Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 891 word(s) 891 2021-03-02 04:53:59 |
2 update layout and reference Meta information modification 891 2021-03-10 03:34:57 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wang, X. Tunneling Nanotubes. Encyclopedia. Available online: https://encyclopedia.pub/entry/7848 (accessed on 30 June 2024).
Wang X. Tunneling Nanotubes. Encyclopedia. Available at: https://encyclopedia.pub/entry/7848. Accessed June 30, 2024.
Wang, Xiang. "Tunneling Nanotubes" Encyclopedia, https://encyclopedia.pub/entry/7848 (accessed June 30, 2024).
Wang, X. (2021, March 09). Tunneling Nanotubes. In Encyclopedia. https://encyclopedia.pub/entry/7848
Wang, Xiang. "Tunneling Nanotubes." Encyclopedia. Web. 09 March, 2021.
Tunneling Nanotubes
Edit

Tunneling nanotubes (TNTs) are recognized long membrane nanotubes connecting distance cells. In the last decade, growing evidence has shown that these subcellular structures mediate the specific transfer of cellular materials, pathogens, and electrical signals between cells. As intercellular bridges, they play a unique role in embryonic development, collective cell migration, injured cell recovery, cancer treatment resistance, and pathogen propagation. Although TNTs have been considered as potential drug targets for treatment, there is still a long way to go to translate the research findings into clinical practice.

tunneling nanotubes intercellular communication heterogeneity communication efficiency

1. Introduction

Multicellular organisms coordinate cell behavior, regulate morphogenesis, and maintain tissue homeostasis by secreting chemical molecules, releasing exosomes, and establishing direct connections such as neuronal synapses and gap junctions [1]. In 2004, the group of Gerdes reported for the first time a new way of long-distance cell–cell communication, tunneling nanotubes (TNTs) [2]. TNTs are tubular membrane structures with diameters of several hundred nanometers, which contain F-actin and, usually, adhesion proteins at one end (Figure 1) [3][4]. Hovering above the substrate, they directly connect adjacent cells up to hundreds of micrometers apart. These unique morphological features make them different from other cellular protrusions, such as filopodia and cytoneme. Numerous studies over the past ten years have shown that TNTs are widely present in various cell types [5][6]. More importantly, TNTs enable the transfer of small molecules, proteins, vesicles, and organelles between cells [2][7][8][9][10][11]. In 2010, we discovered that TNTs mediated depolarization coupling in non-neuronal cells, indicating that TNTs facilitate electrical signal transduction in addition to material transport [12]. Indeed, due to the characteristics of long-distance, high specificity, and multilevel transportation, TNT communication was dubbed the Internet of cells [13].

Figure 1. The schematic representation illustrates the structure and general composition of tunneling nanotubes (TNTs).

2. TNTs Are Heterogeneous Structures

2.1. Difference in Morphology

TNTs exhibit high variability in their morphology in terms of length and thickness [5][14]. Though the length of TNTs usually changes with the cell spacing caused by cell movement, it ranges from 10 to 100 µm in most types of cells. In a few cases, TNTs have been described as long cytoplasmic extensions up to 300 µm in length [6]. However, these fragile structures may break during prolongation if the pulling force by the cells exceeds the mechanical strength of TNTs [2][15]. In any event, the maximum length of a TNT is crucial since it determines the communication distance between cells. By analyzing the electron microscope images, the diameter of the TNTs was measured from hundreds of nanometers to a microscale [16]. One explanation for such a variation is that TNTs containing microtubules display thicker morphology [17][18][19]. Using cryo-electron microscopy, Sartori-Rupp and his colleagues recently revealed that TNTs were composed of a bunch of ultrathin tubes in mouse catecholaminergic CAD cells and human neuroblastoma SH-SY5Y cells, which are hardly distinguished by conventional confocal microscopy [3]. According to this study, the diameter of a TNT lacking microtubules may be determined by the number of ultrathin tubes. Since the identification of TNTs is still based on their morphological characteristics, the morphological diversity of TNTs has brought confusion to the nomenclature and the literature review on TNT research. The establishment of criteria for the classification of TNTs will be of great significance in this field.

2.2. Different Mechanisms of TNTs Formation

The importance of F-actin in TNT formation was first proved by our early study showing that a low dose of cytochalasin B, an actin inhibitor, could reduce the number of TNTs in PC12 cells [20]. Even in the MT-TNTs, F-actin plays a dominant role as well, because the treatment of microtubule inhibitors did not significantly disrupt the TNT structures [17][21]. Due to this line of thought, researchers considered that actin regulators and motor proteins were implicated in the formation of TNTs. Many laboratories successively identified several key proteins and signal pathways regulating TNT formation in different types of cells, such as M-sec/ERp29 [22][23][24], p53/Akt/PI3K/mTOR [25][26][27], Myosin10 [28][29], CDC42/IRSp53/VASP [30], and Rab11a/Rab8a [31][32]. Paradoxically, TNTs were also observed in M-sec or p53-deficient cells [28][33]. Such inconsistent results imply that the biogenesis of TNTs may not have a universal molecular regulation mechanism, probably due to their heterogeneity [34].

2.3. Where and When Do TNTs Form?

Although the evolutionary significance of TNTs biogenesis is completely unknown, cells under specific physiological circumstances may need these unique structures to establish communication that could not be accomplished by other types of intercellular connections: (i) distant cells connection [4][35][36], (ii) cells migration or invasion [37][38][39], and (iii) heterogeneous cells interaction [40][41][42][43]. Interestingly, the discovery of TNT-like structures between bacteria may provide clues in the study of the putative evolution of TNTs from bacteria to mammals [44]. Whether there are extracellular signals that trigger the formation and directionality of TNTs is still an exciting question. At least, quite a lot of studies have described that the number of TNT-like structures increased in inflammatory and stress conditions, such as pathogen infections [45][46][47], oxidative stress [48], high intracellular calcium concentrations [22], inflammatory signals [49][50], misfolded proteins, and pathogenic amyloid aggregates [8][28][51][52]. Moreover, the tumor microenvironment (hypoxia, acidic pH, hyperglycemia, and serum deprivation), as well as chemo- and radiotherapy-induced reactive oxygen species (ROS) production, leads to more TNTs in tumor cells [25][48][53][54][55][56][57][58]. Additionally, exosomes derived from malignant cells or vesicle recycling induce an increased rate in the formation of TNTs [32][59]. Under these circumstances, cells may respond to the stresses or stimulations by activating signaling pathways that initiate cytoskeleton rearrangement and cell movement, which consequently promote the formation of TNTs.

References

  1. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228.
  2. Rustom, A.; Saffrich, R.; Markovic, I.; Walther, P.; Gerdes, H.H. Nanotubular highways for intercellular organelle transport. Science 2004, 303, 1007–1010.
  3. Sartori-Rupp, A.; Cordero Cervantes, D.; Pepe, A.; Gousset, K.; Delage, E.; Corroyer-Dulmont, S.; Schmitt, C.; Krijnse-Locker, J.; Zurzolo, C. Correlative cryo-electron microscopy reveals the structure of TNTs in neuronal cells. Nat. Commun. 2019, 10, 342.
  4. Alarcon-Martinez, L.; Villafranca-Baughman, D.; Quintero, H.; Kacerovsky, J.B.; Dotigny, F.; Murai, K.K.; Prat, A.; Drapeau, P.; Di Polo, A. Interpericyte tunnelling nanotubes regulate neurovascular coupling. Nature 2020, 585, 91–95.
  5. Austefjord, M.W.; Gerdes, H.H.; Wang, X. Tunneling nanotubes: Diversity in morphology and structure. Commun. Integr. Biol. 2014, 7, e27934.
  6. Ariazi, J.; Benowitz, A.; De Biasi, V.; Den Boer, M.L.; Cherqui, S.; Cui, H.; Douillet, N.; Eugenin, E.A.; Favre, D.; Goodman, S.; et al. Tunneling Nanotubes and Gap Junctions-Their Role in Long-Range Intercellular Communication during Development, Health, and Disease Conditions. Front. Mol. Neurosci. 2017, 10, 333.
  7. Gurke, S.; Barroso, J.F.V.; Hodneland, E.; Bukoreshtliev, N.V.; Schlicker, O.; Gerdes, H.-H. Tunneling nanotube (TNT)-like structures facilitate a constitutive, actomyosin-dependent exchange of endocytic organelles between normal rat kidney cells. Exp. Cell Res. 2008, 314, 3669–3683.
  8. Costanzo, M.; Abounit, S.; Marzo, L.; Danckaert, A.; Chamoun, Z.; Roux, P.; Zurzolo, C. Transfer of polyglutamine aggregates in neuronal cells occurs in tunneling nanotubes. J. Cell Sci. 2013, 126 Pt 16, 3678–3685.
  9. Haimovich, G.; Ecker, C.M.; Dunagin, M.C.; Eggan, E.; Raj, A.; Gerst, J.E.; Singer, R.H. Intercellular mRNA trafficking via membrane nanotube-like extensions in mammalian cells. Proc. Natl. Acad. Sci. USA 2017, 114, E9873–E9882.
  10. 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.
  11. Kolba, M.D.; Dudka, W.; Zareba-Koziol, M.; Kominek, A.; Ronchi, P.; Turos, L.; Chroscicki, P.; Wlodarczyk, J.; Schwab, Y.; Klejman, A.; et al. Tunneling nanotube-mediated intercellular vesicle and protein transfer in the stroma-provided imatinib resistance in chronic myeloid leukemia cells. Cell Death Dis. 2019, 10, 817.
  12. Wang, X.; Veruki, M.L.; Bukoreshtliev, N.V.; Hartveit, E.; Gerdes, H.H. Animal cells connected by nanotubes can be electrically coupled through interposed gap-junction channels. Proc. Natl. Acad. Sci. USA 2010, 107, 17194–17199.
  13. Baker, M. How the Internet of cells has biologists buzzing. Nature 2017, 549, 322–324.
  14. Mittal, R.; Karhu, E.; Wang, J.S.; Delgado, S.; Zukerman, R.; Mittal, J.; Jhaveri, V.M. Cell communication by tunneling nanotubes: Implications in disease and therapeutic applications. J. Cell Physiol. 2019, 234, 1130–1146.
  15. Pontes, B.; Viana, N.B.; Campanati, L.; Farina, M.; Neto, V.M.; Nussenzveig, H.M. Structure and elastic properties of tunneling nanotubes. Eur. Biophys. J. Biophys. Lett. 2008, 37, 121–129.
  16. Pinto, G.; Brou, C.; Zurzolo, C. Tunneling Nanotubes: The Fuel of Tumor Progression? Trends Cancer 2020.
  17. Wang, X.; Gerdes, H.H. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ. 2015, 22, 1181–1191.
  18. Weil, S.; Osswald, M.; Solecki, G.; Grosch, J.; Jung, E.; Lemke, D.; Ratliff, M.; Hanggi, D.; Wick, W.; Winkler, F. Tumor microtubes convey resistance to surgical lesions and chemotherapy in gliomas. Neuro Oncol. 2017, 19, 1316–1326.
  19. Guo, L.; Zhang, Y.; Wei, R.; Wang, C.; Feng, M. Lipopolysaccharide-anchored macrophages hijack tumor microtube networks for selective drug transport and augmentation of antitumor effects in orthotopic lung cancer. Theranostics 2019, 9, 6936–6948.
  20. Bukoreshtliev, N.V.; Wang, X.; Hodneland, E.; Gurke, S.; Barroso, J.F.V.; Gerdes, H.-H. Selective block of tunneling nanotube (TNT) formation inhibits intercellular organelle transfer between PC12 cells. FEBS Lett. 2009, 583, 1481–1488.
  21. Hanna, S.J.; 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.
  22. Hase, K.; Kimura, S.; Takatsu, H.; Ohmae, M.; Kawano, S.; Kitamura, H.; Ito, M.; Watarai, H.; Hazelett, C.C.; Yeaman, C.; et al. M-Sec promotes membrane nanotube formation by interacting with Ral and the exocyst complex. Nat. Cell Biol. 2009, 11, 1427–1432.
  23. Hashimoto, M.; Bhuyan, F.; Hiyoshi, M.; Noyori, O.; Nasser, H.; Miyazaki, M.; Saito, T.; Kondoh, Y.; Osada, H.; Kimura, S.; et al. Potential Role of the Formation of Tunneling Nanotubes in HIV-1 Spread in Macrophages. J. Immunol. 2016, 196, 1832–1841.
  24. Pergu, R.; Dagar, S.; Kumar, H.; Kumar, R.; Bhattacharya, J.; Mylavarapu, S.V.S. The chaperone ERp29 is required for tunneling nanotube formation by stabilizing MSec. J. Biol. Chem. 2019, 294, 7177–7193.
  25. Wang, Y.; Cui, J.; Sun, X.; Zhang, Y. Tunneling-nanotube development in astrocytes depends on p53 activation. Cell Death Differ. 2011, 18, 732–742.
  26. Lu, J.J.; Yang, W.M.; Li, F.; Zhu, W.; Chen, Z. Tunneling Nanotubes Mediated microRNA-155 Intercellular Transportation Promotes Bladder Cancer Cells’ Invasive and Proliferative Capacity. Int. J. Nanomed. 2019, 14, 9731–9743.
  27. Sharma, M.; Subramaniam, S. Rhes travels from cell to cell and transports Huntington disease protein via TNT-like protrusion. J. Cell Biol. 2019, 218, 1972–1993.
  28. Gousset, K.; Marzo, L.; Commere, P.H.; Zurzolo, C. Myo10 is a key regulator of TNT formation in neuronal cells. J. Cell Sci. 2013, 126 Pt 19, 4424–4435.
  29. Uhl, J.; Gujarathi, S.; Waheed, A.A.; Gordon, A.; Freed, E.O.; Gousset, K. Myosin-X is essential to the intercellular spread of HIV-1 Nef through tunneling nanotubes. J. Cell Commun. Signal. 2019, 13, 209–224.
  30. Delage, E.; Cervantes, D.C.; Penard, 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.
  31. Zhu, H.; Xue, C.; Xu, X.; Guo, Y.; Li, X.; Lu, J.; Ju, S.; Wang, Y.; Cao, Z.; Gu, X. Rab8a/Rab11a regulate intercellular communications between neural cells via tunneling nanotubes. Cell Death Dis. 2016, 7, e2523.
  32. Zhu, S.; Bhat, S.; Syan, S.; Kuchitsu, Y.; Fukuda, M.; Zurzolo, C. Rab11a-Rab8a cascade regulates the formation of tunneling nanotubes through vesicle recycling. J. Cell Sci. 2018, 131.
  33. Andresen, V.; Wang, X.; Ghimire, S.; Omsland, M.; Gjertsen, B.T.; Gerdes, H.H. Tunneling nanotube (TNT) formation is independent of p53 expression. Cell Death Differ. 2013, 20, 1124.
  34. Ljubojevic, N.; Henderson, J.M.; Zurzolo, C. The Ways of Actin: Why Tunneling Nanotubes Are Unique Cell Protrusions. Trends Cell Biol. 2021, 31, 130–142.
  35. McKinney, M.C.; Stark, D.A.; Teddy, J.; Kulesa, P.M. Neural Crest Cell Communication Involves an Exchange of Cytoplasmic Material Through Cellular Bridges Revealed by Photoconversion of KikGR. Dev. Dyn. 2011, 240, 1391–1401.
  36. Miyazawa, K.; Emmerling, K.; Manuelidis, L. Proliferative Arrest of Neural Cells Induces Prion Protein Synthesis, Nanotube Formation, and Cell-to-Cell Contacts. J. Cell. Biochem. 2010, 111, 239–247.
  37. Osswald, M.; Jung, E.; Sahm, F.; Solecki, G.; Venkataramani, V.; Blaes, J.; Weil, S.; Horstmann, H.; Wiestler, B.; Syed, M.; et al. Brain tumour cells interconnect to a functional and resistant network. Nature 2015, 528, 93.
  38. Zani, B.G.; Laura, I.; Edelman, E.R.; Edward, M.R. Tubular Bridges for Bronchial Epithelial Cell Migration and Communication. PLoS ONE 2010, 5, e8930.
  39. Guo, L.; Zhang, Y.; Yang, Z.; Peng, H.; Wei, R.; Wang, C.; Feng, M. Tunneling Nanotubular Expressways for Ultrafast and Accurate M1 Macrophage Delivery of Anticancer Drugs to Metastatic Ovarian Carcinoma. ACS Nano 2019, 13, 1078–1096.
  40. Islam, M.N.; Das, S.R.; Emin, M.T.; Wei, M.; Sun, L.; Westphalen, K.; Rowlands, D.J.; Quadri, S.K.; Bhattacharya, S.; Bhattacharya, J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 2012, 18, 759–765.
  41. Spees, J.L.; Olson, S.D.; Whitney, M.J.; Prockop, D.J. Mitochondrial transfer between cells can rescue aerobic respiration. Proc. Natl. Acad. Sci. USA 2006, 103, 1283–1288.
  42. Han, H.; Hu, J.; Yan, Q.; Zhu, J.; Zhu, Z.; Chen, Y.; Sun, J.; Zhang, R. Bone marrow-derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. Mol. Med. Rep. 2016, 13, 1517–1524.
  43. Vallabhaneni, K.C.; Haller, H.; Dumler, I. Vascular smooth muscle cells initiate proliferation of mesenchymal stem cells by mitochondrial transfer via tunneling nanotubes. Stem Cells Dev. 2012, 21, 3104–3113.
  44. Dubey, G.P.; Ben-Yehuda, S. Intercellular nanotubes mediate bacterial communication. Cell 2011, 144, 590–600.
  45. Sowinski, S.; Jolly, C.; Berninghausen, O.; Purbhoo, M.A.; Chauveau, A.; Kohler, K.; Oddos, S.; Eissmann, P.; Brodsky, F.M.; Hopkins, C.; et al. Membrane nanotubes physically connect T cells over long distances presenting a novel route for HIV-1 transmission. Nat. Cell Biol. 2008, 10, 211–219.
  46. Onfelt, B.; Nedvetzki, S.; Benninger, R.K.P.; Purbhoo, M.A.; Sowinski, S.; Hume, A.N.; Seabra, M.C.; Neil, M.A.A.; French, P.M.W.; Davis, D.M. Structurally distinct membrane nanotubes between human macrophages support long-distance vesicular traffic or surfing of bacteria. J. Immunol. 2006, 177, 8476–8483.
  47. Eugenin, E.A.; Gaskill, P.J.; Berman, J.W. Tunneling nanotubes (TNT) are induced by HIV-infection of macrophages: A potential mechanism for intercellular HIV trafficking. Cell. Immunol. 2009, 254, 142–148.
  48. Zhu, D.; Tan, K.S.; Zhang, X.; Sun, A.Y.; Sun, G.Y.; Lee, J.C. Hydrogen peroxide alters membrane and cytoskeleton properties and increases intercellular connections in astrocytes. J. Cell Sci. 2005, 118 Pt 16, 3695–3703.
  49. Souriant, S.; Balboa, L.; Dupont, M.; Pingris, K.; Kviatcovsky, D.; Cougoule, C.; Lastrucci, C.; Bah, A.; Gasser, R.; Poincloux, R.; et al. Tuberculosis Exacerbates HIV-1 Infection through IL-10/STAT3-Dependent Tunneling Nanotube Formation in Macrophages. Cell Rep. 2019, 26, 3586–3599.e7.
  50. Filippova, N.; Nabors, L.B. ELAVL1 Role in Cell Fusion and Tunneling Membrane Nanotube Formations with Implication to Treat Glioma Heterogeneity. Cancers 2020, 12, 3069.
  51. Abounit, S.; Bousset, L.; Loria, F.; Zhu, S.; de Chaumont, F.; Pieri, L.; Olivo-Marin, J.C.; Melki, R.; Zurzolo, C. Tunneling nanotubes spread fibrillar alpha-synuclein by intercellular trafficking of lysosomes. EMBO J. 2016, 35, 2120–2138.
  52. Abounit, S.; Wu, J.W.; Duff, K.; Victoria, G.S.; Zurzolo, C. Tunneling nanotubes: A possible highway in the spreading of tau and other prion-like proteins in neurodegenerative diseases. Prion 2016, 10, 344–351.
  53. Lou, E.; Fujisawa, S.; Morozov, A.; Barlas, A.; Romin, Y.; Dogan, Y.; Gholami, S.; Moreira, A.L.; Manova-Todorova, K.; Moore, M.A. Tunneling nanotubes provide a unique conduit for intercellular transfer of cellular contents in human malignant pleural mesothelioma. PLoS ONE 2012, 7, e33093.
  54. Matejka, N.; Reindl, J. Perspectives of cellular communication through tunneling nanotubes in cancer cells and the connection to radiation effects. Radiat. Oncol. 2019, 14, 218.
  55. Desir, S.; Wong, P.; Turbyville, T.; Chen, D.; Shetty, M.; Clark, C.; Zhai, E.; Romin, Y.; Manova-Todorova, K.; Starr, T.K.; et al. Intercellular Transfer of Oncogenic KRAS via Tunneling Nanotubes Introduces Intracellular Mutational Heterogeneity in Colon Cancer Cells. Cancers 2019, 11, 892.
  56. Marlein, C.R.; Piddock, R.E.; Mistry, J.J.; Zaitseva, L.; Hellmich, C.; Horton, R.H.; Zhou, Z.; Auger, M.J.; Bowles, K.M.; Rushworth, S.A. CD38-Driven Mitochondrial Trafficking Promotes Bioenergetic Plasticity in Multiple Myeloma. Cancer Res. 2019, 79, 2285–2297.
  57. Asencio-Barría, C.; Defamie, N.; Sáez, J.C.; Mesnil, M.; Godoy, A.S. Direct Intercellular Communications and Cancer: A Snapshot of the Biological Roles of Connexins in Prostate Cancer. Cancers 2019, 11, 1370.
  58. Kretschmer, A.; Zhang, F.; Somasekharan, S.P.; Tse, C.; Leachman, L.; Gleave, A.; Li, B.; Asmaro, I.; Huang, T.; Kotula, L.; et al. Stress-induced tunneling nanotubes support treatment adaptation in prostate cancer. Sci. Rep. 2019, 9, 7826.
  59. Thayanithy, V.; Babatunde, V.; Dickson, E.L.; Wong, P.; Oh, S.; Ke, X.; Barlas, A.; Fujisawa, S.; Romin, Y.; Moreira, A.L.; et al. Tumor exosomes induce tunneling nanotubes in lipid raft-enriched regions of human mesothelioma cells. Exp. Cell Res. 2014, 323, 178–188.
More
Information
Subjects: Cell Biology; Pathology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 597
Revisions: 2 times (View History)
Update Date: 10 Mar 2021
1000/1000
Video Production Service