151
|
Mittal R, Karhu E, Wang JS, Delgado S, Zukerman R, Mittal J, Jhaveri VM. Cell communication by tunneling nanotubes: Implications in disease and therapeutic applications. J Cell Physiol 2018; 234:1130-1146. [PMID: 30206931 DOI: 10.1002/jcp.27072] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Intercellular communication is essential for the development and maintenance of multicellular organisms. Tunneling nanotubes (TNTs) are a recently recognized means of long and short distance communication between a wide variety of cell types. TNTs are transient filamentous membrane protrusions that connect cytoplasm of neighboring or distant cells. Cytoskeleton fiber-mediated transport of various cargoes occurs through these tubules. These cargoes range from small ions to whole organelles. TNTs have been shown to contribute not only to embryonic development and maintenance of homeostasis, but also to the spread of infectious particles and resistance to therapies. These functions in the development and progression of cancer and infectious disease have sparked increasing scrutiny of TNTs, as their contribution to disease progression lends them a promising therapeutic target. Herein, we summarize the current knowledge of TNT structure and formation as well as the role of TNTs in pathology, focusing on viral, prion, and malignant disease. We then discuss the therapeutic possibilities of TNTs in light of their varied functions. Despite recent progress in the growing field of TNT research, more studies are needed to precisely understand the role of TNTs in pathological conditions and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Elisa Karhu
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Jay-Shing Wang
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Stefanie Delgado
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Ryan Zukerman
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Vasanti M Jhaveri
- Department of Otolaryngology, University of Miami-Miller School of Medicine, Miami, Florida
| |
Collapse
|
152
|
Yamashita YM, Inaba M, Buszczak M. Specialized Intercellular Communications via Cytonemes and Nanotubes. Annu Rev Cell Dev Biol 2018; 34:59-84. [PMID: 30074816 DOI: 10.1146/annurev-cellbio-100617-062932] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years, thin membrane protrusions such as cytonemes and tunneling nanotubes have emerged as a novel mechanism of intercellular communication. Protrusion-based cellular interactions allow for specific communication between participating cells and have a distinct spectrum of advantages compared to secretion- and diffusion-based intercellular communication. Identification of protrusion-based signaling in diverse systems suggests that this mechanism is a ubiquitous and prevailing means of communication employed by many cell types. Moreover, accumulating evidence indicates that protrusion-based intercellular communication is often involved in pathogenesis, including cancers and infections. Here we review our current understanding of protrusion-based intercellular communication.
Collapse
Affiliation(s)
- Yukiko M Yamashita
- Life Sciences Institute, Department of Cell and Developmental Biology, and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Mayu Inaba
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA;
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| |
Collapse
|
153
|
Zhu S, Abounit S, Korth C, Zurzolo C. Transfer of disrupted-in-schizophrenia 1 aggregates between neuronal-like cells occurs in tunnelling nanotubes and is promoted by dopamine. Open Biol 2018; 7:rsob.160328. [PMID: 28275106 PMCID: PMC5376705 DOI: 10.1098/rsob.160328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/10/2017] [Indexed: 12/22/2022] Open
Abstract
The disrupted-in-schizophrenia 1 (DISC1) gene was identified as a genetic risk factor for chronic mental illnesses (CMI) such as schizophrenia, bipolar disorder and severe recurrent depression. Insoluble aggregated DISC1 variants were found in the cingular cortex of sporadic, i.e. non-genetic, CMI patients. This suggests protein pathology as a novel, additional pathogenic mechanism, further corroborated in a recent transgenic rat model presenting DISC1 aggregates. Since the potential role of aggregation of DISC1 in sporadic CMI is unknown, we investigated whether DISC1 undergoes aggregation in cell culture and could spread between neuronal cells in a prion-like manner, as shown for amyloid proteins in neurodegenerative diseases. Co-culture experiments between donor cells forming DISC1 aggregates and acceptor cells showed that 4.5% of acceptor cells contained donor-derived DISC1 aggregates, thus indicating an efficient transfer in vitro. DISC1 aggregates were found inside tunnelling nanotubes (TNTs) and transfer was enhanced by increasing TNT formation and notably by dopamine treatment, which also induces DISC1 aggregation. These data indicate that DISC1 aggregates can propagate between cells similarly to prions, thus providing some molecular basis for the role of protein pathology in CMI.
Collapse
Affiliation(s)
- Seng Zhu
- Institut Pasteur, Membrane Traffic and Pathogenesis Unit, 25-28 rue du Docteur Roux, 75724 Paris, France
| | - Saïda Abounit
- Institut Pasteur, Membrane Traffic and Pathogenesis Unit, 25-28 rue du Docteur Roux, 75724 Paris, France
| | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Chiara Zurzolo
- Institut Pasteur, Membrane Traffic and Pathogenesis Unit, 25-28 rue du Docteur Roux, 75724 Paris, France
| |
Collapse
|
154
|
Maxan A, Cicchetti F. Tau: A Common Denominator and Therapeutic Target for Neurodegenerative Disorders. J Exp Neurosci 2018; 12:1179069518772380. [PMID: 29760562 PMCID: PMC5946355 DOI: 10.1177/1179069518772380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
There is compelling evidence that a number of neurodegenerative diseases share common pathogenic mechanisms. Better understanding these mechanisms will allow us to develop new therapeutic strategies. This commentary follows up on our recent findings that tau pathology can be found in healthy fetal tissue transplanted into the brain of patients with either Huntington or Parkinson disease. We will examine how tau appears to be shared in a number of different conditions and how its expression relates to cognitive decline and disease progression. We will further review pathogenic mechanisms and especially the relevance of the possible prion-like behavior of tau. We will conclude by discussing how all this work opens up novel therapeutic approaches to treating the cognitive impairments related to neurodegenerative diseases using a common strategy.
Collapse
Affiliation(s)
- Alexander Maxan
- Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada.,Département de Psychiatrie et de Neurosciences, Université Laval, Québec, QC, Canada
| |
Collapse
|
155
|
Han Y, Cho U, Kim S, Park IS, Cho JH, Dhanasekaran DN, Song YS. Tumour microenvironment on mitochondrial dynamics and chemoresistance in cancer. Free Radic Res 2018; 52:1271-1287. [PMID: 29607684 DOI: 10.1080/10715762.2018.1459594] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mitochondria, evolutionally acquired symbionts of eukaryotic cells, are essential cytoplasmic organelles. They are structurally dynamic organelles that continually go through fission and fusion processes in response to various stimuli. Tumour tissue is composed of not just cancer cells but also various cell types like fibroblasts, mesenchymal stem and immune cells. Mitochondrial dynamics of cancer cells has been shown to be significantly affected by features of tumour microenvironment such as hypoxia, inflammation and energy deprivation. The interactions of cancer cells with tumour microenvironment like hypoxia give rise to the inter- and intratumoural heterogeneity, causing chemoresistance. In this review, we will focus on the chemoresistance by tumoural heterogeneity in relation to mitochondrial dynamics of cancer cells. Recent findings in molecular mechanisms involved in the control of mitochondrial dynamics as well as the impact of mitochondrial dynamics on drug sensitivity in cancer are highlighted in the current review.
Collapse
Affiliation(s)
- Youngjin Han
- a Biomodulation, Department of Agricultural Biotechnology , Seoul National University , Seoul , Republic of Korea.,b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Untack Cho
- b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea.,c Interdisciplinary Program in Cancer Biology , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Soochi Kim
- b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea.,d Seoul National University Hospital Biomedical Research Institute , Seoul , Republic of Korea
| | - In Sil Park
- b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea.,e Department of Agricultural Biotechnology , Seoul National University , Seoul , Republic of Korea
| | - Jae Hyun Cho
- f Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Danny N Dhanasekaran
- g Stephenson Cancer Center , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Yong Sang Song
- a Biomodulation, Department of Agricultural Biotechnology , Seoul National University , Seoul , Republic of Korea.,b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea.,c Interdisciplinary Program in Cancer Biology , Seoul National University College of Medicine , Seoul , Republic of Korea.,f Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| |
Collapse
|
156
|
Paliwal S, Chaudhuri R, Agrawal A, Mohanty S. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J Biomed Sci 2018; 25:31. [PMID: 29602309 PMCID: PMC5877369 DOI: 10.1186/s12929-018-0429-1] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/14/2018] [Indexed: 12/27/2022] Open
Abstract
The past decade has witnessed an upsurge in studies demonstrating mitochondrial transfer as one of the emerging mechanisms through which mesenchymal stem cells (MSCs) can regenerate and repair damaged cells or tissues. It has been found to play a critical role in healing several diseases related to brain injury, cardiac myopathies, muscle sepsis, lung disorders and acute respiratory disorders. Several studies have shown that various mechanisms are involved in mitochondrial transfer that includes tunnel tube formation, micro vesicle formation, gap junctions, cell fusion and others modes of transfer. Few studies have investigated the mechanisms that contribute to mitochondrial transfer, primarily comprising of signaling pathways involved in tunnel tube formation that facilitates tunnel tube formation for movement of mitochondria from one cell to another. Various stress signals such as release of damaged mitochondria, mtDNA and mitochondrial products along with elevated reactive oxygen species levels trigger the transfer of mitochondria from MSCs to recipient cells. However, extensive cell signaling pathways that lead to mitochondrial transfer from healthy cells are still under investigation and the changes that contribute to restoration of mitochondrial bioenergetics in recipient cells remain largely elusive. In this review, we have discussed the phenomenon of mitochondrial transfer from MSCs to neighboring stressed cells, and how this aids in cellular repair and regeneration of different organs such as lung, heart, eye, brain and kidney. The potential scope of mitochondrial transfer in providing novel therapeutic strategies for treatment of various pathophysiological conditions has also been discussed.
Collapse
Affiliation(s)
- Swati Paliwal
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rituparna Chaudhuri
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anurag Agrawal
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung disease, CSIR-Institute of Genomics and Integrative Biology, Delhi, 110007, India.
| | - Sujata Mohanty
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
157
|
Omsland M, Bruserud Ø, Gjertsen BT, Andresen V. Tunneling nanotube (TNT) formation is downregulated by cytarabine and NF-κB inhibition in acute myeloid leukemia (AML). Oncotarget 2018; 8:7946-7963. [PMID: 27974700 PMCID: PMC5352373 DOI: 10.18632/oncotarget.13853] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 11/21/2016] [Indexed: 01/11/2023] Open
Abstract
Acute myeloid leukemia (AML) is a bone marrow derived blood cancer where intercellular communication in the leukemic bone marrow participates in disease development, progression and chemoresistance. Tunneling nanotubes (TNTs) are intercellular communication structures involved in transport of cellular contents and pathogens, also demonstrated to play a role in both cell death modulation and chemoresistance. Here we investigated the presence of TNTs by live fluorescent microscopy and identified TNT formation between primary AML cells and in AML cell lines. We found that NF-κB activity was involved in TNT regulation and formation. Cytarabine downregulated TNTs and inhibited NF-κB alone and in combination with daunorubicin, providing additional support for involvement of the NF-κB pathway in TNT formation. Interestingly, daunorubicin was found to localize to lysosomes in TNTs connecting AML cells indicating a novel function of TNTs as drug transporting devices. We conclude that TNT communication could reflect important biological features of AML that may be explored in future therapy development.
Collapse
Affiliation(s)
- Maria Omsland
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, Precision Oncology Research Group, University of Bergen, Bergen, Norway
| | - Øystein Bruserud
- Leukaemia Research Group, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Internal Medicine, Haematology Section, Haukeland University Hospital, Bergen, Norway
| | - Bjørn T Gjertsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, Precision Oncology Research Group, University of Bergen, Bergen, Norway.,Department of Internal Medicine, Haematology Section, Haukeland University Hospital, Bergen, Norway
| | - Vibeke Andresen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, Precision Oncology Research Group, University of Bergen, Bergen, Norway
| |
Collapse
|
158
|
D’Aloia A, Berruti G, Costa B, Schiller C, Ambrosini R, Pastori V, Martegani E, Ceriani M. RalGPS2 is involved in tunneling nanotubes formation in 5637 bladder cancer cells. Exp Cell Res 2018; 362:349-361. [DOI: 10.1016/j.yexcr.2017.11.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 11/25/2022]
|
159
|
Tunneling nanotubes (TNT) mediate long-range gap junctional communication: Implications for HIV cell to cell spread. Sci Rep 2017; 7:16660. [PMID: 29192225 PMCID: PMC5709493 DOI: 10.1038/s41598-017-16600-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/15/2017] [Indexed: 12/24/2022] Open
Abstract
Cell-to-cell communication is essen for the development of multicellular systems and is coordinated by soluble factors, exosomes, gap junction (GJ) channels, and the recently described tunneling nanotubes (TNTs). We and others have demonstrated that TNT-like structures are mostly present during pathogenic conditions, including HIV infection. However, the nature, function, and communication properties of TNTs are still poorly understood. In this manuscript, we demonstrate that TNTs induced by HIV infection have functional GJs at the ends of their membrane extensions and that TNTs mediate long-range GJ communication during HIV infection. Blocking or reducing GJ communication during HIV infection resulted in aberrant TNT cell-to-cell contact, compromising HIV spread and replication. Thus, TNTs and associated GJs are required for the efficient cell-to-cell communication and viral spread. Our data indicate that targeting TNTs/GJs may provide new therapeutic opportunities for the treatment of HIV.
Collapse
|
160
|
A function-blocking CD47 antibody modulates extracellular vesicle-mediated intercellular signaling between breast carcinoma cells and endothelial cells. J Cell Commun Signal 2017; 12:157-170. [PMID: 29188480 DOI: 10.1007/s12079-017-0428-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
Tumor cells release extracellular vesicles (EVs) into the tumor microenvironment that may facilitate malignant progression and metastasis. Breast carcinoma EVs express high levels of the thrombospondin-1 and signal regulatory protein-α receptor CD47, which is the target of several experimental therapeutics currently in clinical trials. We analyzed changes in gene expression and function in human umbilical vein endothelial cells (HUVEC) induced by treatment with EVs derived from breast carcinoma cells and the effects of the function-blocking CD47 antibody B6H12 on the resulting intercellular communication. CD47+ EVs exhibited greater uptake by HUVEC compared to CD47- EVs, but the CD47 antibody did not inhibit their uptake. Global and targeted analyses of transcripts demonstrated that treatment of HUVEC with EVs derived from MDA-MB-231 breast carcinomas cells altered pathways associated with tumor necrosis factor-α signaling, angiogenesis, lymphangiogenesis, endothelial-mesenchymal transition, and extracellular matrix. EVs from triple-negative MDA-MB-231 cells were more active than EVs from less metastatic breast carcinoma cell lines. Treatment with MDA-MB-231 EVs down-regulated VEGFR2 mRNA expression and tyrosine phosphorylation while enhancing phosphorylation of the tyrosine phosphatase SHP2. VEGFR2 expression and phosphorylation in HUVEC was further inhibited by the CD47 antibody. Consistent with the observed changes in endothelial-mesenchymal transition genes and SHP2, treatment with MDA-MB-231-derived EVs decreased Zeb1 protein levels in HUVEC, whereas the CD47 antibody increased Zeb1 levels. The induction of E-selectin and other known targets of tumor necrosis factor-α signaling by EVs was also enhanced by the CD47 antibody, and E-selectin was the most up-regulated transcript following CD47 antibody treatment alone. These studies reveal several mechanisms by which therapeutics targeting CD47 could modulate tumor growth by altering the cross talk between cancer-derived EVs and nonmalignant cells in the tumor stroma.
Collapse
|
161
|
Masnata M, Cicchetti F. The Evidence for the Spread and Seeding Capacities of the Mutant Huntingtin Protein in in Vitro Systems and Their Therapeutic Implications. Front Neurosci 2017; 11:647. [PMID: 29234268 PMCID: PMC5712341 DOI: 10.3389/fnins.2017.00647] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/07/2017] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative disorders are not only characterized by specific patterns of cell loss but the presence and accumulation of various pathological proteins—both of which correlate with disease evolution. There is now mounting evidence to suggest that these pathological proteins present with toxic, at times prion-like, properties and can therefore seed pathology in neighboring as well remotely connected healthy neurons as they spread across the brain. What is less clear, at this stage, is how much this actually contributes to, and drives, the core pathogenic events. In this review, we present a comprehensive, up-to-date summary of the reported in vitro studies that support the spreading and seeding capacities of pathological proteins, with an emphasis on mutant huntingtin protein in the context of Huntington's disease, although in vivo work remains to be performed to validate this theory in this particular disease. We have further reviewed these findings in light of their potential implications for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Maria Masnata
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada
| | - Francesca Cicchetti
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Quebec, QC, Canada
| |
Collapse
|
162
|
Dahmke IN, Verch A, Hermannsdörfer J, Peckys DB, Weatherup RS, Hofmann S, de Jonge N. Graphene Liquid Enclosure for Single-Molecule Analysis of Membrane Proteins in Whole Cells Using Electron Microscopy. ACS NANO 2017; 11:11108-11117. [PMID: 29023096 DOI: 10.1021/acsnano.7b05258] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Membrane proteins govern many important functions in cells via dynamic oligomerization into active complexes. However, analytical methods to study their distribution and functional state in relation to the cellular structure are currently limited. Here, we introduce a technique for studying single-membrane proteins within their native context of the intact plasma membrane. SKBR3 breast cancer cells were grown on silicon microchips with thin silicon nitride windows. The cells were fixed, and the epidermal growth factor receptor ErbB2 was specifically labeled with quantum dot (QD) nanoparticles. For correlative fluorescence- and liquid-phase electron microscopy, we enclosed the liquid samples by chemical vapor deposited (CVD) graphene films. Depending on the local cell thickness, QD labels were imaged with a spatial resolution of 2 nm at a low electron dose. The distribution and stoichiometric assembly of ErbB2 receptors were determined at several different cellular locations, including tunneling nanotubes, where we found higher levels of homodimerization at the connecting sites. This experimental approach is applicable to a wide range of cell lines and membrane proteins and particularly suitable for studies involving both inter- and intracellular heterogeneity in protein distribution and expression.
Collapse
Affiliation(s)
- Indra N Dahmke
- INM - Leibniz Institute for New Materials , D-66123 Saarbrücken, Germany
| | - Andreas Verch
- INM - Leibniz Institute for New Materials , D-66123 Saarbrücken, Germany
| | | | - Diana B Peckys
- Department of Biophysics, Saarland University , D-66421 Homburg, Germany
| | - Robert S Weatherup
- Engineering Department, University of Cambridge , Cambridge CB3 0FA, United Kingdom
| | - Stephan Hofmann
- Engineering Department, University of Cambridge , Cambridge CB3 0FA, United Kingdom
| | - Niels de Jonge
- INM - Leibniz Institute for New Materials , D-66123 Saarbrücken, Germany
- Department of Physics, Saarland University , D-66123 Saarbrücken, Germany
| |
Collapse
|
163
|
Abounit S, Wu JW, Duff K, Victoria GS, Zurzolo C. Tunneling nanotubes: A possible highway in the spreading of tau and other prion-like proteins in neurodegenerative diseases. Prion 2017; 10:344-351. [PMID: 27715442 DOI: 10.1080/19336896.2016.1223003] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The mechanisms of intercellular spreading of amyloidogenic proteins involved in neurodegenerative diseases have yet to be fully elucidated. While secretion has been implicated in the transfer of many proteins, including prions and α-synuclein, tunneling nanotubes (TNTs) have also been demonstrated for prions and mutant Huntingtin. Here, we provide further evidence that Tau aggregates, which have been demonstrated to predominantly be transferred via secretion, can also be found in TNTs. Additionally, cells that have taken up Tau have increased TNT formation. Coupled with previous evidence that other amyloidogenic aggregates also induce TNT formation we propose that misfolded protein aggregates can, through a common mechanism, promote the formation of TNTs and thereby their own intercellular transfer, contributing to the propagation of pathology.
Collapse
Affiliation(s)
- Saida Abounit
- a Unité Trafic Membranaire et Pathogenèse , Institut Pasteur , Paris CEDEX 15 , France
| | - Jessica W Wu
- b Taub Institute, Columbia University Medical Center , New York , NY , USA
| | - Karen Duff
- b Taub Institute, Columbia University Medical Center , New York , NY , USA
| | | | - Chiara Zurzolo
- a Unité Trafic Membranaire et Pathogenèse , Institut Pasteur , Paris CEDEX 15 , France
| |
Collapse
|
164
|
Griessinger E, Moschoi R, Biondani G, Peyron JF. Mitochondrial Transfer in the Leukemia Microenvironment. Trends Cancer 2017; 3:828-839. [PMID: 29198439 DOI: 10.1016/j.trecan.2017.10.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/18/2022]
Abstract
The bone marrow microenvironment (BMME) is a complex ecosystem that instructs and protects hematopoietic stem cells (HSCs) and their malignant counterparts, the leukemia-initiating cells (LICs). Within the physical and functional crosstalk that takes place between HSCs, LICs, and the BMME, the transfer of organelles and of mitochondria in particular is an important new intercellular communication mode in addition to adhesion molecules, tunneling nanotubes (TNTs), and the paracrine secretion of cytokines, (onco)metabolites, and extracellular vesicles (EVs). In this review we discuss the functional roles of mitochondrial transfer between BMME and leukemic cells, and give insights into this new mechanism of drug resistance whose understanding will open the way to innovative anticancer adjuvant treatments.
Collapse
Affiliation(s)
- Emmanuel Griessinger
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France.
| | - Ruxanda Moschoi
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France
| | - Giulia Biondani
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France
| | - Jean-François Peyron
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France.
| |
Collapse
|
165
|
α-Synuclein transfer between neurons and astrocytes indicates that astrocytes play a role in degradation rather than in spreading. Acta Neuropathol 2017; 134:789-808. [PMID: 28725967 DOI: 10.1007/s00401-017-1746-2] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/27/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
Recent evidence suggests that disease progression in Parkinson's disease (PD) could occur by the spreading of α-synuclein (α-syn) aggregates between neurons. Here we studied the role of astrocytes in the intercellular transfer and fate of α-syn fibrils, using in vitro and ex vivo models. α-Syn fibrils can be transferred to neighboring cells; however, the transfer efficiency changes depending on the cell types. We found that α-syn is efficiently transferred from astrocytes to astrocytes and from neurons to astrocytes, but less efficiently from astrocytes to neurons. Interestingly, α-syn puncta are mainly found inside the lysosomal compartments of the recipient cells. However, differently from neurons, astrocytes are able to efficiently degrade fibrillar α-syn, suggesting an active role for these cells in clearing α-syn deposits. Astrocytes co-cultured with organotypic brain slices are able to take up α-syn fibrils from the slices. Altogether our data support a role for astrocytes in trapping and clearing α-syn pathological deposits in PD.
Collapse
|
166
|
Keller KE, Bradley JM, Sun YY, Yang YF, Acott TS. Tunneling Nanotubes are Novel Cellular Structures That Communicate Signals Between Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2017; 58:5298-5307. [PMID: 29049733 PMCID: PMC5656416 DOI: 10.1167/iovs.17-22732] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Purpose The actin cytoskeleton of trabecular meshwork (TM) cells plays a role in regulating aqueous humor outflow. Many studies have investigated stress fibers, but F-actin also assembles into other supramolecular structures including filopodia. Recently, specialized filopodia called tunneling nanotubes (TNTs) have been described, which communicate molecular signals and organelles directly between cells. Here, we investigate TNT formation by TM cells. Methods Human TM cells were labeled separately with the fluorescent dyes, DiO and DiD, or with mitochondrial dye. Fixed or live TM cells were imaged using confocal microscopy. Image analysis software was used to track fluorescent vesicles and count the number and length of filopodia. The number of fluorescently labeled vesicles transferred between cells was counted in response to specific inhibitors of the actin cytoskeleton. Human TM tissue was stained with phalloidin. Results Live-cell confocal imaging of cultured TM cells showed transfer of fluorescently labeled vesicles and mitochondria via TNTs. In TM tissue, a long (160 μm) actin-rich cell process bridged an intertrabecular space and did not adhere to the substratum. Treatment of TM cells with CK-666, an Arp2/3 inhibitor, significantly decreased the number and length of filopodia, decreased transfer of fluorescently labeled vesicles and induced thick stress fibers compared to vehicle control. Conversely, inhibiting stress fibers using Y27632 increased transfer of vesicles and induced long cell processes. Conclusions Identification of TNTs provides a means by which TM cells can directly communicate with each other over long distances. This may be particularly important to overcome limitations of diffusion-based signaling in the aqueous humor fluid environment.
Collapse
Affiliation(s)
- Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - John M Bradley
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ted S Acott
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
167
|
Intercellular mRNA trafficking via membrane nanotube-like extensions in mammalian cells. Proc Natl Acad Sci U S A 2017; 114:E9873-E9882. [PMID: 29078295 PMCID: PMC5699038 DOI: 10.1073/pnas.1706365114] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
mRNA molecules convey genetic information within cells, beginning from genes in the nucleus to ribosomes in the cell body, where they are translated into proteins. Here we show a mode of transferring genetic information from one cell to another. Contrary to previous publications suggesting that mRNAs transfer via extracellular vesicles, we provide visual and quantitative data showing that mRNAs transfer via membrane nanotubes and direct cell-to-cell contact. We predict that this process has a major role in regulating local cellular environments with respect to tissue development and maintenance and cellular responses to stress, interactions with parasites, tissue transplants, and the tumor microenvironment. RNAs have been shown to undergo transfer between mammalian cells, although the mechanism behind this phenomenon and its overall importance to cell physiology is not well understood. Numerous publications have suggested that RNAs (microRNAs and incomplete mRNAs) undergo transfer via extracellular vesicles (e.g., exosomes). However, in contrast to a diffusion-based transfer mechanism, we find that full-length mRNAs undergo direct cell–cell transfer via cytoplasmic extensions characteristic of membrane nanotubes (mNTs), which connect donor and acceptor cells. By employing a simple coculture experimental model and using single-molecule imaging, we provide quantitative data showing that mRNAs are transferred between cells in contact. Examples of mRNAs that undergo transfer include those encoding GFP, mouse β-actin, and human Cyclin D1, BRCA1, MT2A, and HER2. We show that intercellular mRNA transfer occurs in all coculture models tested (e.g., between primary cells, immortalized cells, and in cocultures of immortalized human and murine cells). Rapid mRNA transfer is dependent upon actin but is independent of de novo protein synthesis and is modulated by stress conditions and gene-expression levels. Hence, this work supports the hypothesis that full-length mRNAs undergo transfer between cells through a refined structural connection. Importantly, unlike the transfer of miRNA or RNA fragments, this process of communication transfers genetic information that could potentially alter the acceptor cell proteome. This phenomenon may prove important for the proper development and functioning of tissues as well as for host–parasite or symbiotic interactions.
Collapse
|
168
|
Ariazi J, Benowitz A, De Biasi V, Den Boer ML, Cherqui S, Cui H, Douillet N, Eugenin EA, Favre D, Goodman S, Gousset K, Hanein D, Israel DI, Kimura S, Kirkpatrick RB, Kuhn N, Jeong C, Lou E, Mailliard R, Maio S, Okafo G, Osswald M, Pasquier J, Polak R, Pradel G, de Rooij B, Schaeffer P, Skeberdis VA, Smith IF, Tanveer A, Volkmann N, Wu Z, Zurzolo C. Tunneling Nanotubes and Gap Junctions-Their Role in Long-Range Intercellular Communication during Development, Health, and Disease Conditions. Front Mol Neurosci 2017; 10:333. [PMID: 29089870 PMCID: PMC5651011 DOI: 10.3389/fnmol.2017.00333] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/02/2017] [Indexed: 12/15/2022] Open
Abstract
Cell-to-cell communication is essential for the organization, coordination, and development of cellular networks and multi-cellular systems. Intercellular communication is mediated by soluble factors (including growth factors, neurotransmitters, and cytokines/chemokines), gap junctions, exosomes and recently described tunneling nanotubes (TNTs). It is unknown whether a combination of these communication mechanisms such as TNTs and gap junctions may be important, but further research is required. TNTs are long cytoplasmic bridges that enable long-range, directed communication between connected cells. The proposed functions of TNTs are diverse and not well understood but have been shown to include the cell-to-cell transfer of vesicles, organelles, electrical stimuli and small molecules. However, the exact role of TNTs and gap junctions for intercellular communication and their impact on disease is still uncertain and thus, the subject of much debate. The combined data from numerous laboratories indicate that some TNT mediate a long-range gap junctional communication to coordinate metabolism and signaling, in relation to infectious, genetic, metabolic, cancer, and age-related diseases. This review aims to describe the current knowledge, challenges and future perspectives to characterize and explore this new intercellular communication system and to design TNT-based therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Monique L Den Boer
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Stephanie Cherqui
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Haifeng Cui
- GlaxoSmithKline, Collegeville, PA, United States
| | | | - Eliseo A Eugenin
- Public Health Research Institute (PHRI), Newark, NJ, United States.,Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, United States
| | - David Favre
- GlaxoSmithKline, Research Triangle Park, NC, United States
| | - Spencer Goodman
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Karine Gousset
- Department of Biology, College of Science and Math, California State University, Fresno, CA, United States
| | - Dorit Hanein
- Bioinformatics and System Biology Program, Sanford Burnham Prebys Medical Discovery, La Jolla, CA, United States
| | | | - Shunsuke Kimura
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Nastaran Kuhn
- Division of Cancer Biology, Physical Sciences-Oncology Network, Cancer Tissue Engineering Collaborative Research Program, Program Director, Structural Biology and Molecular Applications Branch, National Cancer Institute, Bethesda, MD, United States
| | - Claire Jeong
- GlaxoSmithKline, King of Prussia, PA, United States
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Robbie Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Stephen Maio
- GlaxoSmithKline, King of Prussia, PA, United States
| | | | - Matthias Osswald
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jennifer Pasquier
- Department of Genetic Medicine, Weill Cornell Medical College in Qatar, Qatar Foundation, Ar-Rayyan, Qatar
| | - Roel Polak
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, RWTH Aachen University, Aachen, Germany
| | - Bob de Rooij
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, Netherlands
| | | | - Vytenis A Skeberdis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ian F Smith
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Ahmad Tanveer
- Section of Intracellular Trafficking and Neurovirology, National Institute of Health, Bethesda, MD, United States
| | - Niels Volkmann
- Bioinformatics and System Biology Program, Sanford Burnham Prebys Medical Discovery, La Jolla, CA, United States
| | - Zhenhua Wu
- GlaxoSmithKline, Collegeville, PA, United States
| | - Chiara Zurzolo
- Unit of Membrane Trafficking and Pathogenesis, Department of Cell Biology and Infection, Pasteur Institute, Paris, France
| |
Collapse
|
169
|
Ribeiro-Rodrigues TM, Martins-Marques T, Morel S, Kwak BR, Girão H. Role of connexin 43 in different forms of intercellular communication - gap junctions, extracellular vesicles and tunnelling nanotubes. J Cell Sci 2017; 130:3619-3630. [PMID: 29025971 DOI: 10.1242/jcs.200667] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Communication is important to ensure the correct and efficient flow of information, which is required to sustain active social networks. A fine-tuned communication between cells is vital to maintain the homeostasis and function of multicellular or unicellular organisms in a community environment. Although there are different levels of complexity, intercellular communication, in prokaryotes to mammalians, can occur through secreted molecules (either soluble or encapsulated in vesicles), tubular structures connecting close cells or intercellular channels that link the cytoplasm of adjacent cells. In mammals, these different types of communication serve different purposes, may involve distinct factors and are mediated by extracellular vesicles, tunnelling nanotubes or gap junctions. Recent studies have shown that connexin 43 (Cx43, also known as GJA1), a transmembrane protein initially described as a gap junction protein, participates in all these forms of communication; this emphasizes the concept of adopting strategies to maximize the potential of available resources by reutilizing the same factor in different scenarios. In this Review, we provide an overview of the most recent advances regarding the role of Cx43 in intercellular communication mediated by extracellular vesicles, tunnelling nanotubes and gap junctions.
Collapse
Affiliation(s)
- Teresa M Ribeiro-Rodrigues
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta Comba, 3000-548 Coimbra, Portugal.,CNC.IBILI, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Tânia Martins-Marques
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta Comba, 3000-548 Coimbra, Portugal.,CNC.IBILI, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Sandrine Morel
- Dept. of Pathology and Immunology, and Dept. of Medical Specialties - Cardiology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Brenda R Kwak
- Dept. of Pathology and Immunology, and Dept. of Medical Specialties - Cardiology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Henrique Girão
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta Comba, 3000-548 Coimbra, Portugal .,CNC.IBILI, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
170
|
Winnard PT, Zhang C, Vesuna F, Kang JW, Garry J, Dasari RR, Barman I, Raman V. Organ-specific isogenic metastatic breast cancer cell lines exhibit distinct Raman spectral signatures and metabolomes. Oncotarget 2017; 8:20266-20287. [PMID: 28145887 PMCID: PMC5386761 DOI: 10.18632/oncotarget.14865] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
Molecular characterization of organ-specific metastatic lesions, which distinguish them from the primary tumor, will provide a better understanding of tissue specific adaptations that regulate metastatic progression. Using an orthotopic xenograft model, we have isolated isogenic metastatic human breast cancer cell lines directly from organ explants that are phenotypically distinct from the primary tumor cell line. Label-free Raman spectroscopy was used and informative spectral bands were ascertained as differentiators of organ-specific metastases as opposed to the presence of a single universal marker. Decision algorithms derived from the Raman spectra unambiguously identified these isogenic cell lines as unique biological entities – a finding reinforced through metabolomic analyses that indicated tissue of origin metabolite distinctions between the cell lines. Notably, complementarity of the metabolomics and Raman datasets was found. Our findings provide evidence that metastatic spread generates tissue-specific adaptations at the molecular level within cancer cells, which can be differentiated with Raman spectroscopy.
Collapse
Affiliation(s)
- Paul T Winnard
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chi Zhang
- The Johns Hopkins University, Department of Mechanical Engineering, Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Farhad Vesuna
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeon Woong Kang
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonah Garry
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ramachandra Rao Dasari
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ishan Barman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Johns Hopkins University, Department of Mechanical Engineering, Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Venu Raman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
171
|
Pseudorabies Virus US3-Induced Tunneling Nanotubes Contain Stabilized Microtubules, Interact with Neighboring Cells via Cadherins, and Allow Intercellular Molecular Communication. J Virol 2017; 91:JVI.00749-17. [PMID: 28747498 DOI: 10.1128/jvi.00749-17] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/16/2017] [Indexed: 01/01/2023] Open
Abstract
Tunneling nanotubes (TNTs) are long bridge-like structures that connect eukaryotic cells and mediate intercellular communication. We found earlier that the conserved alphaherpesvirus US3 protein kinase induces long cell projections that contact distant cells and promote intercellular virus spread. In this report, we show that the US3-induced cell projections constitute TNTs. In addition, we report that US3-induced TNTs mediate intercellular transport of information (e.g., green fluorescent protein [GFP]) in the absence of other viral proteins. US3-induced TNTs are remarkably stable compared to most TNTs described in the literature. In line with this, US3-induced TNTs were found to contain stabilized (acetylated and detyrosinated) microtubules. Transmission electron microscopy showed that virus particles are individually transported in membrane-bound vesicles in US3-induced TNTs and are released along the TNT and at the contact area between a TNT and the adjacent cell. Contact between US3-induced TNTs and acceptor cells is very stable, which correlated with a marked enrichment in adherens junction components beta-catenin and E-cadherin at the contact area. These data provide new structural insights into US3-induced TNTs and how they may contribute to intercellular communication and alphaherpesvirus spread.IMPORTANCE Tunneling nanotubes (TNT) represent an important and yet still poorly understood mode of long-distance intercellular communication. We and others reported earlier that the conserved alphaherpesvirus US3 protein kinase induces long cellular protrusions in infected and transfected cells. Here, we show that US3-induced cell projections constitute TNTs, based on structural properties and transport of biomolecules. In addition, we report on different particular characteristics of US3-induced TNTs that help to explain their remarkable stability compared to physiological TNTs. In addition, transmission electron microscopy assays indicate that, in infected cells, virions travel in the US3-induced TNTs in membranous transport vesicles and leave the TNT via exocytosis. These data generate new fundamental insights into the biology of (US3-induced) TNTs and into how they may contribute to intercellular virus spread and communication.
Collapse
|
172
|
Parker I, Evans KT, Ellefsen K, Lawson DA, Smith IF. Lattice light sheet imaging of membrane nanotubes between human breast cancer cells in culture and in brain metastases. Sci Rep 2017; 7:11029. [PMID: 28887508 PMCID: PMC5591308 DOI: 10.1038/s41598-017-11223-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/17/2017] [Indexed: 01/15/2023] Open
Abstract
Membrane nanotubes are cytosolic protrusions with diameters <1 µm that extend between cells separated by tens of µm. They mediate several forms of intercellular communication and are upregulated in diverse diseases. Difficulties in visualizing and studying nanotubes within intact tissues have, however, prompted skepticism regarding their in vivo relevance, and most studies have been confined to cell culture systems. Here, we introduce lattice-light sheet imaging of MDA-MB-231 human breast cancer cells genetically engineered to brightly express membrane-targeted GFP as a promising approach to visualize membrane nanotubes in vitro and in situ. We demonstrate that cultured cells form multiple nanotubes that mediate intercellular communication of Ca2+ signals and actively traffic GFP-tagged membrane vesicles along their length. Furthermore, we directly visualize nanotubes in situ, interconnecting breast cancer cells in live acute brain slices from an experimental mouse model of breast cancer brain metastasis. This amenable experimental system should facilitate the transition of the study of intercellular communication by membrane nanotubes from cell culture to the whole animal.
Collapse
Affiliation(s)
- Ian Parker
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Katrina T Evans
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Kyle Ellefsen
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Devon A Lawson
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Ian F Smith
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
| |
Collapse
|
173
|
Abstract
PURPOSE OF THE REVIEW The connections between allergy, asthma and metabolic syndrome are becoming increasingly clear. Recent research suggests a unifying mitochondrial link between the diverse phenotypes of these interlinked morbidities. The scope of this review is to highlight cellular mechanisms, epidemiology and environmental allergens influencing mitochondrial function and its importance in allergy and asthma. We briefly also consider the potential of mitochondria-targeted therapies in prevention and cure. RECENT FINDINGS Recent research has shown allergy, asthma and metabolic syndrome to be linked to mitochondrial dysfunction. Environmental pollutants and allergens are observed to cause mitochondrial dysfunction, primarily by inducing oxidative stress and ROS production. Malfunctioning mitochondria change the bioenergetics of the cell and its metabolic profile to favour systemic inflammation, which drives all three types of morbidities. Given the existing experimental evidence, approaches targeting mitochondria (e.g. antioxidant therapy and mitochondrial replacement) are being conducted in relevant disease models-with some progressing towards clinical trials, making mitochondrial function the focus of translational therapy research in asthma, allergy and linked metabolic syndrome.
Collapse
Affiliation(s)
- Divyaanka Iyer
- CSIR Institute of Genomics and Integrative Biology, Delhi University campus, Mall Road, Delhi, 110007, India
| | - Navya Mishra
- Indian Institute of Public Health, Gurugram, India.,Chest Research Foundation, Pune, India.,Academy of Scientific and Innovative Research, New Delhi, India
| | - Anurag Agrawal
- CSIR Institute of Genomics and Integrative Biology, Delhi University campus, Mall Road, Delhi, 110007, India. .,Academy of Scientific and Innovative Research, New Delhi, India.
| |
Collapse
|
174
|
Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget 2017; 8:15539-15552. [PMID: 28107184 PMCID: PMC5362504 DOI: 10.18632/oncotarget.14695] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/27/2016] [Indexed: 01/15/2023] Open
Abstract
Intercellular transfer of organelles via tunneling nanotubes (TNTs) is a novel means of cell-to-cell communication. Here we demonstrate the existence of TNTs between co-cultured RT4 and T24 bladder cancer cells using light microscopy, fluorescence imaging, and scanning electron microscopy (SEM). Spontaneous unidirectional transfer of mitochondria from T24 to RT4 cells was detected using fluorescence imaging and flow cytometry. The distribution of mitochondria migrated from T24 cells was in good agreement with the original mitochondria in RT4 cells, which may imply mitochondrial fusion. We detected cytoskeleton reconstruction in RT4-Mito-T24 cells by observing F-actin redistribution. Akt, mTOR, and their downstream mediators were activated and increased. The resultant increase in the invasiveness of bladder cancer cells was detected in vitro and in vivo. These data indicate that TNTs promote intercellular mitochondrial transfer between heterogeneous cells, followed by an increase in the invasiveness of bladder cancer cells.
Collapse
|
175
|
Hanna SJ, McCoy-Simandle K, Miskolci V, Guo P, Cammer M, Hodgson L, Cox D. The Role of Rho-GTPases and actin polymerization during Macrophage Tunneling Nanotube Biogenesis. Sci Rep 2017; 7:8547. [PMID: 28819224 PMCID: PMC5561213 DOI: 10.1038/s41598-017-08950-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 07/17/2017] [Indexed: 01/06/2023] Open
Abstract
Macrophage interactions with other cells, either locally or at distances, are imperative in both normal and pathological conditions. While soluble means of communication can transmit signals between different cells, it does not account for all long distance macrophage interactions. Recently described tunneling nanotubes (TNTs) are membranous channels that connect cells together and allow for transfer of signals, vesicles, and organelles. However, very little is known about the mechanism by which these structures are formed. Here we investigated the signaling pathways involved in TNT formation by macrophages using multiple imaging techniques including super-resolution microscopy (3D-SIM) and live-cell imaging including the use of FRET-based Rho GTPase biosensors. We found that formation of TNTs required the activity and differential localization of Cdc42 and Rac1. The downstream Rho GTPase effectors mediating actin polymerization through Arp2/3 nucleation, Wiskott-Aldrich syndrome protein (WASP) and WASP family verprolin-homologous 2 (WAVE2) proteins are also important, and both pathways act together during TNT biogenesis. Finally, TNT function as measured by transfer of cellular material between cells was reduced following depletion of a single factor demonstrating the importance of these factors in TNTs. Given that the characterization of TNT formation is still unclear in the field; this study provides new insights and would enhance the understanding of TNT formation towards investigating new markers.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Veronika Miskolci
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Gruss MRRC 306, Bronx, NY, 10461, USA
| | - Peng Guo
- Department of Anatomy and Structural 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, Bronx, NY, USA.,Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Cammer
- Microscopy Core, DART, NYU Langone Medical Center, Bronx, NY, 10016, USA
| | - Louis Hodgson
- Department of Anatomy and Structural 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, Bronx, NY, 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, Bronx, NY, USA.
| |
Collapse
|
176
|
Mutant Huntingtin Is Secreted via a Late Endosomal/Lysosomal Unconventional Secretory Pathway. J Neurosci 2017; 37:9000-9012. [PMID: 28821645 DOI: 10.1523/jneurosci.0118-17.2017] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 06/26/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder caused by the expansion of a CAG triplet in the gene encoding for huntingtin (Htt). The resulting mutant protein (mHtt) with extended polyglutamine (polyQ) sequence at the N terminus leads to neuronal degeneration both in a cell-autonomous and a non-cell-autonomous manner. Recent studies identified mHtt in the extracellular environment and suggested that its spreading contributes to toxicity, but the mechanism of mHtt release from the cell of origin remains unknown. In this study, we performed a comprehensive, unbiased analysis of secretory pathways and identified an unconventional lysosomal pathway as an important mechanism for mHtt secretion in mouse neuroblastoma and striatal cell lines, as well as in primary neurons. mHtt secretion was dependent on synaptotagmin 7, a regulator of lysosomal secretion, and inhibited by chemical ablation of late endosomes/lysosomes, suggesting a lysosomal secretory pattern. mHtt was targeted preferentially to the late endosomes/lysosomes compared with wild-type Htt. Importantly, we found that late endosomal/lysosomal targeting and secretion of mHtt could be inhibited efficiently by the phosphatidylinositol 3-kinase and neutral sphingomyelinase chemical inhibitors, Ly294002 and GW4869, respectively. Together, our data suggest a lysosomal mechanism of mHtt secretion and offer potential strategies for pharmacological modulation of its neuronal secretion.SIGNIFICANCE STATEMENT This is the first study examining the mechanism of mutant huntingtin (mHTT) secretion in an unbiased manner. We found that the protein is secreted via a late endosomal/lysosomal unconventional secretory pathway. Moreover, mHtt secretion can be reduced significantly by phosphatidylinositol 3-kinase and neutral sphingomyelinase inhibitors. Understanding and manipulating the secretion of mHtt is important because of its potentially harmful propagation in the brain.
Collapse
|
177
|
Victoria GS, Zurzolo C. The spread of prion-like proteins by lysosomes and tunneling nanotubes: Implications for neurodegenerative diseases. J Cell Biol 2017; 216:2633-2644. [PMID: 28724527 PMCID: PMC5584166 DOI: 10.1083/jcb.201701047] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/03/2017] [Accepted: 07/05/2017] [Indexed: 11/22/2022] Open
Abstract
Victoria and Zurzolo discuss current evidence for the emerging role of lysosomal damage and tunneling nanotubes in the intercellular propagation of prion and prion-like proteins in neurodegenerative disease. Progression of pathology in neurodegenerative diseases is hypothesized to be a non–cell-autonomous process that may be mediated by the productive spreading of prion-like protein aggregates from a “donor cell” that is the source of misfolded aggregates to an “acceptor cell” in which misfolding is propagated by conversion of the normal protein. Although the proteins involved in the various diseases are unrelated, common pathways appear to be used for their intercellular propagation and spreading. Here, we summarize recent evidence of the molecular mechanisms relevant for the intercellular trafficking of protein aggregates involved in prion, Alzheimer’s, Huntington’s, and Parkinson’s diseases. We focus in particular on the common roles that lysosomes and tunneling nanotubes play in the formation and spreading of prion-like assemblies.
Collapse
Affiliation(s)
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur, Paris, France
| |
Collapse
|
178
|
Modulation of oxidative phosphorylation and redox homeostasis in mitochondrial NDUFS4 deficiency via mesenchymal stem cells. Stem Cell Res Ther 2017. [PMID: 28646906 PMCID: PMC5482938 DOI: 10.1186/s13287-017-0601-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Disorders of the oxidative phosphorylation (OXPHOS) system represent a large group among the inborn errors of metabolism. The most frequently observed biochemical defect is isolated deficiency of mitochondrial complex I (CI). No effective treatment strategies for CI deficiency are so far available. The purpose of this study was to investigate whether and how mesenchymal stem cells (MSCs) are able to modulate metabolic function in fibroblast cell models of CI deficiency. Methods We used human and murine fibroblasts with a defect in the nuclear DNA encoded NDUFS4 subunit of CI. Fibroblasts were co-cultured with MSCs under different stress conditions and intercellular mitochondrial transfer was assessed by flow cytometry and fluorescence microscopy. Reactive oxygen species (ROS) levels were measured using MitoSOX-Red. Protein levels of CI were analysed by blue native polyacrylamide gel electrophoresis (BN-PAGE). Results Direct cellular interactions and mitochondrial transfer between MSCs and human as well as mouse fibroblast cell lines were demonstrated. Mitochondrial transfer was visible in 13.2% and 6% of fibroblasts (e.g. fibroblasts containing MSC mitochondria) for human and mouse cell lines, respectively. The transfer rate could be further stimulated via treatment of cells with TNF-α. MSCs effectively lowered cellular ROS production in NDUFS4-deficient fibroblast cell lines (either directly via co-culture or indirectly via incubation of cell lines with cell-free MSC supernatant). However, CI protein expression and activity were not rescued by MSC treatment. Conclusion This study demonstrates the interplay between MSCs and fibroblast cell models of isolated CI deficiency including transfer of mitochondria as well as modulation of cellular ROS levels. Further exploration of these cellular interactions might help to develop MSC-based treatment strategies for human CI deficiency. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0601-7) contains supplementary material, which is available to authorized users.
Collapse
|
179
|
Tóth EA, Oszvald Á, Péter M, Balogh G, Osteikoetxea-Molnár A, Bozó T, Szabó-Meleg E, Nyitrai M, Derényi I, Kellermayer M, Yamaji T, Hanada K, Vígh L, Matkó J. Nanotubes connecting B lymphocytes: High impact of differentiation-dependent lipid composition on their growth and mechanics. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28645851 DOI: 10.1016/j.bbalip.2017.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanotubes (NTs) are thin, long membranous structures forming novel, yet poorly known communication pathways between various cell types. Key mechanisms controlling their growth still remained poorly understood. Since NT-forming capacity of immature and mature B cells was found largely different, we investigated how lipid composition and molecular order of the membrane affect NT-formation. Screening B cell lines with various differentiation stages revealed that NT-growth linearly correlates with membrane ganglioside levels, while it shows maximum as a function of cholesterol level. NT-growth of B lymphocytes is promoted by raftophilic phosphatidylcholine and sphingomyelin species, various glycosphingolipids, and docosahexaenoic acid-containing inner leaflet lipids, through supporting membrane curvature, as demonstrated by comparative lipidomic analysis of mature versus immature B cell membranes. Targeted modification of membrane cholesterol and sphingolipid levels altered NT-forming capacity confirming these findings, and also highlighted that the actual lipid raft number may control NT-growth via defining the number of membrane-F-actin coupling sites. Atomic force microscopic mechano-manipulation experiments further proved that mechanical properties (elasticity or bending stiffness) of B cell NTs also depend on the actual membrane lipid composition. Data presented here highlight importance of the lipid side in controlling intercellular, nanotubular, regulatory communications in the immune system.
Collapse
Affiliation(s)
- Eszter A Tóth
- Department of Immunology, Eötvös Lorand University, Budapest, Hungary
| | - Ádám Oszvald
- Department of Immunology, Eötvös Lorand University, Budapest, Hungary
| | - Mária Péter
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary.
| | | | - Tamás Bozó
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical Faculty, University of Pécs, Pécs, Hungary; Department of Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Miklós Nyitrai
- Department of Biophysics, Medical Faculty, University of Pécs, Pécs, Hungary; Department of Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Imre Derényi
- Department of Biological Physics, Eötvös Lorand University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary; MTA-SE Molecular Biophysics Research Group, Semmelweis University, Budapest, Hungary
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shunjuku-ku, Tokyo, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shunjuku-ku, Tokyo, Japan
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - János Matkó
- Department of Immunology, Eötvös Lorand University, Budapest, Hungary.
| |
Collapse
|
180
|
Tweety-Homolog 1 Drives Brain Colonization of Gliomas. J Neurosci 2017; 37:6837-6850. [PMID: 28607172 DOI: 10.1523/jneurosci.3532-16.2017] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/16/2017] [Accepted: 04/19/2017] [Indexed: 01/25/2023] Open
Abstract
Early and progressive colonization of the healthy brain is one hallmark of diffuse gliomas, including glioblastomas. We recently discovered ultralong (>10 to hundreds of microns) membrane protrusions [tumor microtubes (TMs)] extended by glioma cells. TMs have been associated with the capacity of glioma cells to effectively invade the brain and proliferate. Moreover, TMs are also used by some tumor cells to interconnect to one large, resistant multicellular network. Here, we performed a correlative gene-expression microarray and in vivo imaging analysis, and identified novel molecular candidates for TM formation and function. Interestingly, these genes were previously linked to normal CNS development. One of the genes scoring highest in tests related to the outgrowth of TMs was tweety-homolog 1 (TTYH1), which was highly expressed in a fraction of TMs in mice and patients. Ttyh1 was confirmed to be a potent regulator of normal TM morphology and of TM-mediated tumor-cell invasion and proliferation. Glioma cells with one or two TMs were mainly responsible for effective brain colonization, and Ttyh1 downregulation particularly affected this cellular subtype, resulting in reduced tumor progression and prolonged survival of mice. The remaining Ttyh1-deficient tumor cells, however, had more interconnecting TMs, which were associated with increased radioresistance in those small tumors. These findings imply a cellular and molecular heterogeneity in gliomas regarding formation and function of distinct TM subtypes, with multiple parallels to neuronal development, and suggest that Ttyh1 might be a promising target to specifically reduce TM-associated brain colonization by glioma cells in patients.SIGNIFICANCE STATEMENT In this report, we identify tweety-homolog 1 (Ttyh1), a membrane protein linked to neuronal development, as a potent driver of tumor microtube (TM)-mediated brain colonization by glioma cells. Targeting of Ttyh1 effectively inhibited the formation of invasive TMs and glioma growth, but increased network formation by intercellular TMs, suggesting a functional and molecular heterogeneity of the recently discovered TMs with potential implications for future TM-targeting strategies.
Collapse
|
181
|
Pernpeintner C, Frank JA, Urban P, Roeske CR, Pritzl SD, Trauner D, Lohmüller T. Light-Controlled Membrane Mechanics and Shape Transitions of Photoswitchable Lipid Vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:4083-4089. [PMID: 28361538 DOI: 10.1021/acs.langmuir.7b01020] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Giant unilamellar vesicles (GUVs) represent a versatile model system to emulate the fundamental properties and functions associated with the plasma membrane of living cells. Deformability and shape transitions of lipid vesicles are closely linked to the mechanical properties of the bilayer membrane itself and are typically difficult to control under physiological conditions. Here, we developed a protocol to form cell-sized vesicles from an azobenzene-containing phosphatidylcholine (azo-PC), which undergoes photoisomerization on irradiation with UV-A and visible light. Photoswitching within the photolipid vesicles enabled rapid and precise control of the mechanical properties of the membrane. By varying the intensity and dynamics of the optical stimulus, controlled vesicle shape changes such as budding transitions, invagination, pearling, or the formation of membrane tubes were achieved. With this system, we could mimic the morphology changes normally seen in cells, in the absence of any molecular machines associated with the cytoskeleton. Furthermore, we devised a mechanism to utilize photoswitchable lipid membranes for storing mechanical energy and then releasing it on command as locally usable work.
Collapse
Affiliation(s)
- Carla Pernpeintner
- Photonics and Optoelectronics Group, Department of Physics and CeNS, Ludwig Maximilians University Munich , Amalienstraße 54, 80799 Munich, Germany
- Nanosystems Initiative Munich, Schellingstraße 4, 80799 Munich, Germany
| | - James A Frank
- Department of Chemistry and Center for Integrated Protein Science, Ludwig Maximilians University Munich , Butenandtstraße 5-13, 81377 Munich, Germany
| | - Patrick Urban
- Photonics and Optoelectronics Group, Department of Physics and CeNS, Ludwig Maximilians University Munich , Amalienstraße 54, 80799 Munich, Germany
| | - Christian R Roeske
- Photonics and Optoelectronics Group, Department of Physics and CeNS, Ludwig Maximilians University Munich , Amalienstraße 54, 80799 Munich, Germany
| | - Stefanie D Pritzl
- Photonics and Optoelectronics Group, Department of Physics and CeNS, Ludwig Maximilians University Munich , Amalienstraße 54, 80799 Munich, Germany
| | - Dirk Trauner
- Department of Chemistry and Center for Integrated Protein Science, Ludwig Maximilians University Munich , Butenandtstraße 5-13, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstraße 4, 80799 Munich, Germany
| | - Theobald Lohmüller
- Photonics and Optoelectronics Group, Department of Physics and CeNS, Ludwig Maximilians University Munich , Amalienstraße 54, 80799 Munich, Germany
- Nanosystems Initiative Munich, Schellingstraße 4, 80799 Munich, Germany
| |
Collapse
|
182
|
Willnow TE. Nanotubes, the fast track to treatment of Dent disease? Kidney Int 2017; 91:776-778. [DOI: 10.1016/j.kint.2016.12.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/21/2023]
|
183
|
Bittins M, Wang X. TNT-Induced Phagocytosis: Tunneling Nanotubes Mediate the Transfer of Pro-Phagocytic Signals From Apoptotic to Viable Cells. J Cell Physiol 2017; 232:2271-2279. [PMID: 27591547 PMCID: PMC5485076 DOI: 10.1002/jcp.25584] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 09/01/2016] [Indexed: 12/24/2022]
Abstract
The exposure of phosphatidylserine (PS) on the surface membrane of apoptotic cells triggers the recruitment of phagocytic receptors and subsequently results in uptake by phagocytes. Here we describe how apoptotic cells can use intercellular membrane nanotubes to transfer exposed PS to neighboring viable cells, and thus deposit an "eat-me" tag on the viable cells. Tunneling nanotubes (TNTs) connected UV-treated apoptotic rat pheochromocytoma PC12 cells with neighboring untreated cells. These TNTs were composed of PS-exposed plasma membrane and facilitated the transfer of the membrane from apoptotic to viable cells. Other pro-phagocytic signals, such as oxidized phospholipids and calreticulin, were also transferred to viable cells. In addition, anti-phagocytic signal CD47 presenting on the plasma membrane of viable cells was masked by the transferred PS-membrane. Confocal imaging revealed an increase of phagocytosis of viable PC12 cells by murine RAW264.7 macrophages when the viable PC12 cells were cocultured with UV-treated PC12 cells. Treatment with 50 nM cytochalasin D would abolish TNTs and correspondingly inhibit this phagocytosis of the viable cells. Our study indicates that exposed-PS membrane is delivered from apoptotic to viable cells through TNTs. This transferred membrane may act as a pro-phagocytic signal for macrophages to induce phagocytosis of viable cells in a situation where they are in the vicinity of apoptotic cells. J. Cell. Physiol. 232: 2271-2279, 2017. © 2016 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
| | - Xiang Wang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
184
|
Periz J, Whitelaw J, Harding C, Gras S, Del Rosario Minina MI, Latorre-Barragan F, Lemgruber L, Reimer MA, Insall R, Heaslip A, Meissner M. Toxoplasma gondii F-actin forms an extensive filamentous network required for material exchange and parasite maturation. eLife 2017; 6:e24119. [PMID: 28322189 PMCID: PMC5375643 DOI: 10.7554/elife.24119] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/09/2017] [Indexed: 01/14/2023] Open
Abstract
Apicomplexan actin is important during the parasite's life cycle. Its polymerization kinetics are unusual, permitting only short, unstable F-actin filaments. It has not been possible to study actin in vivo and so its physiological roles have remained obscure, leading to models distinct from conventional actin behaviour. Here a modified version of the commercially available actin-chromobody was tested as a novel tool for visualising F-actin dynamics in Toxoplasma gondii. Cb labels filamentous actin structures within the parasite cytosol and labels an extensive F-actin network that connects parasites within the parasitophorous vacuole and allows vesicles to be exchanged between parasites. In the absence of actin, parasites lack a residual body and inter-parasite connections and grow in an asynchronous and disorganized manner. Collectively, these data identify new roles for actin in the intracellular phase of the parasites lytic cycle and provide a robust new tool for imaging parasitic F-actin dynamics.
Collapse
Affiliation(s)
- Javier Periz
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Jamie Whitelaw
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Clare Harding
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Simon Gras
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Mario Igor Del Rosario Minina
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Fernanda Latorre-Barragan
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Leandro Lemgruber
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Madita Alice Reimer
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Robert Insall
- Cancer Research United Kingdom Beatson Institute, Bearsden, United Kingdom
| | - Aoife Heaslip
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, United States
| | - Markus Meissner
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
185
|
Internalization, axonal transport and release of fibrillar forms of alpha-synuclein. Neurobiol Dis 2017; 109:219-225. [PMID: 28323023 DOI: 10.1016/j.nbd.2017.03.007] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/21/2017] [Accepted: 03/15/2017] [Indexed: 11/20/2022] Open
Abstract
Intra-neuronal protein aggregates made of fibrillar alpha-synuclein (α-syn) are the hallmark of Parkinson's disease (PD). With time, these aggregates spread through the brain following axonal projections. Understanding the mechanism of this spread is central to the study of the progressive nature of PD. Here we review data relevant to the uptake, transport and release of α-syn fibrils. We summarize several cell surface receptors that regulate the uptake of α-syn fibrils by neurons. The aggregates are then transported along axons, both in the anterograde and retrograde direction. The kinetics of transport suggests that they are part of the slow component b of axonal transport. Recent findings indicate that aggregated α-syn is secreted by neurons by non-canonical pathways that may implicate various molecular chaperones including USP19 and the DnaJ/Hsc70 complex. Additionally, α-syn fibrils may also be released and transmitted from neuron-to-neuron via exosomes and tunneling nanotubes. Understanding these different mechanisms and molecular players underlying α-syn spread is crucial for the development of therapies that could halt the progression of α-syn-related degenerative diseases.
Collapse
|
186
|
Okuda S, Uemura N, Takahashi R. Alpha-synuclein fibrils propagate through tunneling nanotubes. Mov Disord 2017; 32:394. [PMID: 28218419 DOI: 10.1002/mds.26909] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 11/10/2022] Open
Affiliation(s)
- Shinya Okuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norihito Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
187
|
C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nature 2017; 542:367-371. [PMID: 28178240 PMCID: PMC5336134 DOI: 10.1038/nature21362] [Citation(s) in RCA: 300] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 01/03/2017] [Indexed: 12/12/2022]
|
188
|
Isola AL, Chen S. Exosomes: The Messengers of Health and Disease. Curr Neuropharmacol 2017; 15:157-165. [PMID: 27568544 PMCID: PMC5327461 DOI: 10.2174/1570159x14666160825160421] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 12/21/2022] Open
Abstract
Exosomes are small vesicles comprised of a lipid bilayer containing various proteins, RNAs and bioactive lipids. They act as intercellular messengers that give the ability to communicate between both cells of the same type and other cell types. They are released by healthy cells, both constitutively and upon cell activation and play an important role in immune system function. Exosomes are essential for healthy physiological conditions, however under pathological circumstances, they act to potentiate cellular stress and damage. This review explores the characteristics, biogenesis, role(s) in the pathogenesis of diseases and role(s) in progression of cancer of these nano-sized messages-in-a-vesicle: exosomes.
Collapse
Affiliation(s)
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers, the State University, Piscataway, NJ, 08854, USA
| |
Collapse
|
189
|
Gabriel SS, Belge H, Gassama A, Debaix H, Luciani A, Fehr T, Devuyst O. Bone marrow transplantation improves proximal tubule dysfunction in a mouse model of Dent disease. Kidney Int 2017; 91:842-855. [PMID: 28143656 DOI: 10.1016/j.kint.2016.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/01/2016] [Accepted: 11/23/2016] [Indexed: 01/02/2023]
Abstract
Dent disease is a rare X-linked tubulopathy caused by mutations in the endosomal chloride-proton exchanger (ClC-5) resulting in defective receptor-mediated endocytosis and severe proximal tubule dysfunction. Bone marrow transplantation has recently been shown to preserve kidney function in cystinosis, a lysosomal storage disease causing proximal tubule dysfunction. Here we test the effects of bone marrow transplantation in Clcn5Y/- mice, a faithful model for Dent disease. Transplantation of wild-type bone marrow in Clcn5Y/- mice significantly improved proximal tubule dysfunction, with decreased low-molecular-weight proteinuria, glycosuria, calciuria, and polyuria four months after transplantation, compared to Clcn5Y/- mice transplanted with ClC-5 knockout bone marrow. Bone marrow-derived cells engrafted in the interstitium, surrounding proximal tubule cells, which showed a rescue of the apical expression of ClC-5 and megalin receptors. The improvement of proximal tubule dysfunction correlated with Clcn5 gene expression in kidneys of mice transplanted with wild-type bone marrow cells. Coculture of Clcn5Y/- proximal tubule cells with bone marrow-derived cells confirmed rescue of ClC-5 and megalin, resulting in improved endocytosis. Nanotubular extensions between the engrafted bone marrow-derived cells and proximal tubule cells were observed in vivo and in vitro. No rescue was found when the formation of the tunneling nanotubes was prevented by actin depolymerization or when cells were physically separated by transwell inserts. Thus, bone marrow transplantation may rescue the epithelial phenotype due to an inherited endosomal defect. Direct contacts between bone marrow-derived cells and diseased tubular cells play a key role in the rescue mechanism.
Collapse
Affiliation(s)
- Sarah S Gabriel
- Institute of Physiology, University of Zürich, Zürich, Switzerland; Division of Nephrology, University Hospital Zürich, Zürich, Switzerland
| | - Hendrica Belge
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Alkaly Gassama
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Huguette Debaix
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | | | - Thomas Fehr
- Institute of Physiology, University of Zürich, Zürich, Switzerland; Division of Nephrology, University Hospital Zürich, Zürich, Switzerland; Department of Internal Medicine, Cantonal Hospital Graubünden, Chur, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
190
|
van Diggelen F, Tepper AWJW, Apetri MM, Otzen DE. α-Synuclein Oligomers: A Study in Diversity. Isr J Chem 2016. [DOI: 10.1002/ijch.201600116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Femke van Diggelen
- Crossbeta Biosciences; Padualaan 8 3584CH Utrecht The Netherlands
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| | | | | | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| |
Collapse
|
191
|
Delage E, Cervantes DC, Pénard E, Schmitt C, Syan S, Disanza A, Scita G, Zurzolo C. Differential identity of Filopodia and Tunneling Nanotubes revealed by the opposite functions of actin regulatory complexes. Sci Rep 2016; 6:39632. [PMID: 28008977 PMCID: PMC5180355 DOI: 10.1038/srep39632] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/25/2016] [Indexed: 11/09/2022] Open
Abstract
Tunneling Nanotubes (TNTs) are actin enriched filopodia-like protrusions that play a pivotal role in long-range intercellular communication. Different pathogens use TNT-like structures as "freeways" to propagate across cells. TNTs are also implicated in cancer and neurodegenerative diseases, making them promising therapeutic targets. Understanding the mechanism of their formation, and their relation with filopodia is of fundamental importance to uncover their physiological function, particularly since filopodia, differently from TNTs, are not able to mediate transfer of cargo between distant cells. Here we studied different regulatory complexes of actin, which play a role in the formation of both these structures. We demonstrate that the filopodia-promoting CDC42/IRSp53/VASP network negatively regulates TNT formation and impairs TNT-mediated intercellular vesicle transfer. Conversely, elevation of Eps8, an actin regulatory protein that inhibits the extension of filopodia in neurons, increases TNT formation. Notably, Eps8-mediated TNT induction requires Eps8 bundling but not its capping activity. Thus, despite their structural similarities, filopodia and TNTs form through distinct molecular mechanisms. Our results further suggest that a switch in the molecular composition in common actin regulatory complexes is critical in driving the formation of either type of membrane protrusion.
Collapse
Affiliation(s)
- Elise Delage
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Diégo Cordero Cervantes
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Esthel Pénard
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Christine Schmitt
- Ultrapole, 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
| | - Andrea Disanza
- FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Giorgio Scita
- FIRC Institute of Molecular Oncology, 20139 Milan, Italy.,Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20122 Milan, Italy
| | - Chiara Zurzolo
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| |
Collapse
|
192
|
Rab8a/Rab11a regulate intercellular communications between neural cells via tunneling nanotubes. Cell Death Dis 2016; 7:e2523. [PMID: 28005071 PMCID: PMC5260982 DOI: 10.1038/cddis.2016.441] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/01/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022]
Abstract
Tunneling nanotubes (TNTs) are F-actin-based membrane tubes, and can form between cultured cells and within vital tissues. TNTs mediate intercellular communications that range from electrical signaling to the transfer of organelles. Following peripheral nerve injury, the orchestrated intercellular communications among neural and non-neural cells are required for effective nerve regeneration. It remains unknown whether TNTs exist between neural cells in the peripheral nerve system and how TNTs affect neural regeneration. To address these interesting questions, we investigated the transfer of neurotropic factors, membrane protein, cytoplasmic protein, mitochondria and RNA in functional TNTs formed between cultured Schwann cells (SCs). TNT-like structures were increased not only in cultured SCs after exposure to serum depletion but also in longitudinal sections of proximal sciatic nerve stump harvested after rat peripheral nerve transection. Meanwhile, downregulation of Rab8a or Rab11a in cultured SCs inhibited the formation of functional TNTs and vesicle transfer and led to decrease in cell migration, increase in SCs apoptosis. Likewise, knockdown of Rab8a or Rab11a in primary SCs also suppressed axonal outgrowth from co-cultured dorsal root ganglion (DRG) neurons. Overall, our results suggested that the gene of Rab8a or Rab11a might be involved in the formation of TNTs structures in the peripheral nerve system, while TNTs structures were likely to affect peripheral nerve regeneration through the regulation of neural cell communications.
Collapse
|
193
|
Popov LD. Mitochondrial networking in diabetic left ventricle cardiomyocytes. Mitochondrion 2016; 34:24-31. [PMID: 28007605 DOI: 10.1016/j.mito.2016.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 10/07/2016] [Accepted: 12/16/2016] [Indexed: 12/17/2022]
Abstract
Cardiomyocyte mitochondria preserve "the quorum sensing" attribute of their aerobic bacterial ancestors, as shown by the transient physical connectivity and communication not only with each other, but also with other intracellular organelles and with cytosol, ensuing cellular homeostasis. In this review, we present original electron microscopy evidence on mitochondrial networking within diabetic left ventricular cardiomyocytes, focusing on: (i) the inter-mitochondrial communication, allowing electrochemical signals transfer and outer membrane components or matrix proteins exchange, (ii) the interplay between mitochondria and the cardiomyocyte nucleus, nucleolus, sarcoplasmic reticulum, lysosomes, and lipid droplets viewed as attributes of mitochondrial "quality control" and "retrograde signaling function", and (iii) the crosstalk between mitochondria and cardiomyocyte cytosol, as part of the adaptive responses that allow cells survival. Confirmation of such interactions in diabetic myocardium and identification of molecules involved are ongoing, foreseeing the alleviation of heart contractile dysfunction in cardiomyopathy.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- "Nicolae Simionescu" Institute of Cellular Biology and Pathology of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest 050568, Romania.
| |
Collapse
|
194
|
Martinez MG, Kielian M. Intercellular Extensions Are Induced by the Alphavirus Structural Proteins and Mediate Virus Transmission. PLoS Pathog 2016; 12:e1006061. [PMID: 27977778 PMCID: PMC5158078 DOI: 10.1371/journal.ppat.1006061] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/13/2016] [Indexed: 11/19/2022] Open
Abstract
Alphaviruses are highly organized enveloped RNA viruses with an internal nucleocapsid surrounded by a membrane containing the E2 and E1 transmembrane proteins. Alphavirus budding takes place at the plasma membrane and requires the interaction of the cytoplasmic domain of E2 with the capsid protein. Here we used WT alphaviruses and Sindbis virus in which E2 was fused to a fluorescent protein to characterize virus exit from host cells. Our results show that alphavirus infection induced striking modifications of the host cell cytoskeleton and resulted in the formation of stable intercellular extensions that emanated exclusively from the infected cell. The intercellular extensions were long (> 10 μM), contained actin and tubulin, and formed flattened contacts with neighboring cells, but did not mediate membrane or cytoplasmic continuity between cells. Receptor down-regulation studies indicated that formation of stable extensions did not require the virus receptor, and that extensions promoted cell-to-cell virus transmission to receptor-depleted cells. Virus mutant experiments demonstrated that formation of extensions required the E2-capsid interaction but not active particle budding, while intercellular transmission of infection required the production of fusion-active virus particles. Protein expression studies showed that even in the absence of virus infection, the viral structural proteins alone induced intercellular extensions, and that these extensions were preferentially targeted to non-expressing cells. Together, our results identify a mechanism for alphavirus cell-to-cell transmission and define the key viral protein interactions that it requires. Alphaviruses are a group of small enveloped RNA viruses that include a number of important human pathogens such as Chikungunya virus and viruses that cause fatal encephalitis. Chikungunya virus emerged recently in a number of countries worldwide including the Americas, where it has caused major outbreaks. Vaccines and anti-viral strategies for these viruses are urgently needed, and basic information on the alphavirus infection pathway will help in targeting critical steps. Here we describe the changes in the alphavirus-infected cell that allow it to transmit virus to neighboring uninfected cells. Infected cells form long extensions that contact neighboring cells and mediate cell-to-cell virus transmission. This mechanism of virus transmission may help to shield virus from neutralization by host antibodies. Surprisingly, expression of the viral structural proteins alone induces these intercellular extensions, which preferentially target non-expressing cells. We used this system to define a critical interaction of the capsid and envelope protein that is required for formation of extensions.
Collapse
Affiliation(s)
- Maria Guadalupe Martinez
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
195
|
El Najjar F, Cifuentes-Muñoz N, Chen J, Zhu H, Buchholz UJ, Moncman CL, Dutch RE. Human metapneumovirus Induces Reorganization of the Actin Cytoskeleton for Direct Cell-to-Cell Spread. PLoS Pathog 2016; 12:e1005922. [PMID: 27683250 PMCID: PMC5040343 DOI: 10.1371/journal.ppat.1005922] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/08/2016] [Indexed: 11/22/2022] Open
Abstract
Paramyxovirus spread generally involves assembly of individual viral particles which then infect target cells. We show that infection of human bronchial airway cells with human metapneumovirus (HMPV), a recently identified paramyxovirus which causes significant respiratory disease, results in formation of intercellular extensions and extensive networks of branched cell-associated filaments. Formation of these structures is dependent on actin, but not microtubule, polymerization. Interestingly, using a co-culture assay we show that conditions which block regular infection by HMPV particles, including addition of neutralizing antibodies or removal of cell surface heparan sulfate, did not prevent viral spread from infected to new target cells. In contrast, inhibition of actin polymerization or alterations to Rho GTPase signaling pathways significantly decreased cell-to-cell spread. Furthermore, viral proteins and viral RNA were detected in intercellular extensions, suggesting direct transfer of viral genetic material to new target cells. While roles for paramyxovirus matrix and fusion proteins in membrane deformation have been previously demonstrated, we show that the HMPV phosphoprotein extensively co-localized with actin and induced formation of cellular extensions when transiently expressed, supporting a new model in which a paramyxovirus phosphoprotein is a key player in assembly and spread. Our results reveal a novel mechanism for HMPV direct cell-to-cell spread and provide insights into dissemination of respiratory viruses. Human metapneumovirus (HMPV) is an important human respiratory pathogen that affects all age groups worldwide. There are currently no vaccines or treatments available for HMPV, and key aspects of its life cycle remain unknown. We examined the late events of the HMPV infection cycle in human bronchial epithelial cells. Our data demonstrate that HMPV infection leads to formation of unique structures, including intercellular extensions connecting cells, and large networks of branched cell-associated filaments. Viral modulation of the cellular cytoskeleton and cellular signaling pathways are important for formation of these structures. Our results are consistent with the intercellular extensions playing a role in direct spread of virus from cell-to-cell, potentially by transfer of virus genetic material without particle formation. We also show that the HMPV phosphoprotein localizes with actin and can promote membrane deformations, suggesting a novel role in viral assembly or spread for paramyxovirus phosphoproteins.
Collapse
Affiliation(s)
- Farah El Najjar
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Nicolás Cifuentes-Muñoz
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Ursula J. Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Carole L. Moncman
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Rebecca Ellis Dutch
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
196
|
Zhang Y, Yu Z, Jiang D, Liang X, Liao S, Zhang Z, Yue W, Li X, Chiu SM, Chai YH, Liang Y, Chow Y, Han S, Xu A, Tse HF, Lian Q. iPSC-MSCs with High Intrinsic MIRO1 and Sensitivity to TNF-α Yield Efficacious Mitochondrial Transfer to Rescue Anthracycline-Induced Cardiomyopathy. Stem Cell Reports 2016; 7:749-763. [PMID: 27641650 PMCID: PMC5063626 DOI: 10.1016/j.stemcr.2016.08.009] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/11/2016] [Accepted: 08/16/2016] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can donate mitochondria and rescue anthracycline-induced cardiomyocyte (CM) damage, although the underlying mechanisms remain elusive. We determined that the superior efficiency of mitochondrial transfer by human induced-pluripotent-stem-cell-derived MSCs (iPSC-MSCs) compared with bone marrow-derived MSCs (BM-MSCs) is due to high expression of intrinsic Rho GTPase 1 (MIRO1). Further, due to a higher level of TNFαIP2 expression, iPSC-MSCs are more responsive to tumor necrosis factor alpha (TNF-α)-induced tunneling nanotube (TNT) formation for mitochondrial transfer to CMs, which is regulated via the TNF-α/NF-κB/TNFαIP2 signaling pathway. Inhibition of TNFαIP2 or MIRO1 in iPSC-MSCs reduced the efficiency of mitochondrial transfer and decreased CMs protection. Compared with BM-MSCs, transplantation of iPSC-MSCs into a mouse model of anthracycline-induced cardiomyopathy resulted in more human mitochondrial retention and bioenergetic preservation in heart tissue. Efficacious transfer of mitochondria from iPSC-MSCs to CMs, due to higher MIRO1 expression and responsiveness to TNF-α-induced nanotube formation, effectively attenuates anthracycline-induced CM damage. Functional mitochondrial transfer of iPSC-MSCs attenuates Dox-induced cardiomyopathy High intrinsic Miro1 in iPSC-MSCs contributes to efficacious mitochondrial transfer iPSC-MSCs are highly responsive to TNF-α-induced tunneling nanotube formation
Collapse
Affiliation(s)
- Yuelin Zhang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhendong Yu
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen 518000, China
| | - Dan Jiang
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, China
| | - Xiaoting Liang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Songyan Liao
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhao Zhang
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, China
| | - Wensheng Yue
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Li
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sin-Ming Chiu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuet-Hung Chai
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Yingmin Liang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yenyen Chow
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shuo Han
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China; Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, The University of Hong Kong, Hong Kong, China.
| | - Qizhou Lian
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Ophthalmology, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China; Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen 518000, China.
| |
Collapse
|
197
|
Fykerud TA, Knudsen LM, Totland MZ, Sørensen V, Dahal-Koirala S, Lothe RA, Brech A, Leithe E. Mitotic cells form actin-based bridges with adjacent cells to provide intercellular communication during rounding. Cell Cycle 2016; 15:2943-2957. [PMID: 27625181 PMCID: PMC5105929 DOI: 10.1080/15384101.2016.1231280] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In order to achieve accurate chromosome segregation, eukaryotic cells undergo a dramatic change in morphology to obtain a spherical shape during mitosis. Interphase cells communicate directly with each other by exchanging ions and small molecules via gap junctions, which have important roles in controlling cell growth and differentiation. As cells round up during mitosis, the gap junctional communication between mitotic cells and adjacent interphase cells ceases. Whether mitotic cells use alternative mechanisms for mediating direct cell-cell communication during rounding is currently unknown. Here, we have studied the mechanisms involved in the remodeling of gap junctions during mitosis. We further demonstrate that mitotic cells are able to form actin-based plasma membrane bridges with adjacent cells during rounding. These structures, termed “mitotic nanotubes,” were found to be involved in mediating the transport of cytoplasm, including Rab11-positive vesicles, between mitotic cells and adjacent cells. Moreover, a subpool of the gap-junction channel protein connexin43 localized in these intercellular bridges during mitosis. Collectively, the data provide new insights into the mechanisms involved in the remodeling of gap junctions during mitosis and identify actin-based plasma membrane bridges as a novel means of communication between mitotic cells and adjacent cells during rounding.
Collapse
Affiliation(s)
- Tone A Fykerud
- a Department of Molecular Oncology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,c Institute for Biosciences, University of Oslo , Oslo , Norway.,d K.G. Jebsen Colorectal Cancer Research Center, Oslo University Hospital , Oslo , Norway
| | - Lars M Knudsen
- a Department of Molecular Oncology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,c Institute for Biosciences, University of Oslo , Oslo , Norway.,d K.G. Jebsen Colorectal Cancer Research Center, Oslo University Hospital , Oslo , Norway
| | - Max Z Totland
- a Department of Molecular Oncology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,c Institute for Biosciences, University of Oslo , Oslo , Norway.,d K.G. Jebsen Colorectal Cancer Research Center, Oslo University Hospital , Oslo , Norway
| | - Vigdis Sørensen
- b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,e Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,f Department of Core Facilities , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Shiva Dahal-Koirala
- a Department of Molecular Oncology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,c Institute for Biosciences, University of Oslo , Oslo , Norway
| | - Ragnhild A Lothe
- a Department of Molecular Oncology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,c Institute for Biosciences, University of Oslo , Oslo , Norway.,d K.G. Jebsen Colorectal Cancer Research Center, Oslo University Hospital , Oslo , Norway
| | - Andreas Brech
- b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,c Institute for Biosciences, University of Oslo , Oslo , Norway.,e Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,f Department of Core Facilities , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Edward Leithe
- a Department of Molecular Oncology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway.,d K.G. Jebsen Colorectal Cancer Research Center, Oslo University Hospital , Oslo , Norway
| |
Collapse
|
198
|
López-Leal R, Alvarez J, Court FA. Origin of axonal proteins: Is the axon-schwann cell unit a functional syncytium? Cytoskeleton (Hoboken) 2016; 73:629-639. [DOI: 10.1002/cm.21319] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/28/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Rodrigo López-Leal
- Faculty of Sciences, Center for Integrative Biology; Universidad Mayor; Santiago Chile
- Geroscience Center for Brain Health and Metabolism; Santiago Chile
- Millenium Nucleus for Regenerative Biology; Santiago Chile
| | - Jaime Alvarez
- Faculty of Sciences, Center for Integrative Biology; Universidad Mayor; Santiago Chile
- Millenium Nucleus for Regenerative Biology; Santiago Chile
| | - Felipe A. Court
- Faculty of Sciences, Center for Integrative Biology; Universidad Mayor; Santiago Chile
- Geroscience Center for Brain Health and Metabolism; Santiago Chile
- Millenium Nucleus for Regenerative Biology; Santiago Chile
| |
Collapse
|
199
|
Abounit S, Bousset L, Loria F, Zhu S, de Chaumont F, Pieri L, Olivo-Marin JC, Melki R, Zurzolo C. Tunneling nanotubes spread fibrillar α-synuclein by intercellular trafficking of lysosomes. EMBO J 2016; 35:2120-2138. [PMID: 27550960 DOI: 10.15252/embj.201593411] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/22/2016] [Indexed: 12/25/2022] Open
Abstract
Synucleinopathies such as Parkinson's disease are characterized by the pathological deposition of misfolded α-synuclein aggregates into inclusions throughout the central and peripheral nervous system. Mounting evidence suggests that intercellular propagation of α-synuclein aggregates may contribute to the neuropathology; however, the mechanism by which spread occurs is not fully understood. By using quantitative fluorescence microscopy with co-cultured neurons, here we show that α-synuclein fibrils efficiently transfer from donor to acceptor cells through tunneling nanotubes (TNTs) inside lysosomal vesicles. Following transfer through TNTs, α-synuclein fibrils are able to seed soluble α-synuclein aggregation in the cytosol of acceptor cells. We propose that donor cells overloaded with α-synuclein aggregates in lysosomes dispose of this material by hijacking TNT-mediated intercellular trafficking. Our findings thus reveal a possible novel role of TNTs and lysosomes in the progression of synucleinopathies.
Collapse
Affiliation(s)
- Saïda Abounit
- Institut Pasteur, Unité Trafic Membranaire et Pathogénèse, Paris Cedex 15, France
| | - Luc Bousset
- Paris-Saclay Institute of Neuroscience, CNRS, Gif-sur-Yvette, France
| | - Frida Loria
- Institut Pasteur, Unité Trafic Membranaire et Pathogénèse, Paris Cedex 15, France
| | - Seng Zhu
- Institut Pasteur, Unité Trafic Membranaire et Pathogénèse, Paris Cedex 15, France
| | - Fabrice de Chaumont
- Laboratoire d'Analyse d'Images Quantitative, Institut Pasteur, Paris Cedex 15, France
| | - Laura Pieri
- Paris-Saclay Institute of Neuroscience, CNRS, Gif-sur-Yvette, France
| | | | - Ronald Melki
- Paris-Saclay Institute of Neuroscience, CNRS, Gif-sur-Yvette, France
| | - Chiara Zurzolo
- Institut Pasteur, Unité Trafic Membranaire et Pathogénèse, Paris Cedex 15, France
| |
Collapse
|
200
|
Sandison ME, Dempster J, McCarron JG. The transition of smooth muscle cells from a contractile to a migratory, phagocytic phenotype: direct demonstration of phenotypic modulation. J Physiol 2016; 594:6189-6209. [PMID: 27393389 PMCID: PMC5088226 DOI: 10.1113/jp272729] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/26/2016] [Indexed: 12/13/2022] Open
Abstract
Key points Smooth muscle cell (SMC) phenotypic conversion from a contractile to a migratory phenotype is proposed to underlie cardiovascular disease but its contribution to vascular remodelling and even its existence have recently been questioned. Tracking the fate of individual SMCs is difficult as no specific markers of migratory SMCs exist. This study used a novel, prolonged time‐lapse imaging approach to continuously track the behaviour of unambiguously identified, fully differentiated SMCs. In response to serum, highly‐elongated, contractile SMCs initially rounded up, before spreading and migrating and these migratory cells displayed clear phagocytic activity. This study provides a direct demonstration of the transition of fully contractile SMCs to a non‐contractile, migratory phenotype with phagocytic capacity that may act as a macrophage‐like cell.
Abstract Atherosclerotic plaques are populated with smooth muscle cells (SMCs) and macrophages. SMCs are thought to accumulate in plaques because fully differentiated, contractile SMCs reprogramme into a ‘synthetic’ migratory phenotype, so‐called phenotypic modulation, whilst plaque macrophages are thought to derive from blood‐borne myeloid cells. Recently, these views have been challenged, with reports that SMC phenotypic modulation may not occur during vascular remodelling and that plaque macrophages may not be of haematopoietic origin. Following the fate of SMCs is complicated by the lack of specific markers for the migratory phenotype and direct demonstrations of phenotypic modulation are lacking. Therefore, we employed long‐term, high‐resolution, time‐lapse microscopy to track the fate of unambiguously identified, fully‐differentiated, contractile SMCs in response to the growth factors present in serum. Phenotypic modulation was clearly observed. The highly elongated, contractile SMCs initially rounded up, for 1–3 days, before spreading outwards. Once spread, the SMCs became motile and displayed dynamic cell‐cell communication behaviours. Significantly, they also displayed clear evidence of phagocytic activity. This macrophage‐like behaviour was confirmed by their internalisation of 1 μm fluorescent latex beads. However, migratory SMCs did not uptake acetylated low‐density lipoprotein or express the classic macrophage marker CD68. These results directly demonstrate that SMCs may rapidly undergo phenotypic modulation and develop phagocytic capabilities. Resident SMCs may provide a potential source of macrophages in vascular remodelling. Smooth muscle cell (SMC) phenotypic conversion from a contractile to a migratory phenotype is proposed to underlie cardiovascular disease but its contribution to vascular remodelling and even its existence have recently been questioned. Tracking the fate of individual SMCs is difficult as no specific markers of migratory SMCs exist. This study used a novel, prolonged time‐lapse imaging approach to continuously track the behaviour of unambiguously identified, fully differentiated SMCs. In response to serum, highly‐elongated, contractile SMCs initially rounded up, before spreading and migrating and these migratory cells displayed clear phagocytic activity. This study provides a direct demonstration of the transition of fully contractile SMCs to a non‐contractile, migratory phenotype with phagocytic capacity that may act as a macrophage‐like cell.
Collapse
Affiliation(s)
- Mairi E Sandison
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - John Dempster
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK.
| |
Collapse
|