201
|
Tunneling nanotubes mediate the transfer of stem cell marker CD133 between hematopoietic progenitor cells. Exp Hematol 2016; 44:1092-1112.e2. [PMID: 27473566 DOI: 10.1016/j.exphem.2016.07.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 12/24/2022]
Abstract
Deciphering all mechanisms of intercellular communication used by hematopoietic progenitors is important, not only for basic stem cell research, but also in view of their therapeutic relevance. Here, we investigated whether these cells can produce the thin F-actin-based plasma membrane protrusions referred to as tunneling nanotubes (TNTs), which are known to bridge cells over long distances without contact with the substratum and transfer cargo molecules along them in various biological processes. We found that human primary CD34+ hematopoietic progenitors and leukemic KG1a cells develop such structures upon culture on primary mesenchymal stromal cells or specific extracellular-matrix-based substrata. Time-lapse video microscopy revealed that cell dislodgement is the primary mechanism responsible for TNT biogenesis. Surprisingly, we found that, among various cluster of differentiation (CD) markers, only the stem cell antigen CD133 is transferred between cells. It is selectively and directionally transported along the surface of TNTs in small clusters, such as cytoplasmic phospho-myosin light chain 2, suggesting that the latter actin motor protein might be implicated in this process. Our data provide new insights into the biology of hematopoietic progenitors that can contribute to our understanding of all facets of intercellular communication in the bone marrow microenvironment under healthy or cancerous conditions.
Collapse
|
202
|
Communication of Ca(2+) signals via tunneling membrane nanotubes is mediated by transmission of inositol trisphosphate through gap junctions. Cell Calcium 2016; 60:266-72. [PMID: 27388952 DOI: 10.1016/j.ceca.2016.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/17/2016] [Accepted: 06/18/2016] [Indexed: 11/22/2022]
Abstract
Tunneling membrane nanotubes (TNTs) are thin membrane projections linking cell bodies separated by many micrometers, which are proposed to mediate signaling and even transfer of cytosolic contents between distant cells. Several reports describe propagation of Ca(2+) signals between distant cells via TNTs, but the underlying mechanisms remain poorly understood. Utilizing a HeLa M-Sec cell line engineered to upregulate TNTs we replicated previous findings that mechanical stimulation elicits robust cytosolic Ca(2+) elevations that propagate to surrounding, physically separate cells. However, whereas this was previously interpreted to involve intercellular communication through TNTs, we found that Ca(2+) signal propagation was abolished - even in TNT-connected cells - after blocking ATP-mediated paracrine signaling with a cocktail of extracellular inhibitors. To then establish whether gap junctions may enable cell-cell signaling via TNTs under these conditions, we expressed sfGFP-tagged connexin-43 (Cx43) in HeLa M-Sec cells. We observed robust communication of mechanically-evoked Ca(2+) signals between distant but TNT-connected cells, but only when both cells expressed Cx43. Moreover, we also observed communication of Ca(2+) signals evoked in one cell by local photorelease of inositol 1,4,5-trisphosphate (IP3). Ca(2+) responses in connected cells began after long latencies at intracellular sites several microns from the TNT connection site, implicating intercellular transfer of IP3 and subsequent IP3-mediated Ca(2+) liberation, and not Ca(2+) itself, as the mediator between TNT-connected, Cx43-expressing cells. Our results emphasize the need to control for paracrine transmission in studies of cell-cell signaling via TNTs and indicate that, in this cell line, TNTs do not establish cytosolic continuity between connected cells but rather point to the crucial importance of connexins to enable communication of cytosolic Ca(2+) signals via TNTs.
Collapse
|
203
|
Affiliation(s)
- Leonardo Elia
- Humanitas Research Hospital, Via Manzoni 113, Rozzano, MI 20089, Italy Milan Unit, Institute of Genetic and Biomedical Research, Via Manzoni 113, Rozzano, MI 20089, Italy Humanitas University, Via Manzoni 113, Rozzano, MI 20089, Italy
| | - Gianluigi Condorelli
- Humanitas Research Hospital, Via Manzoni 113, Rozzano, MI 20089, Italy Milan Unit, Institute of Genetic and Biomedical Research, Via Manzoni 113, Rozzano, MI 20089, Italy Humanitas University, Via Manzoni 113, Rozzano, MI 20089, Italy
| |
Collapse
|
204
|
Scholkmann F. Long range physical cell-to-cell signalling via mitochondria inside membrane nanotubes: a hypothesis. Theor Biol Med Model 2016; 13:16. [PMID: 27267202 PMCID: PMC4896004 DOI: 10.1186/s12976-016-0042-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/27/2016] [Indexed: 02/07/2023] Open
Abstract
Coordinated interaction of single cells by cell-to-cell communication (signalling) enables complex behaviour necessary for the functioning of multicellular organisms. A quite newly discovered cell-to-cell signalling mechanism relies on nanotubular cell-co-cell connections, termed "membrane nanotubes" (MNTs). The present paper presents the hypothesis that mitochondria inside MNTs can form a connected structure (mitochondrial network) which enables the exchange of energy and signals between cells. It is proposed that two modes of energy and signal transmission may occur: electrical/electrochemical and electromagnetic (optical). Experimental work supporting the hypothesis is reviewed, and suggestions for future research regarding the discussed topic are given.
Collapse
Affiliation(s)
- Felix Scholkmann
- Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, Frauenklinikstr. 10, 8091, Zurich, Switzerland.
- Research Office for Complex Physical and Biological Systems (ROCoS), Mutschellenstr. 179, 8038, Zurich, Switzerland.
| |
Collapse
|
205
|
Drosophila cells use nanotube-like structures to transfer dsRNA and RNAi machinery between cells. Sci Rep 2016; 6:27085. [PMID: 27255932 PMCID: PMC4891776 DOI: 10.1038/srep27085] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 05/12/2016] [Indexed: 01/22/2023] Open
Abstract
Tunnelling nanotubes and cytonemes function as highways for the transport of organelles, cytosolic and membrane-bound molecules, and pathogens between cells. During viral infection in the model organism Drosophila melanogaster, a systemic RNAi antiviral response is established presumably through the transport of a silencing signal from one cell to another via an unknown mechanism. Because of their role in cell-cell communication, we investigated whether nanotube-like structures could be a mediator of the silencing signal. Here, we describe for the first time in the context of a viral infection the presence of nanotube-like structures in different Drosophila cell types. These tubules, made of actin and tubulin, were associated with components of the RNAi machinery, including Argonaute 2, double-stranded RNA, and CG4572. Moreover, they were more abundant during viral, but not bacterial, infection. Super resolution structured illumination microscopy showed that Argonaute 2 and tubulin reside inside the tubules. We propose that nanotube-like structures are one of the mechanisms by which Argonaute 2, as part of the antiviral RNAi machinery, is transported between infected and non-infected cells to trigger systemic antiviral immunity in Drosophila.
Collapse
|
206
|
Rehberg M, Nekolla K, Sellner S, Praetner M, Mildner K, Zeuschner D, Krombach F. Intercellular Transport of Nanomaterials is Mediated by Membrane Nanotubes In Vivo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1882-1890. [PMID: 26854197 DOI: 10.1002/smll.201503606] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/22/2015] [Indexed: 06/05/2023]
Abstract
So-called membrane nanotubes are cellular protrusions between cells whose functions include cell communication, environmental sampling, and protein transfer. It has been previously reported that systemically administered carboxyl-modified quantum dots (cQDs) are rapidly taken up by perivascular macrophages in skeletal muscle of healthy mice. Expanding these studies, it is found, by means of in vivo fluorescence microscopy on the mouse cremaster muscle, rapid uptake of cQDs not only by perivascular macrophages but also by tissue-resident cells, which are localized more than 100 μm distant from the closest vessel. Confocal microscopy on muscle tissue, immunostained for the membrane dye DiI, reveals the presence of continuous membranous structures between MHC-II-positive, F4/80-positive cells. These structures contain microtubules, components of the cytoskeleton, which clearly colocalize with cQDs. The cQDs are exclusively found inside endosomal vesicles. Most importantly, by using in vivo fluorescence microscopy, this study detected fast (0.8 μm s(-1) , mean velocity), bidirectional movement of cQDs in such structures, indicating transport of cQD-containing vesicles along microtubule tracks by the action of molecular motors. The findings are the first to demonstrate membrane nanotube function in vivo and they suggest a previously unknown route for the distribution of nanomaterials in tissue.
Collapse
Affiliation(s)
- Markus Rehberg
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katharina Nekolla
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sabine Sellner
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marc Praetner
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Fritz Krombach
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
207
|
Cui W, Lee JY. Arabidopsis callose synthases CalS1/8 regulate plasmodesmal permeability during stress. NATURE PLANTS 2016; 2:16034. [PMID: 27243643 DOI: 10.1038/nplants.2016.34] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 02/19/2016] [Indexed: 05/20/2023]
Abstract
Plants need to cope with biotic and abiotic stress through well-coordinated cell-to-cell communication to survive as sedentary organisms. Environmental challenges such as wounding, low temperature, oxidative states and pathogen infection are known to affect the symplasmic molecular exchange between plant cells determined by plasmodesmal permeability. However, the signalling pathways and mechanisms by which different environmental stressors affect plasmodesmal permeability are not well understood. Here we show that regulating callose accumulation at plasmodesmal channels is a common strategy to alter plasmodesmal permeability under both pathogen infection and mechanical wounding stress. We have identified Arabidopsis callose synthase 1 (CalS1) and CalS8 as key genes involved in this process, and have integrated these new players into both known and novel signalling pathways that control responses to biotic and abiotic stress. Our studies provide experimental data that indicate the presence of specialized pathways tuned to respond to particular stressors, and new insights into how plants regulate plasmodesmata in response to environmental assaults.
Collapse
Affiliation(s)
- Weier Cui
- Department of Plant and Soil Sciences, Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19711, USA
| | - Jung-Youn Lee
- Department of Plant and Soil Sciences, Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19711, USA
| |
Collapse
|
208
|
Lopez-Leal R, Court FA. Schwann Cell Exosomes Mediate Neuron-Glia Communication and Enhance Axonal Regeneration. Cell Mol Neurobiol 2016; 36:429-36. [PMID: 26993502 PMCID: PMC11482438 DOI: 10.1007/s10571-015-0314-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/24/2015] [Indexed: 12/30/2022]
Abstract
The functional and structural integrity of the nervous system depends on the coordinated action of neurons and glial cells. Phenomena like synaptic activity, conduction of action potentials, and neuronal growth and regeneration, to name a few, are fine tuned by glial cells. Furthermore, the active role of glial cells in the regulation of neuronal functions is underscored by several conditions in which specific mutation affecting the glia results in axonal dysfunction. We have shown that Schwann cells (SCs), the peripheral nervous system glia, supply axons with ribosomes, and since proteins underlie cellular programs or functions, this dependence of axons from glial cells provides a new and unexplored dimension to our understanding of the nervous system. Recent evidence has now established a new modality of intercellular communication through extracellular vesicles. We have already shown that SC-derived extracellular vesicles known as exosomes enhance axonal regeneration, and increase neuronal survival after pro-degenerative stimuli. Therefore, the biology nervous system will have to be reformulated to include that the phenotype of a nerve cell results from the contribution of two nuclei, with enormous significance for the understanding of the nervous system in health and disease.
Collapse
Affiliation(s)
- Rodrigo Lopez-Leal
- FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Millennium Nucleus in Regenerative Biology (MINREB), Department of Physiology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Av. B. O'Higgins 340, 8331150, Santiago, Chile
| | - Felipe A Court
- FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
- Millennium Nucleus in Regenerative Biology (MINREB), Department of Physiology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Av. B. O'Higgins 340, 8331150, Santiago, Chile.
| |
Collapse
|
209
|
Gungor-Ordueri NE, Celik-Ozenci C, Cheng CY. Ezrin: a regulator of actin microfilaments in cell junctions of the rat testis. Asian J Androl 2016; 17:653-8. [PMID: 25652626 PMCID: PMC4492059 DOI: 10.4103/1008-682x.146103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ezrin, radixin, moesin and merlin (ERM) proteins are highly homologous actin-binding proteins that share extensive sequence similarity with each other. These proteins tether integral membrane proteins and their cytoplasmic peripheral proteins (e.g., adaptors, nonreceptor protein kinases and phosphatases) to the microfilaments of actin-based cytoskeleton. Thus, these proteins are crucial to confer integrity of the apical membrane domain and its associated junctional complex, namely the tight junction and the adherens junction. Since ectoplasmic specialization (ES) is an F-actin-rich testis-specific anchoring junction-a highly dynamic ultrastructure in the seminiferous epithelium due to continuous transport of germ cells, in particular spermatids, across the epithelium during the epithelial cycle-it is conceivable that ERM proteins are playing an active role in these events. Although these proteins were first reported almost 25 years and have since been extensively studied in multiple epithelia/endothelia, few reports are found in the literature to examine their role in the actin filament bundles at the ES. Studies have shown that ezrin is also a constituent protein of the actin-based tunneling nanotubes (TNT) also known as intercellular bridges, which are transient cytoplasmic tubular ultrastructures that transport signals, molecules and even organelles between adjacent and distant cells in an epithelium to coordinate cell events that occur across an epithelium. Herein, we critically evaluate recent data on ERM in light of recent findings in the field in particular ezrin regarding its role in actin dynamics at the ES in the testis, illustrating additional studies are warranted to examine its physiological significance in spermatogenesis.
Collapse
Affiliation(s)
| | | | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, USA
| |
Collapse
|
210
|
Zaccard CR, Rinaldo CR, Mailliard RB. Linked in: immunologic membrane nanotube networks. J Leukoc Biol 2016; 100:81-94. [PMID: 26931578 DOI: 10.1189/jlb.4vmr0915-395r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/01/2016] [Indexed: 01/01/2023] Open
Abstract
Membrane nanotubes, also termed tunneling nanotubes, are F-actin-based structures that can form direct cytoplasmic connections and support rapid communication between distant cells. These nanoscale conduits have been observed in diverse cell types, including immune, neuronal, stromal, cancer, and stem cells. Until recently, little was known about the mechanisms involved in membrane nanotube development in myeloid origin APCs or how membrane nanotube networks support their ability to bridge innate and adaptive immunity. New research has provided insight into the modes of induction and regulation of the immune process of "reticulation" or the development of multicellular membrane nanotube networks in dendritic cells. Preprogramming by acute type 1 inflammatory mediators at their immature stage licenses mature type 1-polarized dendritic cells to reticulate upon subsequent interaction with CD40 ligand-expressing CD4(+) Th cells. Dendritic cell reticulation can support direct antigen transfer for amplification of specific T cell responses and can be positively or negatively regulated by signals from distinct Th cell subsets. Membrane nanotubes not only enhance the ability of immature dendritic cells to sense pathogens and rapidly mobilize nearby antigen-presenting cells in the peripheral tissues but also likely support communication of pathogen-related information from mature migratory dendritic cells to resident dendritic cells in lymph nodes. Therefore, the reticulation process facilitates a coordinated multicellular response for the efficient initiation of cell-mediated adaptive immune responses. Herein, we discuss studies focused on the molecular mechanisms of membrane nanotube formation, structure, and function in the context of immunity and how pathogens, such as HIV-1, may use dendritic cell reticulation to circumvent host defenses.
Collapse
Affiliation(s)
- C R Zaccard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pennsylvania, USA and
| | - C R Rinaldo
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pennsylvania, USA and Department of Pathology, University of Pittsburgh, Pennsylvania, USA
| | - R B Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pennsylvania, USA and
| |
Collapse
|
211
|
Victoria GS, Arkhipenko A, Zhu S, Syan S, Zurzolo C. Astrocyte-to-neuron intercellular prion transfer is mediated by cell-cell contact. Sci Rep 2016; 6:20762. [PMID: 26857744 PMCID: PMC4746738 DOI: 10.1038/srep20762] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 01/07/2016] [Indexed: 02/04/2023] Open
Abstract
Prion diseases are caused by misfolding of the cellular protein PrP(C) to an infectious conformer, PrP(Sc). Intercellular PrP(Sc) transfer propagates conversion and allows infectivity to move from the periphery to the brain. However, how prions spread between cells of the central nervous system is unclear. Astrocytes are specialized non-neuronal cells within the brain that have a number of functions indispensable for brain homeostasis. Interestingly, they are one of the earliest sites of prion accumulation in the brain. A fundamental question arising from this observation is whether these cells are involved in intercellular prion transfer and thereby disease propagation. Using co-culture systems between primary infected astrocytes and granule neurons or neuronal cell lines, we provide direct evidence that prion-infected astrocytes can disseminate prion to neurons. Though astrocytes are capable of secreting PrP, this is an inefficient method of transferring prion infectivity. Efficient transfer required co-culturing and direct cell contact. Astrocytes form numerous intercellular connections including tunneling nanotubes, containing PrP(Sc), often colocalized with endolysosomal vesicles, which may constitute the major mechanism of transfer. Because of their role in intercellular transfer of prions astrocytes may influence progression of the disease.
Collapse
Affiliation(s)
- Guiliana Soraya Victoria
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Alexander Arkhipenko
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Seng Zhu
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Sylvie Syan
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Chiara Zurzolo
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| |
Collapse
|
212
|
Yang H, Borg TK, Ma Z, Xu M, Wetzel G, Saraf LV, Markwald R, Runyan RB, Gao BZ. Biochip-based study of unidirectional mitochondrial transfer from stem cells to myocytes via tunneling nanotubes. Biofabrication 2016; 8:015012. [PMID: 26844857 DOI: 10.1088/1758-5090/8/1/015012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tunneling nanotubes (TNTs) are small membranous tubes of 50-1000 nm diameter observed to connect cells in culture. Transfer of subcellular organelles through TNTs was observed in vitro and in vivo, but the formation and significance of these structures is not well understood. A polydimethylsiloxane biochip-based coculture model was devised to constrain TNT orientation and explore both TNT-formation and TNT-mediated mitochondrial transfer. Two parallel microfluidic channels connected by an array of smaller microchannels enabled localization of stem cell and cardiomyocyte populations while allowing connections to form between them. Stem cells and cardiomyocytes were deposited in their respective microfluidic channels, and stem cell-cardiomyocyte pairs were formed via the microchannels. Formation of TNTs and transfer of stained mitochondria through TNTs was observed by 24 h real-time video recording. The data show that stem cells are 7.7 times more likely to initiate contact by initial extension of filopodia. By 24 h, 67% of nanotube connections through the microchannels are composed of cardiomyocyte membrane. Filopodial extension and retraction by stem cells draws an extension of TNTs from cardiomyocytes. MitoTracker staining shows that unidirectional transfer of mitochondria between stem cell-cardiomyocyte pairs invariably originates from stem cells. Control experiments with cardiac fibroblasts and cardiomyocytes show little nanotube formation between homotypic or mixed cell pairs and no mitochondrial transfer. These data identify a novel biological process, unidirectional mitochondrial transfer, mediated by heterotypic TNT connections. This suggests that the enhancement of cardiomyocyte function seen after stem-cell injection may be due to a bioenergetic stimulus provided by mitochondrial transfer.
Collapse
Affiliation(s)
- Huaxiao Yang
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Dubey G, Malli Mohan G, Dubrovsky A, Amen T, Tsipshtein S, Rouvinski A, Rosenberg A, Kaganovich D, Sherman E, Medalia O, Ben-Yehuda S. Architecture and Characteristics of Bacterial Nanotubes. Dev Cell 2016; 36:453-61. [DOI: 10.1016/j.devcel.2016.01.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 09/28/2015] [Accepted: 01/22/2016] [Indexed: 01/11/2023]
|
214
|
Rimkutė L, Jotautis V, Marandykina A, Sveikatienė R, Antanavičiūtė I, Skeberdis VA. The role of neural connexins in HeLa cell mobility and intercellular communication through tunneling tubes. BMC Cell Biol 2016; 17:3. [PMID: 26758208 PMCID: PMC4710989 DOI: 10.1186/s12860-016-0080-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 01/08/2016] [Indexed: 01/12/2023] Open
Abstract
Background Membranous tunneling tubes (TTs) are a recently discovered new form of communication between remote cells allowing their electrical synchronization, migration, and transfer of cellular materials. TTs have been identified in the brain and share similarities with neuronal processes. TTs can be open-ended, close-ended or contain functional gap junctions at the membrane interface. Gap junctions are formed of two unapposed hemichannels composed of six connexin (Cx) subunits. There are evidences that Cxs also play channel-independent role in cell adhesion, migration, division, differentiation, formation of neuronal networks and tumorigenicity. These properties of Cxs and TTs may synergetically determine the cellular and intercellular processes. Therefore, we examined the impact of Cxs expressed in the nervous system (Cx36, Cx40, Cx43, Cx45, and Cx47) on: 1) cell mobility; 2) formation and properties of TTs; and 3) transfer of siRNA between remote cells through TTs. Results We have identified two types of TTs between HeLa cells: F-actin rich only and containing F-actin and α-tubulin. The morphology of TTs was not influenced by expression of examined connexins; however, Cx36-EGFP-expressing cells formed more TTs while cells expressing Cx43-EGFP, Cx45, and Cx47 formed fewer TTs between each other compared with wt and Cx40-CFP-expressing cells. Also, Cx36-EGFP and Cx40-CFP-expressing HeLa cells were more mobile compared with wt and other Cxs-expressing cells. TTs containing Cx40-CFP, Cx43-EGFP, or Cx47 gap junctions were capable of transmitting double-stranded small interfering RNA; however, Cx36-EGFP and Cx45 were not permeable to it. In addition, we show that Cx43-EGFP-expressing HeLa cells and laryngeal squamous cell carcinoma cells can couple to the mesenchymal stem cells through TTs. Conclusions Different Cxs may modulate the mobility of cells and formation of TTs in an opposite manner; siRNA transfer through the GJ-containing TTs is Cx isoform-dependent. Electronic supplementary material The online version of this article (doi:10.1186/s12860-016-0080-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lina Rimkutė
- Institute of Cardiology, Lithuanian University of Health Sciences, 17 Sukilėlių Ave., 50009, Kaunas, Lithuania.
| | - Vaidas Jotautis
- Institute of Cardiology, Lithuanian University of Health Sciences, 17 Sukilėlių Ave., 50009, Kaunas, Lithuania.
| | - Alina Marandykina
- Institute of Cardiology, Lithuanian University of Health Sciences, 17 Sukilėlių Ave., 50009, Kaunas, Lithuania.
| | - Renata Sveikatienė
- Institute of Cardiology, Lithuanian University of Health Sciences, 17 Sukilėlių Ave., 50009, Kaunas, Lithuania.
| | - Ieva Antanavičiūtė
- Institute of Cardiology, Lithuanian University of Health Sciences, 17 Sukilėlių Ave., 50009, Kaunas, Lithuania.
| | - Vytenis Arvydas Skeberdis
- Institute of Cardiology, Lithuanian University of Health Sciences, 17 Sukilėlių Ave., 50009, Kaunas, Lithuania.
| |
Collapse
|
215
|
Tang BL. MIRO GTPases in Mitochondrial Transport, Homeostasis and Pathology. Cells 2015; 5:1. [PMID: 26729171 PMCID: PMC4810086 DOI: 10.3390/cells5010001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 01/08/2023] Open
Abstract
The evolutionarily-conserved mitochondrial Rho (MIRO) small GTPase is a Ras superfamily member with three unique features. It has two GTPase domains instead of the one found in other small GTPases, and it also has two EF hand calcium binding domains, which allow Ca(2+)-dependent modulation of its activity and functions. Importantly, it is specifically associated with the mitochondria and via a hydrophobic transmembrane domain, rather than a lipid-based anchor more commonly found in other small GTPases. At the mitochondria, MIRO regulates mitochondrial homeostasis and turnover. In metazoans, MIRO regulates mitochondrial transport and organization at cellular extensions, such as axons, and, in some cases, intercellular transport of the organelle through tunneling nanotubes. Recent findings have revealed a myriad of molecules that are associated with MIRO, particularly the kinesin adaptor Milton/TRAK, mitofusin, PINK1 and Parkin, as well as the endoplasmic reticulum-mitochondria encounter structure (ERMES) complex. The mechanistic aspects of the roles of MIRO and its interactors in mitochondrial homeostasis and transport are gradually being revealed. On the other hand, MIRO is also increasingly associated with neurodegenerative diseases that have roots in mitochondrial dysfunction. In this review, I discuss what is currently known about the cellular physiology and pathophysiology of MIRO functions.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD7, 8 Medical Drive, Singapore 117597, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore.
| |
Collapse
|
216
|
McCoy-Simandle K, Hanna SJ, Cox D. Exosomes and nanotubes: Control of immune cell communication. Int J Biochem Cell Biol 2015; 71:44-54. [PMID: 26704468 DOI: 10.1016/j.biocel.2015.12.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/01/2015] [Accepted: 12/10/2015] [Indexed: 12/19/2022]
Abstract
Cell-cell communication is critical to coordinate the activity and behavior of a multicellular organism. The cells of the immune system not only must communicate with similar cells, but also with many other cell types in the body. Therefore, the cells of the immune system have evolved multiple ways to communicate. Exosomes and tunneling nanotubes (TNTs) are two means of communication used by immune cells that contribute to immune functions. Exosomes are small membrane vesicles secreted by most cell types that can mediate intercellular communication and in the immune system they are proposed to play a role in antigen presentation and modulation of gene expression. TNTs are membranous structures that mediate direct cell-cell contact over several cell diameters in length (and possibly longer) and facilitate the interaction and/or the transfer of signals, material and other cellular organelles between connected cells. Recent studies have revealed additional, but sometimes conflicting, structural and functional features of both exosomes and TNTs. Despite the new and exciting information in exosome and TNT composition, origin and in vitro function, biologically significant functions are still being investigated and determined. In this review, we discuss the current field regarding exosomes and TNTs in immune cells providing evaluation and perspectives of the current literature.
Collapse
Affiliation(s)
- Kessler McCoy-Simandle
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Gruss MRRC 306, Bronx, NY 10461, USA.
| | - Samer J Hanna
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Gruss MRRC 306, Bronx, NY 10461, USA.
| | - Dianne Cox
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Gruss MRRC 306, Bronx, NY 10461, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Gruss MRRC 306, Bronx, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, 1300 Morris Park Ave, Gruss MRRC 306, Bronx, NY 10461, USA.
| |
Collapse
|
217
|
Epperla CP, Mohan N, Chang CW, Chen CC, Chang HC. Nanodiamond-Mediated Intercellular Transport of Proteins through Membrane Tunneling Nanotubes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:6097-105. [PMID: 26479149 DOI: 10.1002/smll.201502089] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/28/2015] [Indexed: 05/20/2023]
Abstract
Recently discovered tunneling nanotubes (TNTs) are capable of creating intercellular communication pathways through which transport of proteins and other cytoplasmic components occurs. Intercellular transport is related to many diseases and nanotubes are potentially useful as drug-delivery channels for cancer therapy. Here, we apply fluorescent nanodiamond (FND) as a photostable tracker, as well as a protein carrier, to illustrate the transport events in TNTs of human cells. Proteins, including bovine serum albumin and green fluorescent protein, are first coated on 100-nm FNDs by physical adsorption and then single-particle tracking of the bioconjugates in the transient membrane connections is carried out by fluorescence microscopy. Stop-and-go and to-and-fro motions mediated by molecular motors are found for the active transport of protein-loaded FNDs trapped in the endosomal vehicles of human embryonic kidney cells (HEK293T). Quantitative analysis of the heterotypical transport between HEK293T and SH-SY5Y neuroblastoma cells by flow cytometry confirm the formation of open-ended nanotubes between them, despite that their TNTs differ in structural components. Our results demonstrate the promising applications of this novel carbon-based nanomaterial for intercellular delivery of biomolecular cargo down to the single-particle level.
Collapse
Affiliation(s)
- Chandra Prakash Epperla
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
- Taiwan International Graduate Program - Molecular Science and Technology, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Nitin Mohan
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
| | - Che-Wei Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
- Department of Chemistry, National Taiwan Normal University, Taipei, 116, Taiwan
| | - Chia-Chun Chen
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
- Department of Chemistry, National Taiwan Normal University, Taipei, 116, Taiwan
| | - Huan-Cheng Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
- Taiwan International Graduate Program - Molecular Science and Technology, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106, Taiwan
| |
Collapse
|
218
|
Extracellular Vesicles: Evolving Factors in Stem Cell Biology. Stem Cells Int 2015; 2016:1073140. [PMID: 26649044 PMCID: PMC4663346 DOI: 10.1155/2016/1073140] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/09/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022] Open
Abstract
Stem cells are proposed to continuously secrete trophic factors that potentially serve as mediators of autocrine and paracrine activities, associated with reprogramming of the tumor microenvironment, tissue regeneration, and repair. Hitherto, significant efforts have been made to understand the level of underlying paracrine activities influenced by stem cell secreted trophic factors, as little is known about these interactions. Recent findings, however, elucidate this role by reporting the effects of stem cell derived extracellular vesicles (EVs) that mimic the phenotypes of the cells from which they originate. Exchange of genetic information utilizing persistent bidirectional communication mediated by stem cell-EVs could regulate stemness, self-renewal, and differentiation in stem cells and their subpopulations. This review therefore discusses stem cell-EVs as evolving communication factors in stem cell biology, focusing on how they regulate cell fates by inducing persistent and prolonged genetic reprogramming of resident cells in a paracrine fashion. In addition, we address the role of stem cell-secreted vesicles in shaping the tumor microenvironment and immunomodulation and in their ability to stimulate endogenous repair processes during tissue damage. Collectively, these functions ensure an enormous potential for future therapies.
Collapse
|
219
|
Understanding Fibroblasts in Order to Comprehend the Osteopathic Treatment of the Fascia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:860934. [PMID: 26357524 PMCID: PMC4556860 DOI: 10.1155/2015/860934] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/11/2014] [Accepted: 09/29/2014] [Indexed: 12/29/2022]
Abstract
The osteopathic treatment of the fascia involves several techniques, each aimed at allowing the various layers of the connective system to slide over each other, improving the responses of the afferents in case of dysfunction. However, before becoming acquainted with a method, one must be aware of the structure and function of the tissue that needs treating, in order to not only better understand the manual approach, but also make a more conscious choice of the therapeutic technique to employ, in order to adjust the treatment to the specific needs of the patient. This paper examines the current literature regarding the function and structure of the fascial system and its foundation, that is, the fibroblasts. These connective cells have many properties, including the ability to contract and to communicate with one another. They play a key role in the transmission of the tension produced by the muscles and in the management of the interstitial fluids. They are a source of nociceptive and proprioceptive information as well, which is useful for proper functioning of the body system. Therefore, the fibroblasts are an invaluable instrument, essential to the understanding of the therapeutic effects of osteopathic treatment. Scientific research should make greater efforts to better understand their functioning and relationships.
Collapse
|
220
|
Affiliation(s)
- Deepak Ramanujam
- From the Institut für Pharmakologie und Toxikologie, Technische Universität München (TUM), Munich, Germany (D.R., S.E.); and DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.E.)
| | - Stefan Engelhardt
- From the Institut für Pharmakologie und Toxikologie, Technische Universität München (TUM), Munich, Germany (D.R., S.E.); and DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.E.).
| |
Collapse
|
221
|
KLF5 promotes breast cancer proliferation, migration and invasion in part by upregulating the transcription of TNFAIP2. Oncogene 2015; 35:2040-51. [PMID: 26189798 DOI: 10.1038/onc.2015.263] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 05/17/2015] [Accepted: 06/05/2015] [Indexed: 12/31/2022]
Abstract
The Kruppel-like factor 5 (KLF5) transcription factor is highly expressed in high-grade and basal-like breast cancers. However, the mechanism by which KLF5 promotes cell migration and invasion is still not completely understood. In this study, we demonstrate that TNFAIP2, a tumor necrosis factor-α (TNFα)-induced gene, is a direct KLF5 target gene. The expression of TNFAIP2 is highly correlated with the expression of KLF5 in breast cancers. The manipulation of KLF5 expression positively alters TNFAIP2 expression levels. KLF5 directly binds to the TNFAIP2 gene promoter and activates its transcription. Functionally, KLF5 promotes cancer cell proliferation, migration and invasion in part through TNFAIP2. TNFAIP2 interacts with the two small GTPases Rac1 and Cdc42, thereby increasing their activities to change actin cytoskeleton and cell morphology. These findings collectively suggest that TNFAIP2 is a direct KLF5 target gene, and both KLF5 and TNFAIP2 promote breast cancer cell proliferation, migration and invasion through Rac1 and Cdc42.
Collapse
|
222
|
Sisakhtnezhad S, Khosravi L. Emerging physiological and pathological implications of tunneling nanotubes formation between cells. Eur J Cell Biol 2015; 94:429-43. [PMID: 26164368 DOI: 10.1016/j.ejcb.2015.06.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 12/21/2022] Open
Abstract
Cell-to-cell communication is a critical requirement to coordinate behaviors of the cells in a community and thereby achieve tissue homeostasis and conservation of the multicellular organisms. Tunneling nanotubes (TNTs), as a cell-to-cell communication over long distance, allow for bi- or uni-directional transfer of cellular components between cells. Identification of inducing agents and the cell and molecular mechanism underling the formation of TNTs and their structural and functional features may lead to finding new important roles for these intercellular bridges in vivo and in vitro. During the last decade, research has shown TNTs have different structural and functional properties, varying between and within cell systems. In this review, we will focus on TNTs and their cell and molecular mechanism of formation. Moreover, the latest findings into their functional roles in physiological and pathological processes, such as signal transduction, micro and nano-particles delivery, immune responses, embryogenesis, cellular reprogramming, apoptosis, cancer, and neurodegenerative diseases initiation and progression and pathogens transfer, will be discussed.
Collapse
Affiliation(s)
| | - Leila Khosravi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
223
|
Astanina K, Koch M, Jüngst C, Zumbusch A, Kiemer AK. Lipid droplets as a novel cargo of tunnelling nanotubes in endothelial cells. Sci Rep 2015; 5:11453. [PMID: 26095213 PMCID: PMC4476149 DOI: 10.1038/srep11453] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Intercellular communication is a fundamental process in the development and functioning of multicellular organisms. Recently, an essentially new type of intercellular communication, based on thin membrane channels between cells, has been reported. These structures, termed intercellular or tunnelling nanotubes (TNTs), permit the direct exchange of various components or signals (e.g., ions, proteins, or organelles) between non-adjacent cells at distances over 100 μm. Our studies revealed the presence of tunnelling nanotubes in microvascular endothelial cells (HMEC-1). The TNTs were studied with live cell imaging, environmental scanning electron microscopy (ESEM), and coherent anti-Stokes Raman scattering spectroscopy (CARS). Tunneling nanotubes showed marked persistence: the TNTs could connect cells over long distances (up to 150 μm) for several hours. Several cellular organelles were present in TNTs, such as lysosomes and mitochondria. Moreover, we could identify lipid droplets as a novel type of cargo in the TNTs. Under angiogenic conditions (VEGF treatment) the number of lipid droplets increased significantly. Arachidonic acid application not only increased the number of lipid droplets but also tripled the extent of TNT formation. Taken together, our results provide the first demonstration of lipid droplets as a cargo of TNTs and thereby open a new field in intercellular communication research.
Collapse
Affiliation(s)
- Ksenia Astanina
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Marcus Koch
- Leibniz Institute for New Materials, Saarbrücken, Germany
| | | | | | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| |
Collapse
|
224
|
Wang X, Yu X, Xie C, Tan Z, Tian Q, Zhu D, Liu M, Guan Y. Rescue of Brain Function Using Tunneling Nanotubes Between Neural Stem Cells and Brain Microvascular Endothelial Cells. Mol Neurobiol 2015; 53:2480-8. [DOI: 10.1007/s12035-015-9225-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/21/2015] [Indexed: 01/19/2023]
|
225
|
Abounit S, Delage E, Zurzolo C. Identification and Characterization of Tunneling Nanotubes for Intercellular Trafficking. ACTA ACUST UNITED AC 2015; 67:12.10.1-12.10.21. [PMID: 26061240 DOI: 10.1002/0471143030.cb1210s67] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Tunneling nanotubes (TNTs) are thin membranous channels providing direct cytoplasmic connection between remote cells. They are commonly observed in different cell cultures and increasing evidence supports their role in intercellular communication and pathogen transfer. However, the study of TNTs presents several pitfalls (e.g., difficulty in preserving such delicate structures, possible confusion with other protrusions, structural and functional heterogeneity, etc.) and therefore requires thoroughly designed approaches. The methods described in this unit represent a guideline for the characterization of TNTs (or TNT-like structures) in cell culture. Specifically, optimized protocols to (1) identify TNTs and the cytoskeletal elements present inside them; (2) evaluate TNT frequency in cell culture; (3) unambiguously distinguish them from other cellular connections or protrusions; and (4) monitor their formation in living cells are provided. Finally, this unit describes how to assess TNT-mediated cell-to-cell transfer of cellular components, which is a fundamental criterion for identifying functional TNTs.
Collapse
Affiliation(s)
- Saïda Abounit
- Unité de Traffic Membranaire et Pathogenèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, Paris, France.,These authors contributed equally to this work
| | - Elise Delage
- Unité de Traffic Membranaire et Pathogenèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, Paris, France.,These authors contributed equally to this work
| | - Chiara Zurzolo
- Unité de Traffic Membranaire et Pathogenèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, Paris, France.,Corresponding author
| |
Collapse
|
226
|
Climent M, Quintavalle M, Miragoli M, Chen J, Condorelli G, Elia L. TGFβ Triggers miR-143/145 Transfer From Smooth Muscle Cells to Endothelial Cells, Thereby Modulating Vessel Stabilization. Circ Res 2015; 116:1753-64. [PMID: 25801897 DOI: 10.1161/circresaha.116.305178] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/23/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE The miR-143/145 cluster is highly expressed in smooth muscle cells (SMCs), where it regulates phenotypic switch and vascular homeostasis. Whether it plays a role in neighboring endothelial cells (ECs) is still unknown. OBJECTIVE To determine whether SMCs control EC functions through passage of miR-143 and miR-145. METHODS AND RESULTS We used cocultures of SMCs and ECs under different conditions, as well as intact vessels to assess the transfer of miR-143 and miR-145 from one cell type to another. Imaging of cocultured cells transduced with fluorescent miRNAs suggested that miRNA transfer involves membrane protrusions known as tunneling nanotubes. Furthermore, we show that miRNA passage is modulated by the transforming growth factor (TGF) β pathway because both a specific transforming growth factor-β (TGFβ) inhibitor (SB431542) and an shRNA against TGFβRII suppressed the passage of miR-143/145 from SMCs to ECs. Moreover, miR-143 and miR-145 modulated angiogenesis by reducing the proliferation index of ECs and their capacity to form vessel-like structures when cultured on matrigel. We also identified hexokinase II (HKII) and integrin β 8 (ITGβ8)-2 genes essential for the angiogenic potential of ECs-as targets of miR-143 and miR-145, respectively. The inhibition of these genes modulated EC phenotype, similarly to miR-143 and miR-145 overexpression in ECs. These findings were confirmed by ex vivo and in vivo approaches, in which it was shown that TGFβ and vessel stress, respectively, triggered miR-143/145 transfer from SMCs to ECs. CONCLUSIONS Our results demonstrate that miR-143 and miR-145 act as communication molecules between SMCs and ECs to modulate the angiogenic and vessel stabilization properties of ECs.
Collapse
Affiliation(s)
- Montserrat Climent
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Manuela Quintavalle
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Michele Miragoli
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Ju Chen
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Gianluigi Condorelli
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.).
| | - Leonardo Elia
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.).
| |
Collapse
|
227
|
Biran A, Perelmutter M, Gal H, Burton DGA, Ovadya Y, Vadai E, Geiger T, Krizhanovsky V. Senescent cells communicate via intercellular protein transfer. Genes Dev 2015; 29:791-802. [PMID: 25854920 PMCID: PMC4403256 DOI: 10.1101/gad.259341.115] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/19/2015] [Indexed: 01/07/2023]
Abstract
Biran et al. show that senescent cells directly transfer proteins to neighboring cells and that this process facilitates immune surveillance of senescent cells by NK cells. The transfer is strictly dependent on cell–cell contact and CDC42-regulated actin polymerization and is mediated at least partially by cytoplasmic bridges. These findings reveal a novel mode of intercellular communication by which senescent cells regulate their immune surveillance and might impact tumorigenesis and tissue aging. Mammalian cells mostly rely on extracellular molecules to transfer signals to other cells. However, in stress conditions, more robust mechanisms might be necessary to facilitate cell–cell communications. Cellular senescence, a stress response associated with permanent exit from the cell cycle and the development of an immunogenic phenotype, limits both tumorigenesis and tissue damage. Paradoxically, the long-term presence of senescent cells can promote tissue damage and aging within their microenvironment. Soluble factors secreted from senescent cells mediate some of these cell-nonautonomous effects. However, it is unknown whether senescent cells impact neighboring cells by other mechanisms. Here we show that senescent cells directly transfer proteins to neighboring cells and that this process facilitates immune surveillance of senescent cells by natural killer (NK) cells. We found that transfer of proteins to NK and T cells is increased in the murine preneoplastic pancreas, a site where senescent cells are present in vivo. Proteomic analysis and functional studies of the transferred proteins revealed that the transfer is strictly dependent on cell–cell contact and CDC42-regulated actin polymerization and is mediated at least partially by cytoplasmic bridges. These findings reveal a novel mode of intercellular communication by which senescent cells regulate their immune surveillance and might impact tumorigenesis and tissue aging.
Collapse
Affiliation(s)
- Anat Biran
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Meirav Perelmutter
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hilah Gal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Dominick G A Burton
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ezra Vadai
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tamar Geiger
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv 69978, Israel
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
228
|
MitoCeption as a new tool to assess the effects of mesenchymal stem/stromal cell mitochondria on cancer cell metabolism and function. Sci Rep 2015; 5:9073. [PMID: 25766410 PMCID: PMC4358056 DOI: 10.1038/srep09073] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 01/28/2015] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial activity is central to tissue homeostasis. Mitochondria dysfunction constitutes a hallmark of many genetic diseases and plays a key role in tumor progression. The essential role of mitochondria, added to their recently documented capacity to transfer from cell to cell, obviously contributes to their current interest. However, determining the proper role of mitochondria in defined biological contexts was hampered by the lack of suitable experimental tools. We designed a protocol (MitoCeption) to directly and quantitatively transfer mitochondria, isolated from cell type A, to recipient cell type B. We validated and quantified the effective mitochondria transfer by imaging, fluorescence-activated cell sorting (FACS) and mitochondrial DNA analysis. We show that the transfer of minute amounts of mesenchymal stem/stromal cell (MSC) mitochondria to cancer cells, a process otherwise occurring naturally in coculture, results in cancer cell enhanced oxidative phosphorylation (OXPHOS) activity and favors cancer cell proliferation and invasion. The MitoCeption technique, which can be applied to different cell systems, will therefore be a method of choice to analyze the metabolic modifications induced by exogenous mitochondria in host cells.
Collapse
|
229
|
Wang X, Gerdes HH. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ 2015; 22:1181-91. [PMID: 25571977 PMCID: PMC4572865 DOI: 10.1038/cdd.2014.211] [Citation(s) in RCA: 347] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/29/2014] [Accepted: 11/14/2014] [Indexed: 12/18/2022] Open
Abstract
Tunneling nanotubes (TNTs) are F-actin-based membrane tubes that form between cells in culture and in tissues. They mediate intercellular communication ranging from electrical signalling to the transfer of organelles. Here, we studied the role of TNTs in the interaction between apoptotic and healthy cells. We found that pheochromocytoma (PC) 12 cells treated with ultraviolet light (UV) were rescued when cocultured with untreated PC12 cells. UV-treated cells formed a different type of TNT with untreated PC12 cells, which was characterized by continuous microtubule localized inside these TNTs. The dynamic behaviour of mCherry-tagged end-binding protein 3 and the accumulation of detyrosinated tubulin in these TNTs indicate that they are regulated structures. In addition, these TNTs show different biophysical properties, for example, increased diameter allowing dye entry, prolonged lifetime and decreased membrane fluidity. Further studies demonstrated that microtubule-containing TNTs were formed by stressed cells, which had lost cytochrome c but did not enter into the execution phase of apoptosis characterized by caspase-3 activation. Moreover, mitochondria colocalized with microtubules in TNTs and transited along these structures from healthy to stressed cells. Importantly, impaired formation of TNTs and untreated cells carrying defective mitochondria were unable to rescue UV-treated cells in the coculture. We conclude that TNT-mediated transfer of functional mitochondria reverse stressed cells in the early stages of apoptosis. This provides new insights into the survival mechanisms of damaged cells in a multicellular context.
Collapse
Affiliation(s)
- X Wang
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen 5009, Norway
| | - H-H Gerdes
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen 5009, Norway
| |
Collapse
|
230
|
Gözen I, Jesorka A. Lipid nanotube networks: Biomimetic Cell-to-Cell Communication and Soft-Matter Technology. NANOFABRICATION 2015. [DOI: 10.1515/nanofab-2015-0003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
231
|
Zhu S, Victoria GS, Marzo L, Ghosh R, Zurzolo C. Prion aggregates transfer through tunneling nanotubes in endocytic vesicles. Prion 2015; 9:125-35. [PMID: 25996400 PMCID: PMC4601206 DOI: 10.1080/19336896.2015.1025189] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 02/07/2015] [Accepted: 02/26/2015] [Indexed: 01/31/2023] Open
Abstract
Transmissible spongiform encephalopathies (TSEs) are a group of neurodegenerative diseases caused by the misfolding of the cellular prion protein to an infectious form PrP(Sc). The intercellular transfer of PrP(Sc) is a question of immediate interest as the cell-to-cell movement of the infectious particle causes the inexorable propagation of disease. We have previously identified tunneling nanotubes (TNTs) as one mechanism by which PrP(Sc) can move between cells. Here we investigate further the details of this mechanism and show that PrP(Sc) travels within TNTs in endolysosomal vesicles. Additionally we show that prion infection of CAD cells increases both the number of TNTs and intercellular transfer of membranous vesicles, thereby possibly playing an active role in its own intercellular transfer via TNTs.
Collapse
Key Words
- Ab, antibody
- CFP, cyan fluorescent protein
- ER, endoplasmic reticulum
- ERC, endocytic recycling compartment
- GFP, green fluorescent protein
- PM, plasma membrane
- PrPC, cellular prion protein
- PrPSc, scrapie prion protein
- RFP, red fluorescent protein
- TNTs, tunneling nanotubes
- TSEs, transmissible spongiform encephalopathies
- endosomes
- neuronal cells
- prion
- transfer
- tunneling nanotubes
Collapse
Affiliation(s)
- Seng Zhu
- Unité Trafic Membranaire et Pathogenese, Institut Pasteur, Paris CEDEX 15, France
| | | | - Ludovica Marzo
- Unité Trafic Membranaire et Pathogenese, Institut Pasteur, Paris CEDEX 15, France
| | - Rupam Ghosh
- Unité Trafic Membranaire et Pathogenese, Institut Pasteur, Paris CEDEX 15, France
| | - Chiara Zurzolo
- Unité Trafic Membranaire et Pathogenese, Institut Pasteur, Paris CEDEX 15, France
| |
Collapse
|
232
|
Antanavičiūtė I, Ereminienė E, Vysockas V, Račkauskas M, Skipskis V, Rysevaitė K, Treinys R, Benetis R, Jurevičius J, Skeberdis VA. Exogenous connexin43-expressing autologous skeletal myoblasts ameliorate mechanical function and electrical activity of the rabbit heart after experimental infarction. Int J Exp Pathol 2014; 96:42-53. [PMID: 25529770 DOI: 10.1111/iep.12109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 10/26/2014] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction is one of the major causes of mortality worldwide. For regeneration of the rabbit heart after experimentally induced infarction we used autologous skeletal myoblasts (SMs) due to their high proliferative potential, resistance to ischaemia and absence of immunological and ethical concerns. The cells were characterized with muscle-specific and myogenic markers. Cell transplantation was performed by injection of cell suspension (0.5 ml) containing approximately 6 million myoblasts into the infarction zone. The animals were divided into four groups: (i) no injection; (ii) sham injected; (iii) injected with wild-type SMs; and (iv) injected with SMs expressing connexin43 fused with green fluorescent protein (Cx43EGFP). Left ventricular ejection fraction (LVEF) was evaluated by 2D echocardiography in vivo before infarction, when myocardium has stabilized after infarction, and 3 months after infarction. Electrical activity in the healthy and infarction zones of the heart was examined ex vivo in Langendorff-perfused hearts by optical mapping using di-4-ANEPPS, a potential sensitive fluorescent dye. We demonstrate that SMs in the coculture can couple electrically not only to abutted but also to remote acutely isolated allogenic cardiac myocytes through membranous tunnelling tubes. The beneficial effect of cellular therapy on LVEF and electrical activity was observed in the group of animals injected with Cx43EGFP-expressing SMs. L-type Ca(2+) current amplitude was approximately fivefold smaller in the isolated SMs compared to healthy myocytes suggesting that limited recovery of LVEF may be related to inadequate expression or function of L-type Ca(2+) channels in transplanted differentiating SMs.
Collapse
Affiliation(s)
- Ieva Antanavičiūtė
- Institute of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Lachambre S, Chopard C, Beaumelle B. Preliminary characterisation of nanotubes connecting T-cells and their use by HIV-1. Biol Cell 2014; 106:394-404. [DOI: 10.1111/boc.201400037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/07/2014] [Indexed: 01/06/2023]
Affiliation(s)
- Simon Lachambre
- CPBS, UMR 5236 CNRS-Université de Montpellier; Route de Mende; Montpellier 34293 France
| | - Christophe Chopard
- CPBS, UMR 5236 CNRS-Université de Montpellier; Route de Mende; Montpellier 34293 France
| | - Bruno Beaumelle
- CPBS, UMR 5236 CNRS-Université de Montpellier; Route de Mende; Montpellier 34293 France
| |
Collapse
|
234
|
Evidence for Transfer of Membranes from Mesenchymal Stem Cells to HL-1 Cardiac Cells. Stem Cells Int 2014; 2014:653734. [PMID: 25295065 PMCID: PMC4175751 DOI: 10.1155/2014/653734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/13/2014] [Accepted: 08/18/2014] [Indexed: 01/01/2023] Open
Abstract
This study examined the interaction of mouse bone marrow mesenchymal stem cells (MSC) with cardiac HL-1 cells during coculture by fluorescent dye labeling and then flow cytometry. MSC were layered onto confluent HL-1 cell cultures in a 1 : 4 ratio. MSC gained gap junction permeant calcein from HL-1 cells after 4 hours which was partially reduced by oleamide. After 20 hours, 99% MSC gained calcein, unaffected by oleamide. Double-labeling HL-1 cells with calcein and the membrane dye DiO resulted in transfer of both calcein and DiO to MSC. When HL-1 cells were labeled with calcein and MSC with DiO, MSC gained calcein while HL-1 cells gained DiO. Very little fusion was observed since more than 90% Sca-1 positive MSC gained DiO from HL-1 cells while less than 9% gained gap junction impermeant CMFDA after 20 hours with no Sca-1 transfer to HL-1 cells. Time dependent transfer of membrane DiD was observed from HL-1 cells to MSC (100%) and vice versa (50%) after 20 hours with more limited transfer of CMFDA. These results demonstrate that MSC and HL-1 cells exchange membrane components which may account for some of the beneficial effect of MSC in the heart after myocardial infarction.
Collapse
|
235
|
Ranzinger J, Rustom A, Heide D, Morath C, Schemmer P, Nawroth PP, Zeier M, Schwenger V. The receptor for advanced glycation end-products (RAGE) plays a key role in the formation of nanotubes (NTs) between peritoneal mesothelial cells and in murine kidneys. Cell Tissue Res 2014; 357:667-679. [PMID: 24870978 DOI: 10.1007/s00441-014-1904-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 04/28/2014] [Indexed: 10/25/2022]
Abstract
The receptor for advanced glycation end-products (RAGE), a multiligand receptor of the immunoglobulin superfamily, takes part in various inflammatory processes. The role of this receptor in the context of intercellular communication, like nanotube (NT)-mediated interaction, is largely unknown. Here, we use cell cultures of human and murine peritoneal mesothelial cells as well as murine kidneys from wild-type and RAGE knockout mouse models to assess the role of RAGE in NT formation and function. We show that loss of RAGE function results in reduced NT numbers under physiological conditions and demonstrate the involvement of MAP kinase signaling in NT formation. Additionally, we show for the first time the existence of NTs in murine kidney tissue and confirm the correlation of RAGE expression and NT numbers. Under elevated oxidative stress conditions like renal ischemia or peritoneal dialysis, we demonstrate that RAGE absence does not prevent NT formation. Rather, increased NT numbers and attenuated kidney tissue damage could be observed, indicating that, depending on the predominant conditions, RAGE affects NT formation with implications for cellular communication.
Collapse
Affiliation(s)
- Julia Ranzinger
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany,
| | | | | | | | | | | | | | | |
Collapse
|
236
|
Macaulay AD, Gilbert I, Caballero J, Barreto R, Fournier E, Tossou P, Sirard MA, Clarke HJ, Khandjian ÉW, Richard FJ, Hyttel P, Robert C. The gametic synapse: RNA transfer to the bovine oocyte. Biol Reprod 2014; 91:90. [PMID: 25143353 DOI: 10.1095/biolreprod.114.119867] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Even after several decades of quiescent storage in the ovary, the female germ cell is capable of reinitiating transcription to build the reserves that are essential to support early embryonic development. In the current model of mammalian oogenesis, there exists bilateral communication between the gamete and the surrounding cells that is limited to paracrine signaling and direct transfer of small molecules via gap junctions existing at the end of the somatic cells' projections that are in contact with the oolemma. The purpose of this work was to explore the role of cumulus cell projections as a means of conductance of large molecules, including RNA, to the mammalian oocyte. By studying nascent RNA with confocal and transmission electron microscopy in combination with transcript detection, we show that the somatic cells surrounding the fully grown bovine oocyte contribute to the maternal reserves by actively transferring large cargo, including mRNA and long noncoding RNA. This occurrence was further demonstrated by the reconstruction of cumulus-oocyte complexes with transfected cumulus cells transferring a synthetic transcript. We propose selective transfer of transcripts occurs, the delivery of which is supported by a remarkable synapselike vesicular trafficking connection between the cumulus cells and the gamete. This unexpected exogenous contribution to the maternal stores offers a new perspective on the determinants of female fertility.
Collapse
Affiliation(s)
- Angus D Macaulay
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Isabelle Gilbert
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Julieta Caballero
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Rodrigo Barreto
- Veterinarian Medicine Department, São Paulo University, São Paulo, Brazil
| | - Eric Fournier
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Prudencio Tossou
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Marc-André Sirard
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University Health Centre, Montréal, Québec, Canada
| | - Édouard W Khandjian
- Département de Psychiatrie et Neurosciences, Institut universitaire en santé mentale de Québec, Université Laval, Québec City, Québec, Canada
| | - Francois J Richard
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claude Robert
- Département des sciences animales, Centre de recherche en biologie de la reproduction, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
237
|
Kaur S, Singh SP, Elkahloun AG, Wu W, Abu-Asab MS, Roberts DD. CD47-dependent immunomodulatory and angiogenic activities of extracellular vesicles produced by T cells. Matrix Biol 2014; 37:49-59. [PMID: 24887393 PMCID: PMC6176487 DOI: 10.1016/j.matbio.2014.05.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 05/15/2014] [Accepted: 05/22/2014] [Indexed: 11/20/2022]
Abstract
Intercellular communication is critical for integrating complex signals in multicellular eukaryotes. Vascular endothelial cells and T lymphocytes closely interact during the recirculation and trans-endothelial migration of T cells. In addition to direct cell-cell contact, we show that T cell derived extracellular vesicles can interact with endothelial cells and modulate their cellular functions. Thrombospondin-1 and its receptor CD47 are expressed on exosomes/ectosomes derived from T cells, and these extracellular vesicles are internalized and modulate signaling in both T cells and endothelial cells. Extracellular vesicles released from cells expressing or lacking CD47 differentially regulate activation of T cells induced by engaging the T cell receptor. Similarly, T cell-derived extracellular vesicles modulate endothelial cell responses to vascular endothelial growth factor and tube formation in a CD47-dependent manner. Uptake of T cell derived extracellular vesicles by recipient endothelial cells globally alters gene expression in a CD47-dependent manner. CD47 also regulates the mRNA content of extracellular vesicles in a manner consistent with some of the resulting alterations in target endothelial cell gene expression. Therefore, the thrombospondin-1 receptor CD47 directly or indirectly regulates intercellular communication mediated by the transfer of extracellular vesicles between vascular cells.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20982, USA
| | - Satya P Singh
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20982, USA
| | - Abdel G Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiwei Wu
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mones S Abu-Asab
- Histopathology Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20982, USA.
| |
Collapse
|
238
|
Antanavičiūtė I, Rysevaitė K, Liutkevičius V, Marandykina A, Rimkutė L, Sveikatienė R, Uloza V, Skeberdis VA. Long-distance communication between laryngeal carcinoma cells. PLoS One 2014; 9:e99196. [PMID: 24945745 PMCID: PMC4063716 DOI: 10.1371/journal.pone.0099196] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/12/2014] [Indexed: 01/22/2023] Open
Abstract
Tunneling nanotubes and epithelial bridges are recently discovered new forms of intercellular communication between remote cells allowing their electrical synchronization, transfer of second messengers and even membrane vesicles and organelles. In the present study, we demonstrate for the first time in primary cell cultures prepared from human laryngeal squamous cell carcinoma (LSCC) samples that these cells communicate with each other over long distances (up to 1 mm) through membranous tunneling tubes (TTs), which can be open-ended or contain functional gap junctions formed of connexin 43. We found two types of TTs, containing F-actin alone or F-actin and α-tubulin. In the LSCC cell culture, we identified 5 modes of TT formation and performed quantitative assessment of their electrical properties and permeability to fluorescent dyes of different molecular weight and charge. We show that TTs, containing F-actin and α-tubulin, transport mitochondria and accommodate small DAPI-positive vesicles suggesting possible transfer of genetic material through TTs. We confirmed this possibility by demonstrating that even TTs, containing gap junctions, were capable of transmitting double-stranded small interfering RNA. To support the idea that the phenomenon of TTs is not only typical of cell cultures, we have examined microsections of samples obtained from human LSCC tissues and identified intercellular structures similar to those found in the primary LSCC cell culture.
Collapse
Affiliation(s)
- Ieva Antanavičiūtė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kristina Rysevaitė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Institute of Anatomy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vykintas Liutkevičius
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Alina Marandykina
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Lina Rimkutė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Renata Sveikatienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Virgilijus Uloza
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | |
Collapse
|
239
|
Arrevillaga-Boni G, Hernández-Ruiz M, Castillo EC, Ortiz-Navarrete V. Intercellular communication through contacts between continuous pseudopodial extensions in a macrophage-like cell line. CELL COMMUNICATION & ADHESION 2014; 21:213-20. [PMID: 24896643 DOI: 10.3109/15419061.2014.923993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cell-to-cell information exchange mediated by membrane protrusions in tunneling nanotubes (TNTs) has been widely described in distinct cell lines. Here, we describe a new form of direct intercellular communication in a murine macrophage-like cell line that is mediated by pseudopodial fusions that form over scraped plastic tissue culture surfaces along scratch lines. These structures are capable of forming intercellular, tunnel-like channels (inter-pseudopodial axis connections) that can be differentiated from TNTs based on length, thickness, tandem arrangement along an axis, pseudopodial origin and permanency. These channels were able to exchange membrane lipids and contain particles 0.5 μm or lesser in diameter between cells and might represent an additional biological function of pseudopodia.
Collapse
Affiliation(s)
- Gerardo Arrevillaga-Boni
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) , Avenida IPN No. 2508, Colonia San Pedro Zacatenco, México, D. F. , Mexico
| | | | | | | |
Collapse
|
240
|
de Cavanagh EMV, González SA, Inserra F, Forcada P, Castellaro C, Chiabaut-Svane J, Obregón S, Casarini MJ, Kempny P, Kotliar C. Sympathetic predominance is associated with impaired endothelial progenitor cells and tunneling nanotubes in controlled-hypertensive patients. Am J Physiol Heart Circ Physiol 2014; 307:H207-15. [PMID: 24858852 DOI: 10.1152/ajpheart.00955.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Early endothelial progenitor cells (early EPC) and late EPC are involved in endothelial repair and can rescue damaged endothelial cells by transferring organelles through tunneling nanotubes (TNT). In rodents, EPC mobilization from the bone marrow depends on sympathetic nervous system activity. Indirect evidence suggests a relation between autonomic derangements and human EPC mobilization. We aimed at testing whether hypertension-related autonomic imbalances are associated with EPC impairment. Thirty controlled-essential hypertensive patients [systolic blood pressure/diastolic blood pressure = 130(120-137)/85(61-88) mmHg; 81.8% male] and 20 healthy normotensive subjects [114(107-119)/75(64-79) mmHg; 80% male] were studied. Mononuclear cells were cultured on fibronectin- and collagen-coated dishes for early EPC and late EPC, respectively. Low (LF)- and high (HF)-frequency components of short-term heart rate variability were analyzed during a 5-min rest, an expiration/inspiration maneuver, and a Stroop color-word test. Modulations of cardiac sympathetic and parasympathetic activities were evaluated by LF/HF (%) and HF power (ms(2)), respectively. In controlled-hypertensive patients, the numbers of early EPC, early EPC that emitted TNT, late EPC, and late EPC that emitted TNT were 41, 77, 50, and 88% lower than in normotensive subjects (P < 0.008), respectively. In controlled-hypertensive patients, late EPC number was positively associated with cardiac parasympathetic reserve during the expiration/inspiration maneuver (rho = 0.45, P = 0.031) and early EPC with brachial flow-mediated dilation (rho = 0.655; P = 0.049); also, late TNT number was inversely related to cardiac sympathetic response during the stress test (rho = -0.426, P = 0.045). EPC exposure to epinephrine or norepinephrine showed negative dose-response relationships on cell adhesion to fibronectin and collagen; both catecholamines stimulated early EPC growth, but epinephrine inhibited late EPC growth. In controlled-hypertensive patients, sympathetic overactivity/parasympathetic underactivity were negatively associated with EPC, suggesting that reducing sympathetic/increasing parasympathetic activation might favor endothelial repair.
Collapse
Affiliation(s)
- Elena M V de Cavanagh
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Sergio A González
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Felipe Inserra
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and
| | - Pedro Forcada
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Carlos Castellaro
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Jorge Chiabaut-Svane
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Sebastián Obregón
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - María Jesús Casarini
- Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Pablo Kempny
- Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| | - Carol Kotliar
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and Arterial Hypertension Center, Cardiology Department, Austral University Hospital, Buenos Aires, Argentina
| |
Collapse
|
241
|
Lee JY. New and old roles of plasmodesmata in immunity and parallels to tunneling nanotubes. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2014; 221-222:13-20. [PMID: 24656331 PMCID: PMC4147083 DOI: 10.1016/j.plantsci.2014.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 05/08/2023]
Abstract
Effective cell-to-cell communication is critical for the survival of both unicellular and multicellular organisms. In multicellular plants, direct cell coupling across the cell wall boundaries is mediated by long membrane-lined cytoplasmic bridges, the plasmodesmata. Exciting recent discoveries suggest that the occurrence of such membrane-lined intercellular channels is not unique to plant lineages but more prevalent across biological kingdoms than previously assumed. Striking functional analogies exist among those channels, in that not only do they all facilitate the exchange of various forms of macromolecules, but also they are exploited by some opportunistic pathogens to spread infection from one host cell to another. However, host cells may have also evolved strategies to offset such exploitation of the critical cellular infrastructure by the pathogen. Indeed, recent studies support an emerging paradigm that cellular connectivity via plasmodesmata plays an important role in innate immune responses. Preliminary hypotheses are proposed as to how various regulatory mechanisms integrating plasmodesmata into immune signaling pathways may have evolved.
Collapse
Affiliation(s)
- Jung-Youn Lee
- Department of Plant and Soil Sciences, Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA.
| |
Collapse
|
242
|
Abstract
The extracellular matrix (ECM) is best known for its function as a structural scaffold for the tissue and more recently as a microenvironment to sequester growth factors and cytokines allowing for rapid and localized changes in their activity in the absence of new protein synthesis. In this review, we explore this and additional new aspects of ECM function in mediating cell-to-cell communications. Fibrillar and nonfibrillar components of ECM can limit and facilitate the transport of molecules through the extracellular space while also regulating interstitial hydrostatic pressure. In turn, transmembrane communications via molecules, such as ECM metalloproteinase inducer, thrombospondins, and integrins, can further mediate cell response to extracellular cues and affect ECM composition and tissue remodeling. Other means of cell-to-cell communication include extracellular microRNA transport and its contribution to gene expression in target cells and the nanotube formation between distant cells, which has recently emerged as a novel conduit for intercellular organelle sharing thereby influencing cell survival and function. The information summarized and discussed here are not limited to the cardiovascular ECM but encompass ECM in general with specific references to the cardiovascular system.
Collapse
Affiliation(s)
- Dong Fan
- From the Department of Physiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (D.F., Z.K.); and Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (E.E.C.)
| | | | | |
Collapse
|
243
|
Austefjord MW, Gerdes HH, Wang X. Tunneling nanotubes: Diversity in morphology and structure. Commun Integr Biol 2014; 7:e27934. [PMID: 24778759 PMCID: PMC3995728 DOI: 10.4161/cib.27934] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 02/06/2023] Open
Abstract
Tunneling nanotubes (TNTs) are recently discovered thin membranous tubes that interconnect cells. During the last decade, research has shown TNTs to be diverse in morphology and composition, varying between and within cell systems. In addition, the discovery of TNT-like extracellular protrusions, as well as observations of TNTs in vivo, has further enriched our knowledge on the diversity of TNT-like structures. Considering the complex molecular mechanisms underlying the formation of TNTs, as well as their different functions in intercellular communication, it is important to decipher how heterogeneity of TNTs is established, and to address what roles the compositional elements have in the execution of various functions. Here, we review the current knowledge on the morphological and structural diversity of TNTs, and address the relation between the formation, the structure, and the function of TNTs.
Collapse
Affiliation(s)
| | | | - Xiang Wang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
244
|
Delage E, Zurzolo C. Exploring the role of lipids in intercellular conduits: breakthroughs in the pipeline. FRONTIERS IN PLANT SCIENCE 2013; 4:504. [PMID: 24368909 PMCID: PMC3857720 DOI: 10.3389/fpls.2013.00504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 11/25/2013] [Indexed: 05/08/2023]
Abstract
It has been known for more than a century that most of the plant cells are connected to their neighbors through membranous pores perforating the cell wall, namely plasmodesmata (PDs). The recent discovery of tunneling nanotubes (TNTs), thin membrane bridges established between distant mammalian cells, suggests that intercellular communication mediated through cytoplasmic continuity could be a conserved feature of eukaryotic organisms. Although TNTs differ from PDs in their formation and architecture, both are characterized by a continuity of the plasma membrane between two cells, delimiting a nanotubular channel supported by actin-based cytoskeleton. Due to this unusual membrane organization, lipids are likely to play critical roles in the formation and stability of intercellular conduits like TNTs and PDs, but also in regulating the transfer through these structures. While it is crucial for a better understanding of those fascinating communication highways, the study of TNT lipid composition and dynamics turned out to be extremely challenging. The present review aims to give an overview of the recent findings in this context. We will also discuss some of the promising imaging approaches, which might be the key for future breakthroughs in the field and could also benefit the research on PDs.
Collapse
Affiliation(s)
- Elise Delage
- *Correspondence: Elise Delage and Chiara Zurzolo, Unité de Trafic Membranaire et Pathogenèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, 25, Rue du Docteur Roux, 75724 Paris Cedex 15, France e-mail: ;
| | - Chiara Zurzolo
- *Correspondence: Elise Delage and Chiara Zurzolo, Unité de Trafic Membranaire et Pathogenèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, 25, Rue du Docteur Roux, 75724 Paris Cedex 15, France e-mail: ;
| |
Collapse
|
245
|
LEHMANN TOMASZP, FILIPIAK KRYSTYNA, JUZWA WOJCIECH, SUJKA-KORDOWSKA PATRYCJA, JAGODZIŃSKI PAWEŁP, ZABEL MACIEJ, GŁOWACKI JAKUB, MISTERSKA EWA, WALCZAK MICHAŁ, GŁOWACKI MACIEJ. Co-culture of human nucleus pulposus cells with multipotent mesenchymal stromal cells from human bone marrow reveals formation of tunnelling nanotubes. Mol Med Rep 2013; 9:574-82. [DOI: 10.3892/mmr.2013.1821] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 11/01/2013] [Indexed: 11/06/2022] Open
|
246
|
Abstract
A wide spectrum of pathogenic bacteria and protozoa has adapted to an intracellular life-style, which presents several advantages, including accessibility to host cell metabolites and protection from the host immune system. Intracellular pathogens have developed strategies to enter and exit their host cells while optimizing survival and replication, progression through the life cycle, and transmission. Over the last decades, research has focused primarily on entry, while the exit process has suffered from neglect. However, pathogen exit is of fundamental importance because of its intimate association with dissemination, transmission, and inflammation. Hence, to fully understand virulence mechanisms of intracellular pathogens at cellular and systemic levels, it is essential to consider exit mechanisms to be a key step in infection. Exit from the host cell was initially viewed as a passive process, driven mainly by physical stress as a consequence of the explosive replication of the pathogen. It is now recognized as a complex, strategic process termed "egress," which is just as well orchestrated and temporally defined as entry into the host and relies on a dynamic interplay between host and pathogen factors. This review compares egress strategies of bacteria, pathogenic yeast, and kinetoplastid and apicomplexan parasites. Emphasis is given to recent advances in the biology of egress in mycobacteria and apicomplexans.
Collapse
|
247
|
Bornschlögl T. How filopodia pull: what we know about the mechanics and dynamics of filopodia. Cytoskeleton (Hoboken) 2013; 70:590-603. [PMID: 23959922 DOI: 10.1002/cm.21130] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 01/04/2023]
Abstract
In recent years, the dynamic, hair-like cell protrusions called filopodia have attracted considerable attention. They have been found in a multitude of different cell types and are often called "sensory organelles," since they seem to sense the mechanical and chemical environment of a cell. Once formed, filopodia can exhibit complex behavior, they can grow and retract, push or pull, and transform into distinct structures. They are often found to make first adhesive contact with the extracellular matrix, pathogens or with adjacent cells, and to subsequently exert pulling forces. Much is known about the cytoskeletal players involved in filopodia formation, but only recently have we started to explore the mechanics of filopodia together with the related cytoskeletal dynamics. This review summarizes current advancements in our understanding of the mechanics and dynamics of filopodia, with a focus on the molecular mechanisms behind filopodial force exertion.
Collapse
Affiliation(s)
- Thomas Bornschlögl
- Institut Curie, Laboratoire, Physico-Chimie UMR CNRS, 168, 11 Rue Pierre et Marie Curie, 75005, Paris, France
| |
Collapse
|
248
|
Intracellular trafficking of solid lipid nanoparticles and their distribution between cells through tunneling nanotubes. Eur J Pharm Sci 2013; 50:139-48. [DOI: 10.1016/j.ejps.2013.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 04/11/2013] [Accepted: 04/12/2013] [Indexed: 12/18/2022]
|
249
|
Chen CC, Liu HP, Chao M, Liang Y, Tsang NM, Huang HY, Wu CC, Chang YS. NF-κB-mediated transcriptional upregulation of TNFAIP2 by the Epstein-Barr virus oncoprotein, LMP1, promotes cell motility in nasopharyngeal carcinoma. Oncogene 2013; 33:3648-59. [PMID: 23975427 DOI: 10.1038/onc.2013.345] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/01/2013] [Accepted: 07/22/2013] [Indexed: 12/21/2022]
Abstract
Nasopharyngeal carcinoma (NPC), which is closely associated with Epstein-Barr virus (EBV), is a metastasis-prone epithelial cancer. We previously showed that tumor necrosis factor α-induced protein 2 (TNFAIP2) is highly expressed in NPC tumor tissues and is correlated with metastasis and poor survival in NPC patients. However, the underlying mechanism remains unclear. In this study, we demonstrate that the EBV oncoprotein, latent membrane protein 1 (LMP1), can transcriptionally induce TNFAIP2 expression via NF-κB. Quantitative RT-PCR and western blotting revealed that LMP1 induces TNFAIP2 expression through its C-terminal-activating region (CTAR2) domain, which is required for transduction of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) signaling. Inhibition of NF-κB activation or depletion of p65 (a component of NF-κB) by RNA interference abolished the LMP1-induced expression of TNFAIP2, whereas ectopic expression of p65 was sufficient to induce TNFAIP2 expression. Luciferase reporter assays showed that LMP1 transcriptionally induces TNFAIP2 expression through a newly identified NF-κB-binding site within the TNFAIP2 promoter (-3,869 to -3,860 bp). Immunohistochemical analysis of NPC biopsy specimens further revealed a significant correlation between the protein levels of TNFAIP2 and activated p65 (R=0.689, P<0.001), indicating that our findings are clinically relevant. Immunofluorescence microscopy and co-immunoprecipitation assays showed that TNFAIP2 associates with actin and is involved in the formation of actin-based membrane protrusions. Furthermore, transwell migration assays demonstrated that TNFAIP2 contributes to LMP1-induced cell motility. Collectively, these findings provide novel insights into the regulation of TNFAIP2 and its role in promoting NPC tumor progression.
Collapse
Affiliation(s)
- C-C Chen
- Chang Gung Molecular Medicine Research Center, Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-Shan, Taiwan
| | - H-P Liu
- Chang Gung Molecular Medicine Research Center, Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-Shan, Taiwan
| | - M Chao
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-Shan, Taiwan
| | - Y Liang
- Chang Gung Molecular Medicine Research Center, Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-Shan, Taiwan
| | - N-M Tsang
- Departments of Radiation Oncology, Chang Gung Memorial Hospital at Lin-Kou, Kwei-Shan, Taiwan
| | - H-Y Huang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-Shan, Taiwan
| | - C-C Wu
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Kwei-Shan, Taiwan
| | - Y-S Chang
- Chang Gung Molecular Medicine Research Center, Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-Shan, Taiwan
| |
Collapse
|
250
|
Gousset K, Marzo L, Commere PH, Zurzolo C. Myo10 is a key regulator of TNT formation in neuronal cells. J Cell Sci 2013; 126:4424-35. [PMID: 23886947 DOI: 10.1242/jcs.129239] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cell-to-cell communication is essential in multicellular organisms. Tunneling nanotubes (TNTs) have emerged as a new type of intercellular spreading mechanism allowing the transport of various signals, organelles and pathogens. Here, we study the role of the unconventional molecular motor myosin-X (Myo10) in the formation of functional TNTs within neuronal CAD cells. Myo10 protein expression increases the number of TNTs and the transfer of vesicles between co-cultured cells. We also show that TNT formation requires both the motor and tail domains of the protein, and identify the F2 lobe of the FERM domain within the Myo10 tail as necessary for TNT formation. Taken together, these results indicate that, in neuronal cells, TNTs can arise from a subset of Myo10-driven dorsal filopodia, independent of its binding to integrins and N-cadherins. In addition our data highlight the existence of different mechanisms for the establishment and regulation of TNTs in neuronal cells and other cell types.
Collapse
Affiliation(s)
- Karine Gousset
- Institut Pasteur, 25 Rue du Dr Roux, Unité de Traffic Membranaire et Pathogenèse, 75724 Paris Cedex 15, France
| | | | | | | |
Collapse
|