
| | - Tony Hunter
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037 and
| |
Collapse
|
63
|
Abstract
Stem cells are critical to maintaining steady-state organ homeostasis and regenerating injured tissues. Recent intriguing reports implicate extracellular vesicles (EVs) as carriers for the distribution of morphogens and growth and differentiation factors from tissue parenchymal cells to stem cells, and conversely, stem cell-derived EVs carrying certain proteins and nucleic acids can support healing of injured tissues. We describe approaches to make use of engineered EVs as technology platforms in therapeutics and diagnostics in the context of stem cells. For some regenerative therapies, natural and engineered EVs from stem cells may be superior to single-molecule drugs, biologics, whole cells, and synthetic liposome or nanoparticle formulations because of the ease of bioengineering with multiple factors while retaining superior biocompatibility and biostability and posing fewer risks for abnormal differentiation or neoplastic transformation. Finally, we provide an overview of current challenges and future directions of EVs as potential therapeutic alternatives to cells for clinical applications.
Collapse
Affiliation(s)
- Milad Riazifar
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697; .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, California 92697.,Department of Biomedical Engineering, University of California, Irvine, California 92697.,Department of Biological Chemistry, University of California, Irvine, California 92697
| | - Egest J Pone
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697; .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, California 92697.,Department of Biomedical Engineering, University of California, Irvine, California 92697.,Department of Biological Chemistry, University of California, Irvine, California 92697
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine, The Sahlgrenska Academy, Göteborg University, SE-405 30 Göteborg, Sweden.,Codiak BioSciences Inc., Woburn, Massachusetts 01801
| | - Weian Zhao
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697; .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, California 92697.,Department of Biomedical Engineering, University of California, Irvine, California 92697.,Department of Biological Chemistry, University of California, Irvine, California 92697
| |
Collapse
|
64
|
Pasquale EB. Exosomes expand the sphere of influence of Eph receptors and ephrins. J Cell Biol 2016; 214:5-7. [PMID: 27354377 PMCID: PMC4932377 DOI: 10.1083/jcb.201606074] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 06/15/2016] [Indexed: 12/21/2022] Open
Abstract
Membrane-anchored Eph receptors and ephrins represent a ubiquitous intercellular communication system that typically engages at sites of cell–cell contact to initiate bidirectional signaling. Gong et al. (2016. J. Cell Biol.http://dx.doi.org/10.1083/jcb.201601085) show that cells can deploy the EphB2 receptor on exosomes to activate ephrinB signaling and collapse the growth cones of distant neurons.
Collapse
Affiliation(s)
- Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| |
Collapse
|
65
|
Wendler F, Stamp GW, Giamas G. Tumor-Stromal Cell Communication: Small Vesicles Signal Big Changes. Trends Cancer 2016; 2:326-329. [PMID: 28741534 DOI: 10.1016/j.trecan.2016.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/16/2016] [Accepted: 05/24/2016] [Indexed: 12/16/2022]
Abstract
Reciprocal interactions between malignant and stromal cells create a local microenvironment that fosters tumor growth. Extracellular vesicles (EVs) such as exosomes, microvesicles, and large oncosomes are involved in tumor-stroma communication by shuttling signaling cargo and other molecules. Here we discuss how EVs released by cancer or stromal cells impact the proliferation, differentiation, and metabolism of tumors.
Collapse
Affiliation(s)
- Franz Wendler
- University of Sussex, School of Life Sciences, John Maynard Smith Building, Falmer, Brighton BN1 9QG, UK.
| | - Gordon W Stamp
- Division of Diabetes, Endocrinology, and Metabolism, Imperial College Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Georgios Giamas
- University of Sussex, School of Life Sciences, John Maynard Smith Building, Falmer, Brighton BN1 9QG, UK.
| |
Collapse
|
66
|
Lunavat TR, Cheng L, Kim DK, Bhadury J, Jang SC, Lässer C, Sharples RA, López MD, Nilsson J, Gho YS, Hill AF, Lötvall J. Small RNA deep sequencing discriminates subsets of extracellular vesicles released by melanoma cells--Evidence of unique microRNA cargos. RNA Biol 2016; 12:810-23. [PMID: 26176991 PMCID: PMC4615768 DOI: 10.1080/15476286.2015.1056975] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Melanoma cells release different types of extracellular vesicles (EVs) into the extracellular milieu that are involved with communication and signaling in the tumor microenvironment. Subsets of EVs include exosomes, microvesicles, and apoptotic bodies that carry protein and genetic (RNA) cargos. To define the contribution of the RNA cargo of melanoma cell derived EVs we performed small RNA sequencing to identify different small RNAs in the EV subsets. Using validated centrifugation protocols, we separated these EV subsets released by the melanoma cell line MML-1, and performed RNA sequencing with the Ion Torrent platform. Various, but different, non-coding RNAs were detected in the EV subsets, including microRNA, mitochondrial associated tRNA, small nucleolar RNA, small nuclear RNA, Ro associated Y-RNA, vault RNA and Y-RNA. We identified in total 1041 miRNAs in cells and EV subsets. Hierarchical clustering showed enrichment of specific miRNAs in exosomes, including hsa-miR-214-3p, hsa-miR-199a-3p and hsa-miR-155-5p, all being associated with melanoma progression. Comparison of exosomal miRNAs with miRNAs in clinical melanoma samples indicate that multiple miRNAs in exosomes also are expressed specifically in melanoma tissues, but not in benign naevi. This study shows for the first time the presence of distinct small RNAs in subsets of EVs released by melanoma cells, with significant similarities to clinical melanoma tissue, and provides unique insights into the contribution of EV associated extracellular RNA in cancer.
Collapse
Affiliation(s)
- Taral R Lunavat
- a Krefting Research Center ; Institute of Medicine; University of Gothenburg ; Gothenburg , Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Abstract
Stroke is one of the leading causes of death and disability worldwide. Stroke recovery is orchestrated by a set of highly interactive processes that involve the neurovascular unit and neural stem cells. Emerging data suggest that exosomes play an important role in intercellular communication by transferring exosomal protein and RNA cargo between source and target cells in the brain. Here, we review these advances and their impact on promoting coupled brain remodeling processes after stroke. The use of exosomes for therapeutic applications in stroke is also highlighted.
Collapse
Affiliation(s)
- Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
68
|
Bovy N, Blomme B, Frères P, Dederen S, Nivelles O, Lion M, Carnet O, Martial JA, Noël A, Thiry M, Jérusalem G, Josse C, Bours V, Tabruyn SP, Struman I. Endothelial exosomes contribute to the antitumor response during breast cancer neoadjuvant chemotherapy via microRNA transfer. Oncotarget 2016; 6:10253-66. [PMID: 25860935 PMCID: PMC4496353 DOI: 10.18632/oncotarget.3520] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/17/2015] [Indexed: 12/21/2022] Open
Abstract
The interaction between tumor cells and their microenvironment is an essential aspect of tumor development. Therefore, understanding how this microenvironment communicates with tumor cells is crucial for the development of new anti-cancer therapies. MicroRNAs (miRNAs) are small non-coding RNAs that inhibit gene expression. They are secreted into the extracellular medium in vesicles called exosomes, which allow communication between cells via the transfer of their cargo. Consequently, we hypothesized that circulating endothelial miRNAs could be transferred to tumor cells and modify their phenotype. Using exogenous miRNA, we demonstrated that endothelial cells can transfer miRNA to tumor cells via exosomes. Using miRNA profiling, we identified miR-503, which exhibited downregulated levels in exosomes released from endothelial cells cultured under tumoral conditions. The modulation of miR-503 in breast cancer cells altered their proliferative and invasive capacities. We then identified two targets of miR-503, CCND2 and CCND3. Moreover, we measured increased plasmatic miR-503 in breast cancer patients after neoadjuvant chemotherapy, which could be partly due to increased miRNA secretion by endothelial cells. Taken together, our data are the first to reveal the involvement of the endothelium in the modulation of tumor development via the secretion of circulating miR-503 in response to chemotherapy treatment.
Collapse
Affiliation(s)
- Nicolas Bovy
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| | - Benoît Blomme
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| | - Pierre Frères
- Laboratory of Human Genetics, GIGA-R, University of Liège, Belgium
| | - Stella Dederen
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| | - Olivier Nivelles
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| | - Michelle Lion
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| | - Oriane Carnet
- Laboratory of Tumor & Development Biology, GIGA-R, University of Liège, Belgium
| | - Joseph A Martial
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| | - Agnès Noël
- Laboratory of Tumor & Development Biology, GIGA-R, University of Liège, Belgium
| | - Marc Thiry
- Laboratory of Cell and Tissues Biology, University of Liège, Belgium
| | | | - Claire Josse
- Laboratory of Human Genetics, GIGA-R, University of Liège, Belgium
| | - Vincent Bours
- Laboratory of Human Genetics, GIGA-R, University of Liège, Belgium
| | | | - Ingrid Struman
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, Belgium
| |
Collapse
|
69
|
Sharma A, Khatun Z, Shiras A. Tumor exosomes: cellular postmen of cancer diagnosis and personalized therapy. Nanomedicine (Lond) 2016; 11:421-37. [PMID: 26784674 DOI: 10.2217/nnm.15.210] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nanosized (30-150 nm) extracellular vesicles 'exosomes' are secreted by cells for intercellular communication during normal and pathological conditions. Exosomes carry biomacromolecules from cell-of-origin and, therefore, represent molecular bioprint of the cell. Tumor-derived exosomes or TDEx modulate tumor microenvironment by transfer of macromolecules locally as well as at distant metastatic sites. Due to their biological stability, TDEx are rich source of biomarkers in cancer patients. TDEx focused cancer diagnosis allows liquid biopsy-based tumor typing and may facilitate therapy response monitoring by developing novel exosomes diagnostics. Therefore, efficient and specific capturing of exosomes for subsequent amplification of the biomessages; for example, DNA, RNA, miRNA can reinvent cancer diagnosis. Here, in this review, we discuss advancements in exosomes isolation strategies, presence of exosomes biomarkers and importance of TDEx in gauging tumor heterogeneity for their potential use in cancer diagnosis, therapy.
Collapse
Affiliation(s)
- Aman Sharma
- ExoCan Healthcare Technologies Pvt Ltd, L4, 100 NCL Innovation Park, Dr Homi Bhabha Road, Pune-411008, India.,National Centre for Cell Science, SP Pune University Campus, Ganeshkhind, Pune411007
| | - Zamila Khatun
- ExoCan Healthcare Technologies Pvt Ltd, L4, 100 NCL Innovation Park, Dr Homi Bhabha Road, Pune-411008, India
| | - Anjali Shiras
- National Centre for Cell Science, SP Pune University Campus, Ganeshkhind, Pune411007
| |
Collapse
|
70
|
Bátiz LF, Castro MA, Burgos PV, Velásquez ZD, Muñoz RI, Lafourcade CA, Troncoso-Escudero P, Wyneken U. Exosomes as Novel Regulators of Adult Neurogenic Niches. Front Cell Neurosci 2016; 9:501. [PMID: 26834560 PMCID: PMC4717294 DOI: 10.3389/fncel.2015.00501] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/14/2015] [Indexed: 01/09/2023] Open
Abstract
Adult neurogenesis has been convincingly demonstrated in two regions of the mammalian brain: the sub-granular zone (SGZ) of the dentate gyrus (DG) in the hippocampus, and the sub-ventricular zone (SVZ) of the lateral ventricles (LV). SGZ newborn neurons are destined to the granular cell layer (GCL) of the DG, while new neurons from the SVZ neurons migrate rostrally into the olfactory bulb (OB). The process of adult neurogenesis persists throughout life and is supported by a pool of neural stem cells (NSCs), which reside in a unique and specialized microenvironment known as "neurogenic niche". Neurogenic niches are structured by a complex organization of different cell types, including the NSC-neuron lineage, glial cells and vascular cells. Thus, cell-to-cell communication plays a key role in the dynamic modulation of homeostasis and plasticity of the adult neurogenic process. Specific cell-cell contacts and extracellular signals originated locally provide the necessary support and regulate the balance between self-renewal and differentiation of NSCs. Furthermore, extracellular signals originated at distant locations, including other brain regions or systemic organs, may reach the niche through the cerebrospinal fluid (CSF) or the vasculature and influence its nature. The role of several secreted molecules, such as cytokines, growth factors, neurotransmitters, and hormones, in the biology of adult NSCs, has been systematically addressed. Interestingly, in addition to these well-recognized signals, a novel type of intercellular messengers has been identified recently: the extracellular vesicles (EVs). EVs, and particularly exosomes, are implicated in the transfer of mRNAs, microRNAs (miRNAs), proteins and lipids between cells and thus are able to modify the function of recipient cells. Exosomes appear to play a significant role in different stem cell niches such as the mesenchymal stem cell niche, cancer stem cell niche and pre-metastatic niche; however, their roles in adult neurogenic niches remain virtually unexplored. This review focuses on the current knowledge regarding the functional relationship between cellular and extracellular components of the adult SVZ and SGZ neurogenic niches, and the growing evidence that supports the potential role of exosomes in the physiology and pathology of adult neurogenesis.
Collapse
Affiliation(s)
- Luis Federico Bátiz
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de ChileValdivia, Chile; Program for Cell Biology and Microscopy, Universidad Austral de ChileValdivia, Chile; Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de ChileValdivia, Chile
| | - Maite A Castro
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de ChileValdivia, Chile; Program for Cell Biology and Microscopy, Universidad Austral de ChileValdivia, Chile; Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de ChileValdivia, Chile
| | - Patricia V Burgos
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de ChileValdivia, Chile; Program for Cell Biology and Microscopy, Universidad Austral de ChileValdivia, Chile; Instituto de Fisiología, Facultad de Medicina, Universidad Austral de ChileValdivia, Chile
| | - Zahady D Velásquez
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de ChileValdivia, Chile; Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de ChileValdivia, Chile
| | - Rosa I Muñoz
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de ChileValdivia, Chile; Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de ChileValdivia, Chile
| | - Carlos A Lafourcade
- Laboratorio de Neurociencias, Facultad de Medicina, Universidad de Los Andes Santiago, Chile
| | - Paulina Troncoso-Escudero
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de ChileValdivia, Chile; Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de ChileValdivia, Chile
| | - Ursula Wyneken
- Laboratorio de Neurociencias, Facultad de Medicina, Universidad de Los Andes Santiago, Chile
| |
Collapse
|
71
|
Garcia NA, Moncayo-Arlandi J, Sepulveda P, Diez-Juan A. Cardiomyocyte exosomes regulate glycolytic flux in endothelium by direct transfer of GLUT transporters and glycolytic enzymes. Cardiovasc Res 2015; 109:397-408. [PMID: 26609058 DOI: 10.1093/cvr/cvv260] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 11/10/2015] [Indexed: 12/25/2022] Open
Abstract
AIMS Cardiomyocytes (CMs) and endothelial cells (ECs) have an intimate anatomical relationship, which is essential for maintaining the metabolic requirements of the heart. Little is known about the mechanisms that regulate nutrient flow from ECs to associated CMs, especially in situations of acute stress when local active processes are required to regulate endothelial transport. We examined whether CM-derived exosomes can modulate glucose transport and metabolism in ECs. METHODS AND RESULTS In conditions of glucose deprivation, CMs increase the synthesis and secretion of exosomes. These exosomes are loaded with functional glucose transporters and glycolytic enzymes, which are internalized by ECs, leading to increased glucose uptake, glycolytic activity, and pyruvate production in recipient cells. CONCLUSION These findings establish CM-derived exosomes as key components of the cardio-endothelial communication system which, through intercellular protein complementation, would allow a rapid response from ECs to increase glucose transport and a putative uptake of metabolic fuels from blood to CMs. This CM-EC protein complementation process might have implications for metabolic regulation in health and disease.
Collapse
Affiliation(s)
- Nahuel A Garcia
- Mixt Unit for Cardiovascular Repair, Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Javier Moncayo-Arlandi
- Cardiovascular Genetics Center, Institut d́Investigació Biomèdica de Girona, Girona, Spain
| | - Pilar Sepulveda
- Mixt Unit for Cardiovascular Repair, Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Adva. Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Antonio Diez-Juan
- Fundación IVI/INCLIVA, Valencia, Spain IGENOMICS, Calle Catedrático Agustín Escardino 9, Paterna, Valencia 46980, Spain
| |
Collapse
|
72
|
Hen G, Nicenboim J, Mayseless O, Asaf L, Shin M, Busolin G, Hofi R, Almog G, Tiso N, Lawson ND, Yaniv K. Venous-derived angioblasts generate organ-specific vessels during zebrafish embryonic development. Development 2015; 142:4266-78. [PMID: 26525671 PMCID: PMC4689221 DOI: 10.1242/dev.129247] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/25/2015] [Indexed: 01/04/2023]
Abstract
Formation and remodeling of vascular beds are complex processes orchestrated by multiple signaling pathways. Although it is well accepted that vessels of a particular organ display specific features that enable them to fulfill distinct functions, the embryonic origins of tissue-specific vessels and the molecular mechanisms regulating their formation are poorly understood. The subintestinal plexus of the zebrafish embryo comprises vessels that vascularize the gut, liver and pancreas and, as such, represents an ideal model in which to investigate the early steps of organ-specific vessel formation. Here, we show that both arterial and venous components of the subintestinal plexus originate from a pool of specialized angioblasts residing in the floor of the posterior cardinal vein (PCV). Using live imaging of zebrafish embryos, in combination with photoconvertable transgenic reporters, we demonstrate that these angioblasts undergo two phases of migration and differentiation. Initially, a subintestinal vein forms and expands ventrally through a Bone Morphogenetic Protein-dependent step of collective migration. Concomitantly, a Vascular Endothelial Growth Factor-dependent shift in the directionality of migration, coupled to the upregulation of arterial markers, is observed, which culminates with the generation of the supraintestinal artery. Together, our results establish the zebrafish subintestinal plexus as an advantageous model for the study of organ-specific vessel development and provide new insights into the molecular mechanisms controlling its formation. More broadly, our findings suggest that PCV-specialized angioblasts contribute not only to the formation of the early trunk vasculature, but also to the establishment of late-forming, tissue-specific vascular beds. Highlighted article: A specialized pool of angioblasts is the origin of the zebrafish subintestinal plexus, a structure that gives rise to the organ-specific vessels of the gut, liver and pancreas.
Collapse
Affiliation(s)
- Gideon Hen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Julian Nicenboim
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Oded Mayseless
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Lihee Asaf
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Masahiro Shin
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Giorgia Busolin
- Department of Biology, University of Padova, Padova I-35131, Italy
| | - Roy Hofi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gabriella Almog
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova I-35131, Italy
| | - Nathan D Lawson
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
73
|
Zeng C, Xing R, Liu J, Xing F. Role of CSL-dependent and independent Notch signaling pathways in cell apoptosis. Apoptosis 2015; 21:1-12. [DOI: 10.1007/s10495-015-1188-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
74
|
Meehan K, Vella LJ. The contribution of tumour-derived exosomes to the hallmarks of cancer. Crit Rev Clin Lab Sci 2015; 53:121-31. [PMID: 26479834 DOI: 10.3109/10408363.2015.1092496] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exosomes are small, biologically active extracellular vesicles and over the last decade, both stromal and tumour-derived exosomes (TDE) have been implicated in cancer onset, progression and metastases. Cancer is a complex disease that is underpinned by several "cancer hallmarks", originally described by Hanahan and Weinberg in 2000 and then revised in 2011. The hallmarks of cancer comprise six biological capabilities, along with two emerging hallmarks and two enabling characteristics that facilitate tumour growth and metastatic dissemination. Ample evidence supports a clear role for TDE in four of the original biological hallmarks (sustaining proliferative signalling, resisting cell death, inducing angiogenesis and activating invasion and metastases). A less-defined role exists for TDE in evading growth suppressors, and currently, there is no evidence to suggest a role for TDE in enabling replicative immortality. TDE are intimately involved in the newly defined hallmarks of cancer and enabling characteristics, most evidently in immune inhibition and tumour-promoting inflammation, which ultimately enable escape from immune destruction and tumour progression. Herein, we discuss the role of TDE in the context of the hallmarks and enabling characteristics of cancer as defined by Hanahan and Weinberg.
Collapse
Affiliation(s)
- Katie Meehan
- a School of Pathology and Laboratory Medicine, University of Western Australia , Crawley , Australia and
| | - Laura J Vella
- b Olivia Newton-John Cancer Research Institute, Level 5 Olivia Newton-John Cancer and Wellness Centre , Heidelberg , Australia
| |
Collapse
|
75
|
Hu J, Qiu J, Zheng Y, Zhang T, Yin T, Xie X, Wang G. AAMP Regulates Endothelial Cell Migration and Angiogenesis Through RhoA/Rho Kinase Signaling. Ann Biomed Eng 2015; 44:1462-74. [PMID: 26350504 DOI: 10.1007/s10439-015-1442-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/01/2015] [Indexed: 01/23/2023]
Abstract
Angiogenesis is a complicated process including endothelial cell proliferation, migration and tube formation. AAMP plays a role in regulating cell migration of multiple cell types. The purpose of this study was to investigate whether AAMP regulates angiogenesis, and to clarify the role of AAMP in the VEGF-induced angiogenesis. We found that AAMP expressed in multiple cell types and mainly localized in cytoplasm and membrane in vascular endothelial cells. Using tube formation assay in vitro and aortic ring assay, siRNA-mediated knockdown and antibody blockade of AAMP impaired VEGF-induced endothelial cell tube formation and aortic ring angiogenic sprouting. Mechanistic studies showed that AAMP expression was significantly upregulated by VEGF in a concentration and time-dependent manner. Moreover, VEGF recruited AAMP to the cell membrane protrusions. AAMP regulates angiogenesis by mediating the spreading and migration of vascular endothelial cells. AAMP knock-down reduced VEGF-induced actin stress fibers and collagen gel contraction. Furthermore, we identified RhoA/Rho kinase signaling as an important factor that contributes to the action of AAMP in regulating endothelial cell migration and angiogenesis. Altogether, these data demonstrated the critical role of AAMP in angiogenesis and suggested blocking AAMP could serve as a potential therapeutic strategy for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Jianjun Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yiming Zheng
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tao Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Xiang Xie
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory For Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
76
|
Emerging roles of exosomes during epithelial-mesenchymal transition and cancer progression. Semin Cell Dev Biol 2015; 40:60-71. [PMID: 25721809 DOI: 10.1016/j.semcdb.2015.02.008] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/02/2015] [Accepted: 02/18/2015] [Indexed: 02/08/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a highly conserved process defined by the loss of epithelial characteristics, and acquisition of the mesenchymal phenotype. In addition to its central role in development, EMT has been implicated as a cellular process during tumourigenesis which facilitates tumour cell invasion and metastasis. The EMT process has been largely defined by signal transduction networks and transcriptional factors that activate mesenchymal-associated gene expression. Knowledge of secretome components that influence EMT including secreted proteins/peptides and membrane-derived extracellular vesicles (EVs) (i.e., exosomes) has emerged. Here we review EV cargo associated with inducing the hallmarks of EMT and cancer progression, modulators of cell transformation, invasion/migration, angiogenesis, and components involved in establishing the metastatic niche.
Collapse
|
77
|
Saleem SN, Abdel-Mageed AB. Tumor-derived exosomes in oncogenic reprogramming and cancer progression. Cell Mol Life Sci 2015; 72:1-10. [PMID: 25156068 PMCID: PMC4282952 DOI: 10.1007/s00018-014-1710-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 12/21/2022]
Abstract
In multicellular organisms, effective communication between cells is a crucial part of cellular and tissue homeostasis. This communication mainly involves direct cell-cell contact as well as the secretion of molecules that bind to receptors at the recipient cells. However, a more recently characterized mode of intercellular communication-the release of membrane vesicles known as exosomes-has been the subject of increasing interest and intensive research over the past decade. Following the discovery of the exosome-mediated immune activation, the pathophysiological roles of exosomes have been recognized in different diseases, including cancer. In this review, we describe the biogenesis and main physical characteristics that define exosomes as a specific population of secreted vesicles, with a special focus on their role in oncogenic transformation and cancer progression.
Collapse
Affiliation(s)
- Sarmad N Saleem
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
78
|
Rostama B, Peterson SM, Vary CPH, Liaw L. Notch signal integration in the vasculature during remodeling. Vascul Pharmacol 2014; 63:97-104. [PMID: 25464152 PMCID: PMC4304902 DOI: 10.1016/j.vph.2014.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/06/2014] [Accepted: 10/10/2014] [Indexed: 02/06/2023]
Abstract
Notch signaling plays many important roles in homeostasis and remodeling in the vessel wall, and serves a critical role in the communication between endothelial cells and smooth muscle cells. Within blood vessels, Notch signaling integrates with multiple pathways by mechanisms including direct protein–protein interaction, cooperative or synergistic regulation of signal cascades, and co-regulation of transcriptional targets. After establishment of the mature blood vessel, the spectrum and intensity of Notch signaling change during phases of active remodeling or disease progression. These changes can be mediated by regulation via microRNAs and protein stability or signaling, and corresponding changes in complementary signaling pathways. Notch also affects endothelial cells on a system level by regulating key metabolic components. This review will outline the most recent findings of Notch activity in blood vessels, with a focus on how Notch signals integrate with other molecular signaling pathways controlling vascular phenotype.
Collapse
Affiliation(s)
- Bahman Rostama
- Center for Molecular Medicine, Maine Medical Center Research Institute, USA
| | | | | | | |
Collapse
|
79
|
Abstract
Branching morphogenesis is the developmental program that builds the ramified epithelial trees of various organs, including the airways of the lung, the collecting ducts of the kidney, and the ducts of the mammary and salivary glands. Even though the final geometries of epithelial trees are distinct, the molecular signaling pathways that control branching morphogenesis appear to be conserved across organs and species. However, despite this molecular homology, recent advances in cell lineage analysis and real-time imaging have uncovered surprising differences in the mechanisms that build these diverse tissues. Here, we review these studies and discuss the cellular and physical mechanisms that can contribute to branching morphogenesis.
Collapse
Affiliation(s)
- Victor D Varner
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|