51
|
Resnik N, Baraga D, Glažar P, Jokhadar Zemljič Š, Derganc J, Sepčić K, Veranič P, Kreft ME. Molecular, morphological and functional properties of tunnelling nanotubes between normal and cancer urothelial cells: New insights from the in vitro model mimicking the situation after surgical removal of the urothelial tumor. Front Cell Dev Biol 2022; 10:934684. [PMID: 36601539 PMCID: PMC9806176 DOI: 10.3389/fcell.2022.934684] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Tunnelling nanotubes (TNTs) are membranous connections that represent a unique type of intercellular communication in different cell types. They are associated with cell physiology and cancer pathology. The possible existence of tunnelling nanotubes communication between urothelial cancer and normal cells has not yet been elucidated. Therefore, we analyzed TNTs formed by T24 cells (human invasive cancer urothelial cells) and normal porcine urothelial (NPU) cells, which serve as surrogate models for healthy human urothelial cells. Monocultures and cocultures of NPU and T24 cells were established and analyzed using live-cell imaging, optical tweezers, fluorescence microscopy, and scanning electron microscopy. TNTs of NPU cells differed significantly from tunnelling nanotubes of T24 cells in number, length, diameter, lipid composition, and elastic properties. Membrane domains enriched in cholesterol/sphingomyelin were present in tunnelling nanotubes of T24 cells but not in NPU cells. The tunnelling nanotubes in T24 cells were also easier to bend than the tunnelling nanotubes in NPU cells. The tunnelling nanotubes of both cell types were predominantly tricytoskeletal, and contained actin filaments, intermediate filaments, and microtubules, as well as the motor proteins myosin Va, dynein, and kinesin 5B. Mitochondria were transported within tunnelling nanotubes in living cells, and were colocalized with microtubules and the microtubule-associated protein dynamin 2. In cocultures, heterocellular tunnelling nanotubes were formed between NPU cells and T24 cells and vice versa. The presence of connexin 43 at the end of urothelial tunnelling nanotubes suggests a junctional connection and the involvement of tunnelling nanotube in signal transduction. In this study, we established a novel urothelial cancer-normal coculture model and showed cells in the minority tend to form tunnelling nanotubes with cells in the majority. The condition with cancer cells in the minority is an attractive model to mimic the situation after surgical resection with remaining cancer cells and may help to understand cancer progression and recurrence. Our results shed light on the biological activity of tunnelling nanotubes and have the potential to advance the search for anticancer drugs that target tunnelling nanotubes.
Collapse
Affiliation(s)
- Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Diana Baraga
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Polona Glažar
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Špela Jokhadar Zemljič
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Derganc
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,*Correspondence: Mateja Erdani Kreft,
| |
Collapse
|
52
|
Tarasiuk O, Scuteri A. Role of Tunneling Nanotubes in the Nervous System. Int J Mol Sci 2022; 23:12545. [PMID: 36293396 PMCID: PMC9604327 DOI: 10.3390/ijms232012545] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/20/2022] Open
Abstract
Cellular communication and the transfer of information from one cell to another is crucial for cell viability and homeostasis. During the last decade, tunneling nanotubes (TNTs) have attracted scientific attention, not only as a means of direct intercellular communication, but also as a possible system to transport biological cargo between distant cells. Peculiar TNT characteristics make them both able to increase cellular survival capacities, as well as a potential target of neurodegenerative disease progression. Despite TNT formation having been documented in a number of cell types, the exact mechanisms triggering their formation are still not completely known. In this review, we will summarize and highlight those studies focusing on TNT formation in the nervous system, as well as their role in neurodegenerative diseases. Moreover, we aim to stress some possible mechanisms and important proteins probably involved in TNT formation in the nervous system.
Collapse
Affiliation(s)
| | - Arianna Scuteri
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
53
|
Tozzi A, Mariniello L. Unusual Mathematical Approaches Untangle Nervous Dynamics. Biomedicines 2022; 10:biomedicines10102581. [PMID: 36289843 PMCID: PMC9599563 DOI: 10.3390/biomedicines10102581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
The massive amount of available neurodata suggests the existence of a mathematical backbone underlying neuronal oscillatory activities. For example, geometric constraints are powerful enough to define cellular distribution and drive the embryonal development of the central nervous system. We aim to elucidate whether underrated notions from geometry, topology, group theory and category theory can assess neuronal issues and provide experimentally testable hypotheses. The Monge’s theorem might contribute to our visual ability of depth perception and the brain connectome can be tackled in terms of tunnelling nanotubes. The multisynaptic ascending fibers connecting the peripheral receptors to the neocortical areas can be assessed in terms of knot theory/braid groups. Presheaves from category theory permit the tackling of nervous phase spaces in terms of the theory of infinity categories, highlighting an approach based on equivalence rather than equality. Further, the physical concepts of soft-matter polymers and nematic colloids might shed new light on neurulation in mammalian embryos. Hidden, unexpected multidisciplinary relationships can be found when mathematics copes with neural phenomena, leading to novel answers for everlasting neuroscientific questions. For instance, our framework leads to the conjecture that the development of the nervous system might be correlated with the occurrence of local thermal changes in embryo–fetal tissues.
Collapse
Affiliation(s)
- Arturo Tozzi
- Center for Nonlinear Science, University of North Texas, Denton, TX 76203-5017, USA
- Correspondence:
| | - Lucio Mariniello
- Department of Pediatrics, University Federico II, 80131 Naples, Italy
| |
Collapse
|
54
|
Huang L, Zhang J, Wu Z, Zhou L, Yu B, Jing Y, Lin D, Qu J. Revealing the structure and organization of intercellular tunneling nanotubes (TNTs) by STORM imaging. NANOSCALE ADVANCES 2022; 4:4258-4262. [PMID: 36321151 PMCID: PMC9552758 DOI: 10.1039/d2na00415a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/08/2022] [Indexed: 06/16/2023]
Abstract
Tunneling nanotubes (TNTs) are nanoscale, actin-rich, transient intercellular tubes for cell-to-cell communication, which transport various cargoes between distant cells. The structural complexity and spatial organization of the involved components of TNTs remain unknown. In this work, the STORM super-resolution imaging technique was applied to elucidate the structural organization of microfilaments and microtubules in intercellular TNTs at the nanometer scale. Our results reveal different distributions of microfilaments and intertwined structures of microtubules in TNTs, which promote the knowledge of TNT communications.
Collapse
Affiliation(s)
- Lilin Huang
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Jiao Zhang
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Zekai Wu
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Liangliang Zhou
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Bin Yu
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Yingying Jing
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Danying Lin
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| | - Junle Qu
- Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University Shenzhen 518060 P. R. China
| |
Collapse
|
55
|
Intercellular communication in the tumour microecosystem: Mediators and therapeutic approaches for hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166528. [PMID: 36007784 DOI: 10.1016/j.bbadis.2022.166528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
Hepatocellular carcinoma (HCC), one of the most common tumours worldwide, is one of the main causes of mortality in cancer patients. There are still numerous problems hindering its early diagnosis, which lead to late patients receiving treatment, and these problems need to be solved urgently. The tumour microecosystem is a complex network system comprising seven parts: the hypoxia niche, immune microenvironment, metabolic microenvironment, acidic niche, innervated niche, mechanical microenvironment, and microbial microenvironment. Intercellular communication is divided into direct contact and indirect communication. Direct contact communication includes gap junctions, tunneling nanotubes, and receptor-ligand interactions, whereas indirect communication includes exosomes, apoptotic vesicles, and soluble factors. Mechanical communication and cytoplasmic exchange are further means of intercellular communication. Intercellular communication mediates the crosstalk between the tumour microecosystem and the host as well as that between cells and cell-free components in the tumour microecosystem, causing changes in the tumour hallmarks of the HCC microecosystem such as changes in tumour proliferation, invasion, apoptosis, angiogenesis, metastasis, inflammatory response, gene mutation, immune escape, metabolic reprogramming, and therapeutic resistance. Here, we review the role of the above-mentioned intercellular communication in the HCC microecosystem and discuss the advantages of targeted intercellular communication in the clinical diagnosis and treatment of HCC. Finally, the current problems and prospects are discussed.
Collapse
|
56
|
Ritter A, Kreis NN, Hoock SC, Solbach C, Louwen F, Yuan J. Adipose Tissue-Derived Mesenchymal Stromal/Stem Cells, Obesity and the Tumor Microenvironment of Breast Cancer. Cancers (Basel) 2022; 14:3908. [PMID: 36010901 PMCID: PMC9405791 DOI: 10.3390/cancers14163908] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most frequently diagnosed cancer and a common cause of cancer-related death in women. It is well recognized that obesity is associated with an enhanced risk of more aggressive breast cancer as well as reduced patient survival. Adipose tissue is the major microenvironment of breast cancer. Obesity changes the composition, structure, and function of adipose tissue, which is associated with inflammation and metabolic dysfunction. Interestingly, adipose tissue is rich in ASCs/MSCs, and obesity alters the properties and functions of these cells. As a key component of the mammary stroma, ASCs play essential roles in the breast cancer microenvironment. The crosstalk between ASCs and breast cancer cells is multilateral and can occur both directly through cell-cell contact and indirectly via the secretome released by ASC/MSC, which is considered to be the main effector of their supportive, angiogenic, and immunomodulatory functions. In this narrative review, we aim to address the impact of obesity on ASCs/MSCs, summarize the current knowledge regarding the potential pathological roles of ASCs/MSCs in the development of breast cancer, discuss related molecular mechanisms, underline the possible clinical significance, and highlight related research perspectives. In particular, we underscore the roles of ASCs/MSCs in breast cancer cell progression, including proliferation and survival, angiogenesis, migration and invasion, the epithelial-mesenchymal transition, cancer stem cell development, immune evasion, therapy resistance, and the potential impact of breast cancer cells on ASCS/MSCs by educating them to become cancer-associated fibroblasts. We conclude that ASCs/MSCs, especially obese ASCs/MSCs, may be key players in the breast cancer microenvironment. Targeting these cells may provide a new path of effective breast cancer treatment.
Collapse
Affiliation(s)
- Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| | | | | | | | | | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| |
Collapse
|
57
|
Luchetti F, Carloni S, Nasoni MG, Reiter RJ, Balduini W. Tunneling nanotubes and mesenchymal stem cells: New insights into the role of melatonin in neuronal recovery. J Pineal Res 2022; 73:e12800. [PMID: 35419879 PMCID: PMC9540876 DOI: 10.1111/jpi.12800] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 11/28/2022]
Abstract
Efficient cell-to-cell communication is essential for tissue development, homeostasis, and the maintenance of cellular functions after injury. Tunneling nanotubes (TNTs) have emerged as a new important method of cell-to-cell communication. TNTs are primarily established between stressed and unstressed cells and can transport a variety of cellular components. Mitochondria are important trafficked entities through TNTs. Transcellular mitochondria transfer permits the incorporation of healthy mitochondria into the endogenous network of recipient cells, changing the bioenergetic profile and other functional properties of the recipient and may allow the recipient cells to recuperate from apoptotic processes and return to a normal operating state. Mesenchymal cells (MSCs) can form TNTs and transfer mitochondria and other constituents to target cells. This occurs under both physiological and pathological conditions, leading to changes in cellular energy metabolism and functions. This review summarizes the newly described capacity of melatonin to improve mitochondrial fusion/fission dynamics and promote TNT formation. This new evidence suggests that melatonin's protective effects could be attributed to its ability to prevent mitochondrial damage in injured cells, reduce senescence, and promote anastasis, a natural cell recovery phenomenon that rescues cells from the brink of death. The modulation of these new routes of intercellular communication by melatonin could play a key role in increasing the therapeutic potential of MSCs.
Collapse
Affiliation(s)
- Francesca Luchetti
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Silvia Carloni
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Maria G. Nasoni
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Russel J. Reiter
- Department of Cell Systems and AnatomyLong School of Medicine, UT HealthSan AntonioTexasUSA
| | - Walter Balduini
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| |
Collapse
|
58
|
Pepe A, Pietropaoli S, Vos M, Barba-Spaeth G, Zurzolo C. Tunneling nanotubes provide a route for SARS-CoV-2 spreading. SCIENCE ADVANCES 2022; 8:eabo0171. [PMID: 35857849 PMCID: PMC9299553 DOI: 10.1126/sciadv.abo0171] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/08/2022] [Indexed: 05/10/2023]
Abstract
Neurological manifestations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection represent a major issue in long coronavirus disease. How SARS-CoV-2 gains access to the brain and how infection leads to neurological symptoms are not clear because the principal means of viral entry by endocytosis, the angiotensin-converting enzyme 2 receptor, are barely detectable in the brain. We report that human neuronal cells, nonpermissive to infection through the endocytic pathway, can be infected when cocultured with permissive infected epithelial cells. SARS-CoV-2 induces the formation of tunneling nanotubes (TNTs) and exploits this route to spread to uninfected cells. In cellulo correlative fluorescence and cryo-electron tomography reveal that SARS-CoV-2 is associated with TNTs between permissive cells. Furthermore, multiple vesicular structures such as double-membrane vesicles, sites of viral replication, are observed inside TNTs between permissive and nonpermissive cells. Our data highlight a previously unknown mechanism of SARS-CoV-2 spreading, likely used as a route to invade nonpermissive cells and potentiate infection in permissive cells.
Collapse
Affiliation(s)
- Anna Pepe
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, CNRS UMR3691, Université Paris Cité, 75015 Paris, France
| | - Stefano Pietropaoli
- Unité de Virologie Structurale, CNRS UMR 3569 Département de Virologie, Institut Pasteur 28 rue du Docteur Roux, Université Paris Cité, 75015 Paris, France
- Catalent Pharma Solutions, Strada Provinciale 12 Casilina, 41, 03012 Anagni, Frosinone, Italy
| | - Matthijn Vos
- Plateforme Technologique Nanoimagerie Institut Pasteur, 25 rue du Docteur Roux, 75015 Paris, France
| | - Giovanna Barba-Spaeth
- Unité de Virologie Structurale, CNRS UMR 3569 Département de Virologie, Institut Pasteur 28 rue du Docteur Roux, Université Paris Cité, 75015 Paris, France
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, CNRS UMR3691, Université Paris Cité, 75015 Paris, France
| |
Collapse
|
59
|
Role of pericytes in blood-brain barrier preservation during ischemia through tunneling nanotubes. Cell Death Dis 2022; 13:582. [PMID: 35790716 PMCID: PMC9256725 DOI: 10.1038/s41419-022-05025-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/07/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
Crosstalk mechanisms between pericytes, endothelial cells, and astrocytes preserve integrity and function of the blood-brain-barrier (BBB) under physiological conditions. Long intercellular channels allowing the transfer of small molecules and organelles between distant cells called tunneling nanotubes (TNT) represent a potential substrate for energy and matter exchanges between the tripartite cellular compartments of the BBB. However, the role of TNT across BBB cells under physiological conditions and in the course of BBB dysfunction is unknown. In this work, we analyzed the TNT's role in the functional dialog between human brain endothelial cells, and brain pericytes co-cultured with human astrocytes under normal conditions or after exposure to ischemia/reperfusion, a condition in which BBB breakdown occurs, and pericytes participate in the BBB repair. Using live time-lapse fluorescence microscopy and laser-scanning confocal microscopy, we found that astrocytes form long TNT with pericytes and endothelial cells and receive functional mitochondria from both cell types through this mechanism. The mitochondrial transfer also occurred in multicellular assembloids of human BBB that reproduce the three-dimensional architecture of the BBB. Under conditions of ischemia/reperfusion, TNT formation is upregulated, and astrocytes exposed to oxygen-glucose deprivation were rescued from apoptosis by healthy pericytes through TNT-mediated transfer of functional mitochondria, an effect that was virtually abolished in the presence of TNT-destroying drugs. The results establish a functional role of TNT in the crosstalk between BBB cells and demonstrate that TNT-mediated mitochondrial transfer from pericytes rescues astrocytes from ischemia/reperfusion-induced apoptosis. Our data confirm that the pericytes might play a pivotal role in preserving the structural and functional integrity of BBB under physiological conditions and participate in BBB repair in brain diseases.
Collapse
|
60
|
Cruz JVR, Batista C, Afonso BDH, Alexandre-Moreira MS, Dubois LG, Pontes B, Moura Neto V, Mendes FDA. Obstacles to Glioblastoma Treatment Two Decades after Temozolomide. Cancers (Basel) 2022; 14:cancers14133203. [PMID: 35804976 PMCID: PMC9265128 DOI: 10.3390/cancers14133203] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Glioblastomas are the most common and aggressive brain tumors in adults, with a median survival of 15 months. Treatment is surgical removal, followed by chemotherapy and/or radiotherapy. Current chemotherapeutics do not kill all the tumor cells and some cells survive, leading to the appearance of a new tumor resistant to the treatment. These treatment-resistant cells are called tumor stem cells. In addition, glioblastoma cells have a high capacity for migration, forming new tumors in areas distant from the original tumor. Studies are now focused on understanding the molecular mechanisms of chemoresistance and controlling drug entry into the brain to improve drug performance. Another promising therapeutic approach is the use of viruses that specifically destroy glioblastoma cells, preserving the neural tissue around the tumor. In this review, we summarize the main biological features of glioblastoma and the therapeutic targets that are currently under study for new clinical trials. Abstract Glioblastomas are considered the most common and aggressive primary brain tumor in adults, with an average of 15 months’ survival rate. The treatment is surgery resection, followed by chemotherapy with temozolomide, and/or radiotherapy. Glioblastoma must have wild-type IDH gene and some characteristics, such as TERT promoter mutation, EGFR gene amplification, microvascular proliferation, among others. Glioblastomas have great heterogeneity at cellular and molecular levels, presenting distinct phenotypes and diversified molecular signatures in each tumor mass, making it difficult to define a specific therapeutic target. It is believed that the main responsibility for the emerge of these distinct patterns lies in subcellular populations of tumor stem cells, capable of tumor initiation and asymmetric division. Studies are now focused on understanding molecular mechanisms of chemoresistance, the tumor microenvironment, due to hypoxic and necrotic areas, cytoskeleton and extracellular matrix remodeling, and in controlling blood brain barrier permeabilization to improve drug delivery. Another promising therapeutic approach is the use of oncolytic viruses that are able to destroy specifically glioblastoma cells, preserving the neural tissue around the tumor. In this review, we summarize the main biological characteristics of glioblastoma and the cutting-edge therapeutic targets that are currently under study for promising new clinical trials.
Collapse
Affiliation(s)
- João Victor Roza Cruz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
| | - Carolina Batista
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
| | - Bernardo de Holanda Afonso
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
- Instituto Estadual do Cérebro Paulo Niemeyer, Rua do Rezende 156, Rio de Janeiro 20231-092, Brazil
| | - Magna Suzana Alexandre-Moreira
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Campus A.C. Simões, Avenida Lourival Melo Mota, Maceio 57072-970, Brazil;
| | - Luiz Gustavo Dubois
- UFRJ Campus Duque de Caxias Professor Geraldo Cidade, Rodovia Washington Luiz, n. 19.593, km 104.5, Santa Cruz da Serra, Duque de Caxias 25240-005, Brazil;
| | - Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
| | - Vivaldo Moura Neto
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
- Instituto Estadual do Cérebro Paulo Niemeyer, Rua do Rezende 156, Rio de Janeiro 20231-092, Brazil
| | - Fabio de Almeida Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
- Correspondence:
| |
Collapse
|
61
|
Eugenin E, Camporesi E, Peracchia C. Direct Cell-Cell Communication via Membrane Pores, Gap Junction Channels, and Tunneling Nanotubes: Medical Relevance of Mitochondrial Exchange. Int J Mol Sci 2022; 23:6133. [PMID: 35682809 PMCID: PMC9181466 DOI: 10.3390/ijms23116133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/28/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023] Open
Abstract
The history of direct cell-cell communication has evolved in several small steps. First discovered in the 1930s in invertebrate nervous systems, it was thought at first to be an exception to the "cell theory", restricted to invertebrates. Surprisingly, however, in the 1950s, electrical cell-cell communication was also reported in vertebrates. Once more, it was thought to be an exception restricted to excitable cells. In contrast, in the mid-1960s, two startling publications proved that virtually all cells freely exchange small neutral and charged molecules. Soon after, cell-cell communication by gap junction channels was reported. While gap junctions are the major means of cell-cell communication, in the early 1980s, evidence surfaced that some cells might also communicate via membrane pores. Questions were raised about the possible artifactual nature of the pores. However, early in this century, we learned that communication via membrane pores exists and plays a major role in medicine, as the structures involved, "tunneling nanotubes", can rescue diseased cells by directly transferring healthy mitochondria into compromised cells and tissues. On the other hand, pathogens/cancer could also use these communication systems to amplify pathogenesis. Here, we describe the evolution of the discovery of these new communication systems and the potential therapeutic impact on several uncurable diseases.
Collapse
Affiliation(s)
- Eliseo Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), 105 11th Street, Galveston, TX 77555, USA
| | - Enrico Camporesi
- Department of Surgery and TEAM Health Anesthesia, University of South Florida, 2 Tampa General Circle, Tampa, FL 33606, USA;
| | - Camillo Peracchia
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA;
| |
Collapse
|
62
|
Kit O, Frantsiyants E, Neskubina I, Shikhlyarova A, Kaplieva I. Mitochondrial therapy: a vision of the outlooks for treatment of main twenty-first-century diseases. CARDIOMETRY 2022. [DOI: 10.18137/cardiometry.2022.22.1827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are dynamic organelles which constantly change their shape, size, and location within the cells. Mitochondrial dynamics is associated with mesenchymal metabolism or epithelial-mesenchymal transition to regulate the stem cell differentiation, proliferation, migration, and apoptosis. The transfer of mitochondria from one cell to another is necessary to improve and maintain homeostasis in an organism. Mitochondrial transplantation is a therapeutic approach that involves an introduction of healthy mitochondria into damaged organs. Recent evidence data have shown that the physiological properties of healthy mitochondria provide their ability to replace damaged mitochondria, with suggesting that replacing damaged mitochondria with healthy mitochondria may protect cells from further damage. Moreover, mitochondria can also be actively released into the extracellular space and potentially be transferred between the cells in the central nervous system. This increased interest in mitochondrial therapy calls for a deeper understanding of the mechanisms, which build the basis for mitochondrial transfer, uptake, and cellular defense. In this review, questions related to the involvement of mitochondria in the pathogenesis of cancer will be discussed. Particular attention will be paid to mitochondrial transplantation as a therapeutic approach to treat the mitochondrial dysfunction under some pathological conditions.
Collapse
|
63
|
Couteaudier M, Montange T, Njouom R, Bilounga-Ndongo C, Gessain A, Buseyne F. Plasma antibodies from humans infected with zoonotic simian foamy virus do not inhibit cell-to-cell transmission of the virus despite binding to the surface of infected cells. PLoS Pathog 2022; 18:e1010470. [PMID: 35605011 PMCID: PMC9166401 DOI: 10.1371/journal.ppat.1010470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/03/2022] [Accepted: 03/25/2022] [Indexed: 01/23/2023] Open
Abstract
Zoonotic simian foamy viruses (SFV) establish lifelong infection in their human hosts. Despite repeated transmission of SFV from nonhuman primates to humans, neither transmission between human hosts nor severe clinical manifestations have been reported. We aim to study the immune responses elicited by chronic infection with this retrovirus and previously reported that SFV-infected individuals generate potent neutralizing antibodies that block cell infection by viral particles. Here, we assessed whether human plasma antibodies block SFV cell-to-cell transmission and present the first description of cell-to-cell spreading of zoonotic gorilla SFV. We set-up a microtitration assay to quantify the ability of plasma samples from 20 Central African individuals infected with gorilla SFV and 9 uninfected controls to block cell-associated transmission of zoonotic gorilla SFV strains. We used flow-based cell cytometry and fluorescence microscopy to study envelope protein (Env) localization and the capacity of plasma antibodies to bind to infected cells. We visualized the cell-to-cell spread of SFV by real-time live imaging of a GFP-expressing prototype foamy virus (CI-PFV) strain. None of the samples neutralized cell-associated SFV infection, despite the inhibition of cell-free virus. We detected gorilla SFV Env in the perinuclear region, cytoplasmic vesicles and at the cell surface. We found that plasma antibodies bind to Env located at the surface of cells infected with primary gorilla SFV strains. Extracellular labeling of SFV proteins by human plasma samples showed patchy staining at the base of the cell and dense continuous staining at the cell apex, as well as staining in the intercellular connections that formed when previously connected cells separated from each other. In conclusion, SFV-specific antibodies from infected humans do not block cell-to-cell transmission, at least in vitro, despite their capacity to bind to the surface of infected cells. Trial registration: Clinical trial registration: www.clinicaltrials.gov, https://clinicaltrials.gov/ct2/show/NCT03225794/. Foamy viruses are the oldest known retroviruses and have been mostly described to be nonpathogenic in their natural animal hosts. Simian foamy viruses (SFVs) can be transmitted to humans, in whom they establish persistent infection, as have the simian viruses that led to the emergence of two major human pathogens, human immunodeficiency virus type 1 (HIV-1) and human T lymphotropic virus type 1 (HTLV-1). Such cross-species transmission of SFV is ongoing in many parts of the world where humans have contact with nonhuman primates. We previously showed high titers of neutralizing antibodies in the plasma of most SFV-infected individuals. These antiviral antibodies can inhibit cell-free virus entry. However, SFV efficiently spread from one cell to another. Here, we demonstrate that plasma antibodies do not block such cell-to-cell transmission, despite their capacity to bind to the surface of infected cells. In addition, we document for the first time the cell-to-cell spread of primary zoonotic gorilla SFV.
Collapse
Affiliation(s)
- Mathilde Couteaudier
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Unité d’Epidémiologie et Physiopathologie des Virus Oncogènes, Paris, France
| | - Thomas Montange
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Unité d’Epidémiologie et Physiopathologie des Virus Oncogènes, Paris, France
| | | | | | - Antoine Gessain
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Unité d’Epidémiologie et Physiopathologie des Virus Oncogènes, Paris, France
| | - Florence Buseyne
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Unité d’Epidémiologie et Physiopathologie des Virus Oncogènes, Paris, France
- * E-mail:
| |
Collapse
|
64
|
Driscoll J, Gondaliya P, Patel T. Tunneling Nanotube-Mediated Communication: A Mechanism of Intercellular Nucleic Acid Transfer. Int J Mol Sci 2022; 23:5487. [PMID: 35628298 PMCID: PMC9143920 DOI: 10.3390/ijms23105487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 12/19/2022] Open
Abstract
Tunneling nanotubes (TNTs) are thin, F-actin-based membranous protrusions that connect distant cells and can provide e a novel mechanism for intercellular communication. By establishing cytoplasmic continuity between interconnected cells, TNTs enable the bidirectional transfer of nuclear and cytoplasmic cargo, including organelles, nucleic acids, drugs, and pathogenic molecules. TNT-mediated nucleic acid transfer provides a unique opportunity for donor cells to directly alter the genome, transcriptome, and metabolome of recipient cells. TNTs have been reported to transport DNA, mitochondrial DNA, mRNA, viral RNA, and non-coding RNAs, such as miRNA and siRNA. This mechanism of transfer is observed in physiological as well as pathological conditions, and has been implicated in the progression of disease. Herein, we provide a concise overview of TNTs' structure, mechanisms of biogenesis, and the functional effects of TNT-mediated intercellular transfer of nucleic acid cargo. Furthermore, we highlight the potential translational applications of TNT-mediated nucleic acid transfer in cancer, immunity, and neurological diseases.
Collapse
Affiliation(s)
| | | | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (J.D.); (P.G.)
| |
Collapse
|
65
|
Reiter RJ, Sharma R, Rosales-Corral S, de Campos Zuccari DAP, de Almeida Chuffa LG. Melatonin: A mitochondrial resident with a diverse skill set. Life Sci 2022; 301:120612. [PMID: 35523285 DOI: 10.1016/j.lfs.2022.120612] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/25/2022] [Accepted: 04/30/2022] [Indexed: 12/12/2022]
Abstract
Melatonin is an ancient molecule that originated in bacteria. When these prokaryotes were phagocytized by early eukaryotes, they eventually developed into mitochondria and chloroplasts. These new organelles retained the melatonin synthetic capacity of their forerunners such that all present-day animal and plant cells may produce melatonin in their mitochondria and chloroplasts. Melatonin concentrations are higher in mitochondria than in other subcellular compartments. Isolated mouse oocyte mitochondria form melatonin when they are incubated with serotonin, a necessary precursor. Oocyte mitochondria subsequently give rise to these organelles in all adult vertebrate cells where they continue to synthesize melatonin. The enzymes that convert serotonin to melatonin, i.e., arylalkylamine-N-acetyltransferase (AANAT) and acetylserotonin-O-methyltransferase, have been identified in brain mitochondria which, when incubated with serotonin, also form melatonin. Melatonin is a potent antioxidant and anti-cancer agent and is optimally positioned in mitochondria to aid in the maintenance of oxidative homeostasis and to reduce cancer cell transformation. Melatonin stimulates the transfer of mitochondria from healthy cells to damaged cells via tunneling nanotubes. Melatonin also regulates the major NAD+-dependent deacetylase, sirtuin 3, in the mitochondria. Disruptions of mitochondrial melatonin synthesis may contribute to a number of mitochondria-related diseases, as discussed in this review.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Sergio Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco CP45150, Mexico
| | | | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP-São Paulo State University, Botucatu, São Paulo 18618-689, Brazil
| |
Collapse
|
66
|
Tunneling Nanotubes between Cells Migrating in ECM Mimicking Fibrous Environments. Cancers (Basel) 2022; 14:cancers14081989. [PMID: 35454893 PMCID: PMC9030013 DOI: 10.3390/cancers14081989] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Tumor cells grow, spread, and invade in a three-dimensional manner, but most experimental approaches in cancer cell biology focus on the behavior of cells in two-dimensional spaces. Patterns of cell invasion and spread in 2D may not accurately depict cell behavior in 3D tumors. We cultivated and investigated malignant mesothelioma cells on a 3D scaffold composed of nanofibers positioned in parallel and crosshatched network configurations to overcome this barrier. We focused on studying long extensions called tunneling nanotubes (TNTs) protruding from cells and connecting with other cells, which have been shown to transmit signals from cell to cell and extend into the tumor microenvironments in a 3D manner. This manuscript describes the biophysics of the formation and function of cancer cell TNTs. These findings will impact the research community by accurately assessing how TNTs affect cancer cell invasion and migration in their natural 3D microenvironment using a novel bioengineered platform. Abstract Tunneling nanotubes (TNTs) comprise a unique class of actin-rich nanoscale membranous protrusions. They enable long-distance intercellular communication and may play an integral role in tumor formation, progression, and drug resistance. TNTs are three-dimensional, but nearly all studies have investigated them using two-dimensional cell culture models. Here, we applied a unique 3D culture platform consisting of crosshatched and aligned fibers to fabricate synthetic suspended scaffolds that mimic the native fibrillar architecture of tumoral extracellular matrix (ECM) to characterize TNT formation and function in its native state. TNTs are upregulated in malignant mesothelioma; we used this model to analyze the biophysical properties of TNTs in this 3D setting, including cell migration in relation to TNT dynamics, rate of TNT-mediated intercellular transport of cargo, and conformation of TNT-forming cells. We found that highly migratory elongated cells on aligned fibers formed significantly longer but fewer TNTs than uniformly spread cells on crossing fibers. We developed new quantitative metrics for the classification of TNT morphologies based on shape and cytoskeletal content using confocal microscopy. In sum, our strategy for culturing cells in ECM-mimicking bioengineered scaffolds provides a new approach for accurate biophysical and biologic assessment of TNT formation and structure in native fibrous microenvironments.
Collapse
|
67
|
Yeh YT, Skinner DE, Criado-Hidalgo E, Chen NS, Garcia-De Herreros A, El-Sakkary N, Liu L, Zhang S, Kandasamy A, Chien S, Lasheras JC, del Álamo JC, Caffrey CR. Biomechanical interactions of Schistosoma mansoni eggs with vascular endothelial cells facilitate egg extravasation. PLoS Pathog 2022; 18:e1010309. [PMID: 35316298 PMCID: PMC8939816 DOI: 10.1371/journal.ppat.1010309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/26/2022] [Indexed: 12/03/2022] Open
Abstract
The eggs of the parasitic blood fluke, Schistosoma, are the main drivers of the chronic pathologies associated with schistosomiasis, a disease of poverty afflicting approximately 220 million people worldwide. Eggs laid by Schistosoma mansoni in the bloodstream of the host are encapsulated by vascular endothelial cells (VECs), the first step in the migration of the egg from the blood stream into the lumen of the gut and eventual exit from the body. The biomechanics associated with encapsulation and extravasation of the egg are poorly understood. We demonstrate that S. mansoni eggs induce VECs to form two types of membrane extensions during encapsulation; filopodia that probe eggshell surfaces and intercellular nanotubes that presumably facilitate VEC communication. Encapsulation efficiency, the number of filopodia and intercellular nanotubes, and the length of these structures depend on the egg’s vitality and, to a lesser degree, its maturation state. During encapsulation, live eggs induce VEC contractility and membranous structures formation in a Rho/ROCK pathway-dependent manner. Using elastic hydrogels embedded with fluorescent microbeads as substrates to culture VECs, live eggs induce VECs to exert significantly greater contractile forces during encapsulation than dead eggs, which leads to 3D deformations on both the VEC monolayer and the flexible substrate underneath. These significant mechanical deformations cause the VEC monolayer tension to fluctuate with the eventual rupture of VEC junctions, thus facilitating egg transit out of the blood vessel. Overall, our data on the mechanical interplay between host VECs and the schistosome egg improve our understanding of how this parasite manipulates its immediate environment to maintain disease transmission. Schistosomiasis, which infects over 200 million people, is a painful disease of poverty that is caused by inflammatory responses to the Schistosoma blood fluke’s eggs. To continue the parasite’s life cycle, eggs must escape the blood vessels and migrate through tissues of the host to the alimentary canal for exit into the environment. The biomechanical processes that help the immobile eggs to cross the blood vessel’s vascular endothelial cells (VECs) as the first step in this migration are not understood. We found that live but not dead eggs induce VECs to crawl over and encapsulate them. VECs in contact with live eggs make membranous extensions (filopodia) to explore the egg’s surface and also form long intercellular nanotubes to communicate with neighboring cells. VECs stimulate particular (Rho/ROCK) biochemical pathways to increase cell contractility and the forces generated are large enough to eventually break the junctions between cells and allow passage of the eggs into the underlying tissue. Our findings show how schistosome eggs activate and interact with VECs to initiate their escape from the bloodstream.
Collapse
Affiliation(s)
- Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (YTY); (JCdA); (CRC)
| | - Danielle E. Skinner
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Natalie Shee Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Antoni Garcia-De Herreros
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Nelly El-Sakkary
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Lawrence Liu
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Shun Zhang
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Adithan Kandasamy
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle Washington, United States of America
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle Washington, United States of America
- * E-mail: (YTY); (JCdA); (CRC)
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (YTY); (JCdA); (CRC)
| |
Collapse
|
68
|
Merolli A, Kasaei L, Ramasamy S, Kolloli A, Kumar R, Subbian S, Feldman LC. An intra-cytoplasmic route for SARS-CoV-2 transmission unveiled by Helium-ion microscopy. Sci Rep 2022; 12:3794. [PMID: 35260703 PMCID: PMC8904465 DOI: 10.1038/s41598-022-07867-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/23/2022] [Indexed: 12/11/2022] Open
Abstract
SARS-CoV-2 virions enter the host cells by docking their spike glycoproteins to the membrane-bound Angiotensin Converting Enzyme 2. After intracellular assembly, the newly formed virions are released from the infected cells to propagate the infection, using the extra-cytoplasmic ACE2 docking mechanism. However, the molecular events underpinning SARS-CoV-2 transmission between host cells are not fully understood. Here, we report the findings of a scanning Helium-ion microscopy study performed on Vero E6 cells infected with mNeonGreen-expressing SARS-CoV-2. Our data reveal, with unprecedented resolution, the presence of: (1) long tunneling nanotubes that connect two or more host cells over submillimeter distances; (2) large scale multiple cell fusion events (syncytia); and (3) abundant extracellular vesicles of various sizes. Taken together, these ultrastructural features describe a novel intra-cytoplasmic connection among SARS-CoV-2 infected cells that may act as an alternative route of viral transmission, disengaged from the well-known extra-cytoplasmic ACE2 docking mechanism. Such route may explain the elusiveness of SARS-CoV-2 to survive from the immune surveillance of the infected host.
Collapse
Affiliation(s)
- Antonio Merolli
- Department of Physics and Astronomy, School of Arts and Sciences, Rutgers University, 136 Frelinghuysen Road, Piscataway, NJ, 08854, USA. .,Department Physics and Astronomy, Rutgers University, DLS Building, 145 Bevier Road, Room 108, Piscataway, NJ, 08854, USA.
| | - Leila Kasaei
- Department of Physics and Astronomy, School of Arts and Sciences, Rutgers University, 136 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Santhamani Ramasamy
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ, 07103, USA
| | - Afsal Kolloli
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ, 07103, USA
| | - Ranjeet Kumar
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ, 07103, USA
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ, 07103, USA
| | - Leonard C Feldman
- Department of Physics and Astronomy, School of Arts and Sciences, Rutgers University, 136 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
69
|
Mentor S, Fisher D. The Ism between Endothelial Cilia and Endothelial Nanotubules Is an Evolving Concept in the Genesis of the BBB. Int J Mol Sci 2022; 23:ijms23052457. [PMID: 35269595 PMCID: PMC8910322 DOI: 10.3390/ijms23052457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
The blood–brain barrier (BBB) is fundamental in maintaining central nervous system (CNS) homeostasis by regulating the chemical environment of the underlying brain parenchyma. Brain endothelial cells (BECs) constitute the anatomical and functional basis of the BBB. Communication between adjacent BECs is critical for establishing BBB integrity, and knowledge of its nanoscopic landscape will contribute to our understanding of how juxtaposed zones of tight-junction protein interactions between BECs are aligned. The review discusses and critiques types of nanostructures contributing to the process of BBB genesis. We further critically evaluate earlier findings in light of novel high-resolution electron microscopy descriptions of nanoscopic tubules. One such phenotypic structure is BEC cytoplasmic projections, which, early in the literature, is postulated as brain capillary endothelial cilia, and is evaluated and compared to the recently discovered nanotubules (NTs) formed in the paracellular spaces between BECs during barrier-genesis. The review attempts to elucidate a myriad of unique topographical ultrastructures that have been reported to be associated with the development of the BBB, viz., structures ranging from cilia to BEC tunneling nanotubules (TUNTs) and BEC tethering nanotubules (TENTs).
Collapse
Affiliation(s)
- Shireen Mentor
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Bellville, Cape Town 7535, South Africa;
| | - David Fisher
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Bellville, Cape Town 7535, South Africa;
- School of Health Professions, University of Missouri, Columbia, MO 65211, USA
- Correspondence:
| |
Collapse
|
70
|
Specialized Intercellular Communications via Tunnelling Nanotubes in Acute and Chronic Leukemia. Cancers (Basel) 2022; 14:cancers14030659. [PMID: 35158927 PMCID: PMC8833474 DOI: 10.3390/cancers14030659] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Tunneling nanotubes (TNTs) are cytoplasmic channels which regulate the contacts between cells and allow the transfer of several elements, including ions, mitochondria, microvesicles, exosomes, lysosomes, proteins, and microRNAs. Through this transport, TNTs are implicated in different physiological and pathological phenomena, such as immune response, cell proliferation and differentiation, embryogenesis, programmed cell death, and angiogenesis. TNTs can promote cancer progression, transferring substances capable of altering apoptotic dynamics, modifying the metabolism and energy balance, inducing changes in immunosurveillance, or affecting the response to chemotherapy. In this review, we evaluated their influence on hematologic malignancies’ progression and resistance to therapies, focusing on acute and chronic myeloid and acute lymphoid leukemia. Abstract Effectual cell-to-cell communication is essential to the development and differentiation of organisms, the preservation of tissue tasks, and the synchronization of their different physiological actions, but also to the proliferation and metastasis of tumor cells. Tunneling nanotubes (TNTs) are membrane-enclosed tubular connections between cells that carry a multiplicity of cellular loads, such as exosomes, non-coding RNAs, mitochondria, and proteins, and they have been identified as the main participants in healthy and tumoral cell communication. TNTs have been described in numerous tumors in in vitro, ex vivo, and in vivo models favoring the onset and progression of tumors. Tumor cells utilize TNT-like membranous channels to transfer information between themselves or with the tumoral milieu. As a result, tumor cells attain novel capabilities, such as the increased capacity of metastasis, metabolic plasticity, angiogenic aptitude, and chemoresistance, promoting tumor severity. Here, we review the morphological and operational characteristics of TNTs and their influence on hematologic malignancies’ progression and resistance to therapies, focusing on acute and chronic myeloid and acute lymphoid leukemia. Finally, we examine the prospects and challenges for TNTs as a therapeutic approach for hematologic diseases by examining the development of efficient and safe drugs targeting TNTs.
Collapse
|
71
|
Mentor S, Makhathini KB, Fisher D. The Role of Cytoskeletal Proteins in the Formation of a Functional In Vitro Blood-Brain Barrier Model. Int J Mol Sci 2022; 23:ijms23020742. [PMID: 35054928 PMCID: PMC8775705 DOI: 10.3390/ijms23020742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 02/05/2023] Open
Abstract
The brain capillary endothelium is highly regulatory, maintaining the chemical stability of the brain’s microenvironment. The role of cytoskeletal proteins in tethering nanotubules (TENTs) during barrier-genesis was investigated using the established immortalized mouse brain endothelial cell line (bEnd5) as an in vitro blood-brain barrier (BBB) model. The morphology of bEnd5 cells was evaluated using both high-resolution scanning electron microscopy and immunofluorescence to evaluate treatment with depolymerizing agents Cytochalasin D for F-actin filaments and Nocodazole for α-tubulin microtubules. The effects of the depolymerizing agents were investigated on bEnd5 monolayer permeability by measuring the transendothelial electrical resistance (TEER). The data endorsed that during barrier-genesis, F-actin and α-tubulin play a cytoarchitectural role in providing both cell shape dynamics and cytoskeletal structure to TENTs forming across the paracellular space to provide cell-cell engagement. Western blot analysis of the treatments suggested a reduced expression of both proteins, coinciding with a reduction in the rates of cellular proliferation and decreased TEER. The findings endorsed that TENTs provide alignment of the paracellular (PC) spaces and tight junction (TJ) zones to occlude bEnd5 PC spaces. The identification of specific cytoskeletal structures in TENTs endorsed the postulate of their indispensable role in barrier-genesis and the maintenance of regulatory permeability across the BBB.
Collapse
Affiliation(s)
- Shireen Mentor
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa; (S.M.); (K.B.M.)
| | - Khayelihle Brian Makhathini
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa; (S.M.); (K.B.M.)
| | - David Fisher
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa; (S.M.); (K.B.M.)
- School of Health Professions, University of Missouri, Columbia, MO 65211, USA
- Correspondence: ; Tel.: +27-21-959-2185
| |
Collapse
|
72
|
Therapeutic applications of mitochondrial transplantation. Biochimie 2022; 195:1-15. [DOI: 10.1016/j.biochi.2022.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
|
73
|
D’Aloia A, Arrigoni E, Costa B, Berruti G, Martegani E, Sacco E, Ceriani M. RalGPS2 Interacts with Akt and PDK1 Promoting Tunneling Nanotubes Formation in Bladder Cancer and Kidney Cells Microenvironment. Cancers (Basel) 2021; 13:cancers13246330. [PMID: 34944949 PMCID: PMC8699646 DOI: 10.3390/cancers13246330] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Cell-to-cell communication in the tumor microenvironment is a crucial process to orchestrate the different components of the tumoral infrastructure. Among the mechanisms of cellular interplay in cancer cells, tunneling nanotubes (TNTs) are dynamic connections that play an important role. The mechanism of the formation of TNTs among cells and the molecules involved in the process remain to be elucidated. In this study, we analyze several bladder cancer cell lines, representative of tumors at different stages and grades. We demonstrate that TNTs are formed only by mid or high-stage cell lines that show muscle-invasive properties and that they actively transport mitochondria and proteins. The formation of TNTs is triggered by stressful conditions and starts with the assembly of a specific multimolecular complex. In this study, we characterize some of the protein components of the TNTs complex, as they are potential novel molecular targets for future therapies aimed at counteracting tumor progression. Abstract RalGPS2 is a Ras-independent Guanine Nucleotide Exchange Factor for RalA GTPase that is involved in several cellular processes, including cytoskeletal organization. Previously, we demonstrated that RalGPS2 also plays a role in the formation of tunneling nanotubes (TNTs) in bladder cancer 5637 cells. In particular, TNTs are a novel mechanism of cell–cell communication in the tumor microenvironment, playing a central role in cancer progression and metastasis formation. However, the molecular mechanisms involved in TNTs formation still need to be fully elucidated. Here we demonstrate that mid and high-stage bladder cancer cell lines have functional TNTs, which can transfer mitochondria. Moreover, using confocal fluorescence time-lapse microscopy, we show in 5637 cells that TNTs mediate the trafficking of RalA protein and transmembrane MHC class III protein leukocyte-specific transcript 1 (LST1). Furthermore, we show that RalGPS2 is essential for nanotubes generation, and stress conditions boost its expression both in 5637 and HEK293 cell lines. Finally, we prove that RalGPS2 interacts with Akt and PDK1, in addition to LST1 and RalA, leading to the formation of a complex that promotes nanotubes formation. In conclusion, our findings suggest that in the tumor microenvironment, RalGPS2 orchestrates the assembly of multimolecular complexes that drive the formation of TNTs.
Collapse
Affiliation(s)
- Alessia D’Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Edoardo Arrigoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Giovanna Berruti
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy;
| | - Enzo Martegani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milano, Italy
- Correspondence: ; Tel.: +39-0264483544
| |
Collapse
|
74
|
Hirashima S, Ohta K, Togo A, Nakamura KI. 3D Mesoscopic Architecture of a Heterogeneous Cellular Network in the Cementum-Periodontal Ligament-Alveolar Bone Complex. Microscopy (Oxf) 2021; 71:22-33. [PMID: 34850074 DOI: 10.1093/jmicro/dfab051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 11/14/2022] Open
Abstract
Cell-to-cell communication orchestrates various cell and tissue functions. This communication enables cells to form cellular networks with each other through direct contact via intercellular junctions. Because these cellular networks are closely related to tissue and organ functions, elucidating the morphological characteristics of cellular networks could lead to the development of novel therapeutic approaches. The tooth, periodontal ligament (PDL), and alveolar bone form a complex via collagen fibres. Teeth depend on the co-ordinated activity of this complex to maintain their function, with cellular networks in each of its three components. Imaging methods for three-dimensional (3D) mesoscopic architectural analysis include focused ion beam/scanning electron microscopy (FIB/SEM), which is characterised by its ability to select observation points and acquire data from complex tissue after extensive block-face imaging, without the need to prepare numerous ultrathin sections. Previously, we employed FIB/SEM to analyse the 3D mesoscopic architecture of hard tissue including the PDL, which exists between the bone and tooth root. The imaging results showed that the cementum, PDL, and alveolar bone networks are in contact and form a heterogeneous cellular network. This cellular network may orchestrate mechanical loading-induced remodelling of the cementum-PDL-alveolar bone complex as the remodelling of each complex component is coordinated, as exemplified by tooth movement due to orthodontic treatment and tooth dislocation due to occlusal loss. In this review, we summarise and discuss the 3D mesoscopic architecture of cellular networks in the cementum, PDL, and alveolar bone as observed in our recent mesoscopic and morphological studies.
Collapse
Affiliation(s)
- Shingo Hirashima
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, 830-0011, Japan.,Dental and Oral Medical Center, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Keisuke Ohta
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, 830-0011, Japan.,Advanced Imaging Research Center, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Kei-Ichiro Nakamura
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, 830-0011, Japan.,Cognitive and Molecular Research Institute of Brain Diseases, Kurume University School of Medicine, Kurume, 830-0011, Japan
| |
Collapse
|
75
|
Tunneling nanotubes and related structures: molecular mechanisms of formation and function. Biochem J 2021; 478:3977-3998. [PMID: 34813650 DOI: 10.1042/bcj20210077] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 10/12/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022]
Abstract
Tunneling nanotubes (TNTs) are F-actin-based, membrane-enclosed tubular connections between animal cells that transport a variety of cellular cargo. Over the last 15 years since their discovery, TNTs have come to be recognized as key players in normal cell communication and organism development, and are also exploited for the spread of various microbial pathogens and major diseases like cancer and neurodegenerative disorders. TNTs have also been proposed as modalities for disseminating therapeutic drugs between cells. Despite the rapidly expanding and wide-ranging relevance of these structures in both health and disease, there is a glaring dearth of molecular mechanistic knowledge regarding the formation and function of these important but enigmatic structures. A series of fundamental steps are essential for the formation of functional nanotubes. The spatiotemporally controlled and directed modulation of cortical actin dynamics would be required to ensure outward F-actin polymerization. Local plasma membrane deformation to impart negative curvature and membrane addition at a rate commensurate with F-actin polymerization would enable outward TNT elongation. Extrinsic tactic cues, along with cognate intrinsic signaling, would be required to guide and stabilize the elongating TNT towards its intended target, followed by membrane fusion to create a functional TNT. Selected cargoes must be transported between connected cells through the action of molecular motors, before the TNT is retracted or destroyed. This review summarizes the current understanding of the molecular mechanisms regulating these steps, also highlighting areas that deserve future attention.
Collapse
|
76
|
Moreira R, Mendonça LS, Pereira de Almeida L. Extracellular Vesicles Physiological Role and the Particular Case of Disease-Spreading Mechanisms in Polyglutamine Diseases. Int J Mol Sci 2021; 22:ijms222212288. [PMID: 34830171 PMCID: PMC8621536 DOI: 10.3390/ijms222212288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Recent research demonstrated pathological spreading of the disease-causing proteins from one focal point across other brain regions for some neurodegenerative diseases, such as Parkinson's and Alzheimer's disease. Spreading mediated by extracellular vesicles is one of the proposed disease-spreading mechanisms. Extracellular vesicles are cell membrane-derived vesicles, used by cells for cell-to-cell communication and excretion of toxic components. Importantly, extracellular vesicles carrying pathological molecules, when internalized by "healthy" cells, may trigger pathological pathways and, consequently, promote disease spreading to neighboring cells. Polyglutamine diseases are a group of genetic neurodegenerative disorders characterized by the accumulation of mutant misfolded proteins carrying an expanded tract of glutamines, including Huntington's and Machado-Joseph disease. The pathological spread of the misfolded proteins or the corresponding mutant mRNA has been explored. The understanding of the disease-spreading mechanism that plays a key role in the pathology progression of these diseases can result in the development of effective therapeutic approaches to stop disease progression, arresting the spread of the toxic components and disease aggravation. Therefore, the present review's main focus is the disease-spreading mechanisms with emphasis on polyglutamine diseases and the putative role played by extracellular vesicles in this process.
Collapse
Affiliation(s)
- Ricardo Moreira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana S. Mendonça
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Correspondence: (L.S.M.); (L.P.d.A.); Tel.: +351-239-820-190 (L.S.M.)
| | - Luís Pereira de Almeida
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence: (L.S.M.); (L.P.d.A.); Tel.: +351-239-820-190 (L.S.M.)
| |
Collapse
|
77
|
Kato K, Nguyen KT, Decker CW, Silkwood KH, Eck SM, Hernandez JB, Garcia J, Han D. Tunneling nanotube formation promotes survival against 5-fluorouracil in MCF-7 breast cancer cells. FEBS Open Bio 2021; 12:203-210. [PMID: 34738322 PMCID: PMC8727926 DOI: 10.1002/2211-5463.13324] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/28/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Tunneling nanotubes (TNTs) are F-actin-based open-ended tubular extensions that form following stresses, such as nutritional deprivation and oxidative stress. The chemotherapy agent 5-fluorouracil (5-FU) represents a significant stressor to cancer cells and induces thymidine deficiency, a state similar to nutritional deprivation. However, the ability of 5-FU to induce TNT formation in cancer cells and potentially enhance survival has not been explored. In this study, we examined whether 5-FU can induce TNT formation in MCF-7 breast cancer cells. Cytotoxic doses of 5-FU (150-350 μm) were observed to significantly induce TNT formation beginning at 24 h after exposure. TNTs formed following 5-FU treatment probably originated as extensions of gap junctions as MCF-7 cells detach from cell clusters. TNTs act as conduits for exchange of cellular components and we observed mitochondrial exchange through TNTs following 5-FU treatment. 5-FU-induced TNT formation was inhibited by over 80% following treatment with the F-actin-depolymerizing agent, cytochalasin B (cytoB). The inhibition of TNTs by cytoB corresponded with increased 5-FU-induced cytotoxicity by 30-62% starting at 48 h, suggesting TNT formation aides in MCF-7 cell survival against 5-FU. Two other widely used chemotherapy agents, docetaxel and doxorubicin induced TNT formation at much lower levels than 5-FU. Our work suggests that the therapeutic targeting of TNTs may increase 5-FU chemotherapy efficacy and decrease drug resistance in cancer cells, and these findings merits further investigation.
Collapse
Affiliation(s)
- Kaylyn Kato
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - Kim Tho Nguyen
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - Carl W Decker
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - Kai H Silkwood
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - Sydney M Eck
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - Jeniffer B Hernandez
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| | - Jerome Garcia
- Department of Biology, University of LaVerne, CA, USA
| | - Derick Han
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, USA
| |
Collapse
|
78
|
Mitochondria and the Tumour Microenvironment in Blood Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:181-203. [PMID: 34664240 DOI: 10.1007/978-3-030-73119-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The bone marrow (BM) is a complex organ located within the cavities of bones. The main function of the BM is to produce all the blood cells required for a normal healthy blood system. As with any major organ, many diseases can arise from errors in bone marrow function, including non-malignant disorders such as anaemia and malignant disorders such as leukaemias. This article will explore the role of the bone marrow, in normal and diseased haematopoiesis, with an emphasis on the requirement for intercellular mitochondrial transfer in leukaemia.
Collapse
|
79
|
Matkó J, Tóth EA. Membrane nanotubes are ancient machinery for cell-to-cell communication and transport. Their interference with the immune system. Biol Futur 2021; 72:25-36. [PMID: 34554502 PMCID: PMC7869423 DOI: 10.1007/s42977-020-00062-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/21/2020] [Indexed: 12/27/2022]
Abstract
Nanotubular connections between mammalian cell types came into the focus only two decades ago, when “live cell super-resolution imaging” was introduced. Observations of these long-time overlooked structures led to understanding mechanisms of their growth/withdrawal and exploring some key genetic and signaling factors behind their formation. Unbelievable level of multiple supportive collaboration between tumor cells undergoing cytotoxic chemotherapy, cross-feeding” between independent bacterial strains or “cross-dressing” collaboration of immune cells promoting cellular immune response, all via nanotubes, have been explored recently. Key factors and "calling signals" determining the spatial directionality of their growth and their overall in vivo significance, however, still remained debated. Interestingly, prokaryotes, including even ancient archaebacteria, also seem to use such NT connections for intercellular communication. Herein, we will give a brief overview of current knowledge of membrane nanotubes and depict a simple model about their possible “historical role”.
Collapse
Affiliation(s)
- János Matkó
- Department of Immunology, Institute of Biology, Eötvös Loránd University, H-1117 Pázmány Péter sétány 1/C, Budapest, Hungary.
| | - Eszter Angéla Tóth
- ATRC Aurigon Toxicological Research Center, H-2120 Pálya utca 2, Dunakeszi, Hungary
| |
Collapse
|
80
|
miR-329-containing exosomes derived from breast tumor cells suppress VEGF and KDM1A expression in endothelial cells. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
81
|
Montes de Oca Balderas P. Mitochondria-plasma membrane interactions and communication. J Biol Chem 2021; 297:101164. [PMID: 34481840 PMCID: PMC8503596 DOI: 10.1016/j.jbc.2021.101164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 11/28/2022] Open
Abstract
Mitochondria are known as the powerhouses of eukaryotic cells; however, they perform many other functions besides oxidative phosphorylation, including Ca2+ homeostasis, lipid metabolism, antiviral response, and apoptosis. Although other hypotheses exist, mitochondria are generally thought as descendants of an α-proteobacteria that adapted to the intracellular environment within an Asgard archaebacteria, which have been studied for decades as an organelle subdued by the eukaryotic cell. Nevertheless, several early electron microscopy observations hinted that some mitochondria establish specific interactions with certain plasma membrane (PM) domains in mammalian cells. Furthermore, recent findings have documented the direct physical and functional interaction of mitochondria and the PM, the organization of distinct complexes, and their communication through vesicular means. In yeast, some molecular players mediating this interaction have been elucidated, but only a few works have studied this interaction in mammalian cells. In addition, mitochondria can be translocated among cells through tunneling nanotubes or by other mechanisms, and free, intact, functional mitochondria have been reported in the blood plasma. Together, these findings challenge the conception of mitochondria as organelles subdued by the eukaryotic cell. This review discusses the evidence of the mitochondria interaction with the PM that has been long disregarded despite its importance in cell function, pathogenesis, and evolution. It also proposes a scheme of mitochondria–PM interactions with the intent to promote research and knowledge of this emerging pathway that promises to shift the current paradigms of cell biology.
Collapse
Affiliation(s)
- Pavel Montes de Oca Balderas
- Unidad de Neurobiología Dinámica, Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico; Lab. BL-305, Instituto de Fisiología Celular, Department of Cognitive Neuroscience, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
82
|
Ganti K, Han J, Manicassamy B, Lowen AC. Rab11a mediates cell-cell spread and reassortment of influenza A virus genomes via tunneling nanotubes. PLoS Pathog 2021; 17:e1009321. [PMID: 34473799 PMCID: PMC8443049 DOI: 10.1371/journal.ppat.1009321] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 09/15/2021] [Accepted: 08/22/2021] [Indexed: 11/18/2022] Open
Abstract
Influenza A virus [IAV] genomes comprise eight negative strand RNAs packaged into virions in the form of viral ribonucleoproteins [vRNPs]. Rab11a plays a crucial role in the transport of vRNPs from the nucleus to the plasma membrane via microtubules, allowing assembly and virus production. Here, we identify a novel function for Rab11a in the inter-cellular transport of IAV vRNPs using tunneling nanotubes [TNTs]as molecular highways. TNTs are F-Actin rich tubules that link the cytoplasm of nearby cells. In IAV-infected cells, Rab11a was visualized together with vRNPs in these actin-rich intercellular connections. To better examine viral spread via TNTs, we devised an infection system in which conventional, virion-mediated, spread was not possible. Namely, we generated HA-deficient reporter viruses which are unable to produce progeny virions but whose genomes can be replicated and trafficked. In this system, vRNP transfer to neighboring cells was observed and this transfer was found to be dependent on both actin and Rab11a. Generation of infectious virus via TNT transfer was confirmed using donor cells infected with HA-deficient virus and recipient cells stably expressing HA protein. Mixing donor cells infected with genetically distinct IAVs furthermore revealed the potential for Rab11a and TNTs to serve as a conduit for genome mixing and reassortment in IAV infections. These data therefore reveal a novel role for Rab11a in the IAV life cycle, which could have significant implications for within-host spread, genome reassortment and immune evasion.
Collapse
Affiliation(s)
- Ketaki Ganti
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Julianna Han
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa School of Medicine, Iowa City, Iowa, United States of America
| | - Anice C. Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory-UGA Centers of Excellence for Influenza Research and Surveillance [CEIRS]
| |
Collapse
|
83
|
Haas OA. Somatic Sex: On the Origin of Neoplasms With Chromosome Counts in Uneven Ploidy Ranges. Front Cell Dev Biol 2021; 9:631946. [PMID: 34422788 PMCID: PMC8373647 DOI: 10.3389/fcell.2021.631946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 06/22/2021] [Indexed: 01/09/2023] Open
Abstract
Stable aneuploid genomes with nonrandom numerical changes in uneven ploidy ranges define distinct subsets of hematologic malignancies and solid tumors. The idea put forward herein suggests that they emerge from interactions between diploid mitotic and G0/G1 cells, which can in a single step produce all combinations of mono-, di-, tri-, tetra- and pentasomic paternal/maternal homologue configurations that define such genomes. A nanotube-mediated influx of interphase cell cytoplasm into mitotic cells would thus be responsible for the critical nondisjunction and segregation errors by physically impeding the proper formation of the cell division machinery, whereas only a complete cell fusion can simultaneously generate pentasomies, uniparental trisomies as well as biclonal hypo- and hyperdiploid cell populations. The term "somatic sex" was devised to accentuate the similarities between germ cell and somatic cell fusions. A somatic cell fusion, in particular, recapitulates many processes that are also instrumental in the formation of an abnormal zygote that involves a diploid oocyte and a haploid sperm, which then may further develop into a digynic triploid embryo. Despite their somehow deceptive differences and consequences, the resemblance of these two routes may go far beyond of what has hitherto been appreciated. Based on the arguments put forward herein, I propose that embryonic malignancies of mesenchymal origin with these particular types of aneuploidies can thus be viewed as the kind of flawed somatic equivalent of a digynic triploid embryo.
Collapse
Affiliation(s)
- Oskar A Haas
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| |
Collapse
|
84
|
The 3.0 Cell Communication: New Insights in the Usefulness of Tunneling Nanotubes for Glioblastoma Treatment. Cancers (Basel) 2021; 13:cancers13164001. [PMID: 34439156 PMCID: PMC8392307 DOI: 10.3390/cancers13164001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/05/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Communication between cells helps tumors acquire resistance to chemotherapy and makes the struggle against cancer more challenging. Tunneling nanotubes (TNTs) are long channels able to connect both nearby and distant cells, contributing to a more malignant phenotype. This finding might be useful in designing novel strategies of drug delivery exploiting these systems of connection. This would be particularly important to reach tumor niches, where glioblastoma stem cells proliferate and provoke immune escape, thereby increasing metastatic potential and tumor recurrence a few months after surgical resection of the primary mass. Along with the direct inhibition of TNT formation, TNT analysis, and targeting strategies might be useful in providing innovative tools for the treatment of this tumor. Abstract Glioblastoma (GBM) is a particularly challenging brain tumor characterized by a heterogeneous, complex, and multicellular microenvironment, which represents a strategic network for treatment escape. Furthermore, the presence of GBM stem cells (GSCs) seems to contribute to GBM recurrence after surgery, and chemo- and/or radiotherapy. In this context, intercellular communication modalities play key roles in driving GBM therapy resistance. The presence of tunneling nanotubes (TNTs), long membranous open-ended channels connecting distant cells, has been observed in several types of cancer, where they emerge to steer a more malignant phenotype. Here, we discuss the current knowledge about the formation of TNTs between different cellular types in the GBM microenvironment and their potential role in tumor progression and recurrence. Particularly, we highlight two prospective strategies targeting TNTs as possible therapeutics: (i) the inhibition of TNT formation and (ii) a boost in drug delivery between cells through these channels. The latter may require future studies to design drug delivery systems that are exchangeable through TNTs, thus allowing for access to distant tumor niches that are involved in tumor immune escape, maintenance of GSC plasticity, and increases in metastatic potential.
Collapse
|
85
|
Schnatz A, Müller C, Brahmer A, Krämer‐Albers E. Extracellular Vesicles in neural cell interaction and CNS homeostasis. FASEB Bioadv 2021; 3:577-592. [PMID: 34377954 PMCID: PMC8332475 DOI: 10.1096/fba.2021-00035] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
Central nervous system (CNS) homeostasis critically depends on the interaction between neurons and glia cells. Extracellular vesicles (EVs) recently emerged as versatile messengers in CNS cell communication. EVs are released by neurons and glia in activity-dependent manner and address multiple target cells within and outside the nervous system. Here, we summarize the recent advances in understanding the physiological roles of EVs in the nervous system and their ability to deliver signals across the CNS barriers. In addition to the disposal of cellular components via EVs and clearance by phagocytic cells, EVs are involved in plasticity-associated processes, mediate trophic support and neuroprotection, promote axonal maintenance, and modulate neuroinflammation. While individual functional components of the EV cargo are becoming progressively identified, the role of neural EVs as compound multimodal signaling entities remains to be elucidated. Novel transgenic models and imaging technologies allow EV tracking in vivo and provide further insight into EV targeting and their mode of action. Overall, EVs represent key players in the maintenance of CNS homeostasis essential for the lifelong performance of neural networks and thus provide a wide spectrum of biomedical applications.
Collapse
Affiliation(s)
- Andrea Schnatz
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Christina Müller
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Alexandra Brahmer
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Eva‐Maria Krämer‐Albers
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| |
Collapse
|
86
|
Valdebenito S, Malik S, Luu R, Loudig O, Mitchell M, Okafo G, Bhat K, Prideaux B, Eugenin EA. Tunneling nanotubes, TNT, communicate glioblastoma with surrounding non-tumor astrocytes to adapt them to hypoxic and metabolic tumor conditions. Sci Rep 2021; 11:14556. [PMID: 34267246 PMCID: PMC8282675 DOI: 10.1038/s41598-021-93775-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-to-cell communication is essential for the development and proper function of multicellular systems. We and others demonstrated that tunneling nanotubes (TNT) proliferate in several pathological conditions such as HIV, cancer, and neurodegenerative diseases. However, the nature, function, and contribution of TNT to cancer pathogenesis are poorly understood. Our analyses demonstrate that TNT structures are induced between glioblastoma (GBM) cells and surrounding non-tumor astrocytes to transfer tumor-derived mitochondria. The mitochondrial transfer mediated by TNT resulted in the adaptation of non-tumor astrocytes to tumor-like metabolism and hypoxia conditions. In conclusion, TNT are an efficient cell-to-cell communication system used by cancer cells to adapt the microenvironment to the invasive nature of the tumor.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Shaily Malik
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Ross Luu
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Megan Mitchell
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | | | - Krishna Bhat
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, M.D. Anderson, Houston, TX, USA
| | - Brendan Prideaux
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, Fifth Floor, 105 11th Street, Galveston, TX, 77555, USA.
| |
Collapse
|
87
|
Dilsizoglu Senol A, Samarani M, Syan S, Guardia CM, Nonaka T, Liv N, Latour-Lambert P, Hasegawa M, Klumperman J, Bonifacino JS, Zurzolo C. α-Synuclein fibrils subvert lysosome structure and function for the propagation of protein misfolding between cells through tunneling nanotubes. PLoS Biol 2021; 19:e3001287. [PMID: 34283825 PMCID: PMC8291706 DOI: 10.1371/journal.pbio.3001287] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/13/2021] [Indexed: 01/06/2023] Open
Abstract
The accumulation of α-synuclein (α-syn) aggregates in specific brain regions is a hallmark of synucleinopathies including Parkinson disease (PD). α-Syn aggregates propagate in a "prion-like" manner and can be transferred inside lysosomes to recipient cells through tunneling nanotubes (TNTs). However, how lysosomes participate in the spreading of α-syn aggregates is unclear. Here, by using super-resolution (SR) and electron microscopy (EM), we find that α-syn fibrils affect the morphology of lysosomes and impair their function in neuronal cells. In addition, we demonstrate that α-syn fibrils induce peripheral redistribution of lysosomes, likely mediated by transcription factor EB (TFEB), increasing the efficiency of α-syn fibrils' transfer to neighboring cells. We also show that lysosomal membrane permeabilization (LMP) allows the seeding of soluble α-syn in cells that have taken up α-syn fibrils from the culture medium, and, more importantly, in healthy cells in coculture, following lysosome-mediated transfer of the fibrils. Moreover, we demonstrate that seeding occurs mainly at lysosomes in both donor and acceptor cells, after uptake of α-syn fibrils from the medium and following their transfer, respectively. Finally, by using a heterotypic coculture system, we determine the origin and nature of the lysosomes transferred between cells, and we show that donor cells bearing α-syn fibrils transfer damaged lysosomes to acceptor cells, while also receiving healthy lysosomes from them. These findings thus contribute to the elucidation of the mechanism by which α-syn fibrils spread through TNTs, while also revealing the crucial role of lysosomes, working as a Trojan horse for both seeding and propagation of disease pathology.
Collapse
Affiliation(s)
- Aysegul Dilsizoglu Senol
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et de l’Infection, Institut Pasteur, Paris, France
| | - Maura Samarani
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et de l’Infection, Institut Pasteur, Paris, France
| | - Sylvie Syan
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et de l’Infection, Institut Pasteur, Paris, France
| | - Carlos M. Guardia
- Neurosciences and Cellular and Structural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Takashi Nonaka
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Nalan Liv
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Patricia Latour-Lambert
- Dynamique des Interaction Hôte–Pathogène, Département de Biologie Cellulaire et de l’Infection, Institut Pasteur, Paris, France
| | - Masato Hasegawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Juan S. Bonifacino
- Neurosciences and Cellular and Structural Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et de l’Infection, Institut Pasteur, Paris, France
| |
Collapse
|
88
|
Barutta F, Kimura S, Hase K, Bellini S, Corbetta B, Corbelli A, Fiordaliso F, Barreca A, Papotti MG, Ghiggeri GM, Salvidio G, Roccatello D, Audrito V, Deaglio S, Gambino R, Bruno S, Camussi G, Martini M, Hirsch E, Durazzo M, Ohno H, Gruden G. Protective Role of the M-Sec-Tunneling Nanotube System in Podocytes. J Am Soc Nephrol 2021; 32:1114-1130. [PMID: 33722931 PMCID: PMC8259684 DOI: 10.1681/asn.2020071076] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 01/21/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocyte dysfunction and loss are major determinants in the development of proteinuria. FSGS is one of the most common causes of proteinuria, but the mechanisms leading to podocyte injury or conferring protection against FSGS remain poorly understood. The cytosolic protein M-Sec has been involved in the formation of tunneling nanotubes (TNTs), membrane channels that transiently connect cells and allow intercellular organelle transfer. Whether podocytes express M-Sec is unknown and the potential relevance of the M-Sec-TNT system in FSGS has not been explored. METHODS We studied the role of the M-Sec-TNT system in cultured podocytes exposed to Adriamycin and in BALB/c M-Sec knockout mice. We also assessed M-Sec expression in both kidney biopsies from patients with FSGS and in experimental FSGS (Adriamycin-induced nephropathy). RESULTS Podocytes can form TNTs in a M-Sec-dependent manner. Consistent with the notion that the M-Sec-TNT system is cytoprotective, podocytes overexpressed M-Sec in both human and experimental FSGS. Moreover, M-Sec deletion resulted in podocyte injury, with mitochondrial abnormalities and development of progressive FSGS. In vitro, M-Sec deletion abolished TNT-mediated mitochondria transfer between podocytes and altered mitochondrial bioenergetics. Re-expression of M-Sec reestablishes TNT formation and mitochondria exchange, rescued mitochondrial function, and partially reverted podocyte injury. CONCLUSIONS These findings indicate that the M-Sec-TNT system plays an important protective role in the glomeruli by rescuing podocytes via mitochondrial horizontal transfer. M-Sec may represent a promising therapeutic target in FSGS, and evidence that podocytes can be rescued via TNT-mediated horizontal transfer may open new avenues of research.
Collapse
Affiliation(s)
- Federica Barutta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Shunsuke Kimura
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Stefania Bellini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Alessandro Corbelli
- Department of Cardiovascular Medicine, Institute of Pharmacological Research Mario Negri, Scientific Institute for Hospitalization and Care (IRCCS), Milan, Italy
| | - Fabio Fiordaliso
- Department of Cardiovascular Medicine, Institute of Pharmacological Research Mario Negri, Scientific Institute for Hospitalization and Care (IRCCS), Milan, Italy
| | | | | | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis, Transplantation, Gaslini Children’s Hospital, Genoa, Italy
| | - Gennaro Salvidio
- Scientific Institute for Hospitalization and Care (IRCCS), San Martino University Hospital Clinic, Genoa, Italy
| | - Dario Roccatello
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d’Aosta Network for Rare Diseases, S. Giovanni Bosco Hospital, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy,Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy
| | | | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Marilena Durazzo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
89
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
90
|
Patient-derived glioblastoma stem cells transfer mitochondria through tunneling nanotubes in tumor organoids. Biochem J 2021; 478:21-39. [PMID: 33245115 PMCID: PMC7800365 DOI: 10.1042/bcj20200710] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most aggressive brain cancer and its relapse after surgery, chemo and radiotherapy appears to be led by GBM stem cells (GSCs). Also, tumor networking and intercellular communication play a major role in driving GBM therapy-resistance. Tunneling Nanotubes (TNTs), thin membranous open-ended channels connecting distant cells, have been observed in several types of cancer, where they emerge to drive a more malignant phenotype. Here, we investigated whether GBM cells are capable to intercommunicate by TNTs. Two GBM stem-like cells (GSLCs) were obtained from the external and infiltrative zone of one GBM from one patient. We show, for the first time, that both GSLCs, grown in classical 2D culture and in 3D-tumor organoids, formed functional TNTs which allowed mitochondria transfer. In the organoid model, recapitulative of several tumor's features, we observed the formation of a network between cells constituted of both Tumor Microtubes (TMs), previously observed in vivo, and TNTs. In addition, the two GSLCs exhibited different responses to irradiation in terms of TNT induction and mitochondria transfer, although the correlation with the disease progression and therapy-resistance needs to be further addressed. Thus, TNT-based communication is active in different GSLCs derived from the external tumoral areas associated to GBM relapse, and we propose that they participate together with TMs in tumor networking.
Collapse
|
91
|
Zurzolo C. Tunneling nanotubes: Reshaping connectivity. Curr Opin Cell Biol 2021; 71:139-147. [PMID: 33866130 DOI: 10.1016/j.ceb.2021.03.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/22/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022]
Abstract
Tunneling nanotubes (TNTs), open membranous channels between connected cells, represent a novel direct way of communication between distant cells for the diffusion of various cellular material, including survival or death signals, genetic material, organelles, and pathogens. Their discovery prompted us to review our understanding of many physiological and pathological processes involving cellular communication but also allowed us to discover new mechanisms of communication at a distance. While this has enriched the field, it has also generated some confusion, as different TNT-like protrusions have been described, and it is not clear whether they have the same structure-function. Most studies have been based on low-resolution imaging methods, and one of the major problems is the inconsistency in demonstrating the capacity of these various connections to transfer material between cells belonging to different populations. This brief review examines the fundamental properties of TNTs. In adult tissues, TNTs are stimulated by different diseases, stresses, and inflammatory signals. 'Moreover', based on the similarity of the processes of development of synaptic spines and TNT formation, we argue that TNTs in the brain predate synaptic transmission, being instrumental in the orchestration of the immature neuronal circuit.
Collapse
Affiliation(s)
- Chiara Zurzolo
- Membrane Traffic and Pathogenesis, Institut Pasteur, UMR3691 CNRS, 75015, Paris, France.
| |
Collapse
|
92
|
Cordero Cervantes D, Zurzolo C. Peering into tunneling nanotubes-The path forward. EMBO J 2021; 40:e105789. [PMID: 33646572 PMCID: PMC8047439 DOI: 10.15252/embj.2020105789] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/21/2020] [Accepted: 01/15/2021] [Indexed: 12/19/2022] Open
Abstract
The identification of Tunneling Nanotubes (TNTs) and TNT-like structures signified a critical turning point in the field of cell-cell communication. With hypothesized roles in development and disease progression, TNTs' ability to transport biological cargo between distant cells has elevated these structures to a unique and privileged position among other mechanisms of intercellular communication. However, the field faces numerous challenges-some of the most pressing issues being the demonstration of TNTs in vivo and understanding how they form and function. Another stumbling block is represented by the vast disparity in structures classified as TNTs. In order to address this ambiguity, we propose a clear nomenclature and provide a comprehensive overview of the existing knowledge concerning TNTs. We also discuss their structure, formation-related pathways, biological function, as well as their proposed role in disease. Furthermore, we pinpoint gaps and dichotomies found across the field and highlight unexplored research avenues. Lastly, we review the methods employed to date and suggest the application of new technologies to better understand these elusive biological structures.
Collapse
Affiliation(s)
| | - Chiara Zurzolo
- Institut PasteurMembrane Traffic and PathogenesisParisFrance
| |
Collapse
|
93
|
Erdogan B, Whited JL. Engineered myosins drive filopodial transport. Nat Cell Biol 2021; 23:113-115. [PMID: 33526903 DOI: 10.1038/s41556-021-00632-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Burcu Erdogan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
94
|
Biodistribution of surfactant-free poly(lactic-acid) nanoparticles and uptake by endothelial cells and phagocytes in zebrafish: Evidence for endothelium to macrophage transfer. J Control Release 2021; 331:228-245. [PMID: 33444668 DOI: 10.1016/j.jconrel.2021.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/16/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
In the development of therapeutic nanoparticles (NP), there is a large gap between in vitro testing and in vivo experimentation. Despite its prominence as a model, the mouse shows severe limitations for imaging NP and the cells with which they interact. Recently, the transparent zebrafish larva, which is well suited for high-resolution live-imaging, has emerged as a powerful alternative model to investigate the in vivo behavior of NP. Poly(D,L lactic acid) (PLA) is widely accepted as a safe polymer to prepare therapeutic NP. However, to prevent aggregation, many NP require surfactants, which may have undesirable biological effects. Here, we evaluate 'safe-by-design', surfactant-free PLA-NP that were injected intravenously into zebrafish larvae. Interaction of fluorescent NPs with different cell types labelled in reporter animals could be followed in real-time at high resolution; furthermore, by encapsulating colloidal gold into the matrix of PLA-NP we could follow their fate in more detail by electron microscopy, from uptake to degradation. The rapid clearance of fluorescent PLA-NP from the circulation coincided with internalization by endothelial cells lining the whole vasculature and macrophages. After 30 min, when no NP remained in circulation, we observed that macrophages continued to internalize significant amounts of NP. More detailed video-imaging revealed a new mechanism of NP transfer where NP are transmitted along with parts of the cytoplasm from endothelial cells to macrophages.
Collapse
|
95
|
Opportunities and Challenges in Tunneling Nanotubes Research: How Far from Clinical Application? Int J Mol Sci 2021; 22:ijms22052306. [PMID: 33669068 PMCID: PMC7956326 DOI: 10.3390/ijms22052306] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/21/2021] [Accepted: 02/21/2021] [Indexed: 02/08/2023] Open
Abstract
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. Herein, we emphasize the heterogeneous nature of TNTs by systemically summarizing the current knowledge on their morphology, structure, and biogenesis in different types of cells. Furthermore, we address the communication efficiency and biological outcomes of TNT-dependent transport related to diseases. Finally, we discuss the opportunities and challenges of TNTs as an exciting therapeutic approach by focusing on the development of efficient and safe drugs targeting TNTs.
Collapse
|
96
|
Shahar M, Szalat A, Rosen H. Pathogenic Stress Induces Human Monocyte to Express an Extracellular Web of Tunneling Nanotubes. Front Immunol 2021; 12:620734. [PMID: 33679763 PMCID: PMC7933571 DOI: 10.3389/fimmu.2021.620734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Actin-based tunneling nanotubes are a means of intercellular communication between remote cells. In the last decade, this type of nanotube was described in a wide variety of cell types and it became widely accepted that communication through these nanotubes is related to response to environmental changes. Few reports, however, are available regarding the expression of similar nanotubes in vivo or in primary cells. Moreover, the functional significance of this intercellular communication for health and disease is largely unknown. In this context, and as a first step in unraveling these questions, we examined the formation of similar nanotubes in primary peripheral human monocytes. To that end, we combined the use of a live cell imaging system along with advanced methods of fluorescent and scanning electron microscopy. This experimental approach reveals for the first time that the bacterial lipopolysaccharide endotoxin induces a transient expression of an unexpected abundance of actin-based tunneling nanotubes associated with vesicles. In addition, it was found that a similar response can be achieved by treating human monocytes with various bacterial and yeast membrane components, as well as with a viral component analog. In all these cases, this response is mediated by distinct complexes of toll-like receptors. Therefore, we suggest that the observed phenomena are related to a broad type of monocyte pathogen response, and raise the possibility that the phenomena described above may be involved in many clinical situations related to inflammation as a new topic of study.
Collapse
Affiliation(s)
- Michal Shahar
- The Department of Microbiology and Molecular Genetics, Institute for Medical Research-Israel-Canada, Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| | - Auryan Szalat
- Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haim Rosen
- The Kuvin Center for the Study of Infectious and Tropical Diseases, Institute for Medical Research-Israel-Canada, Hebrew University - Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
97
|
Zhu C, Shi Y, You J. Immune Cell Connection by Tunneling Nanotubes: The Impact of Intercellular Cross-Talk on the Immune Response and Its Therapeutic Applications. Mol Pharm 2021; 18:772-786. [PMID: 33529022 DOI: 10.1021/acs.molpharmaceut.0c01248] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Direct intercellular communication is an important prerequisite for the development of multicellular organisms, the regeneration of tissue, and the maintenance of various physiological activities. Tunnel nanotubes (TNTs), which have diameters of approximately 50-1500 nm and lengths of up to several cell diameters, can connect cells over long distances and have emerged as one of the most important recently discovered types of efficient communication between cells. Moreover, TNTs can also directly transfer organelles, vehicles, proteins, genetic material, ions, and small molecules from one cell to adjacent and even distant cells. However, the mechanism of intercellular communication between various immune cells within the complex immune system has not been fully elucidated. Studies in the past decades have confirmed the existence of TNTs in many types of cells, especially in various kinds of immune cells. TNTs display different structural and functional characteristics between and within different immunocytes, playing a major role in the transmission of signals across various kinds of immune cells. In this review, we introduce the discovery and structure of TNTs, as well as their different functional properties within different immune cells. We also discuss the roles of TNTs in potentiating the immune response and their potential therapeutic applications.
Collapse
Affiliation(s)
- Chunqi Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| |
Collapse
|
98
|
Peters WS, Jensen KH, Stone HA, Knoblauch M. Plasmodesmata and the problems with size: Interpreting the confusion. JOURNAL OF PLANT PHYSIOLOGY 2021; 257:153341. [PMID: 33388666 DOI: 10.1016/j.jplph.2020.153341] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 05/14/2023]
Abstract
Plant tissues exhibit a symplasmic organization; the individual protoplasts are connected to their neighbors via cytoplasmic bridges that extend through pores in the cell walls. These bridges may have diameters of a micrometer or more, as in the sieve pores of the phloem, but in most cell types they are smaller. Historically, botanists referred to cytoplasmic bridges of all sizes as plasmodesmata. The meaning of the term began to shift when the transmission electron microscope (TEM) became the preferred tool for studying these structures. Today, a plasmodesma is widely understood to be a 'nano-scale' pore. Unfortunately, our understanding of these nanoscopic channels suffers from methodological limitations. This is exemplified by the fact that state-of-the-art EM techniques appear to reveal plasmodesmal pore structures that are much smaller than the tracer molecules known to diffuse through these pores. In general, transport processes in pores that have dimensions in the size range of the transported molecules are governed by different physical parameters than transport process in the macroscopic realm. This can lead to unexpected effects, as experience in nanofluidic technologies demonstrates. Our discussion of problems of size in plasmodesma research leads us to conclude that the field will benefit from technomimetic reasoning - the utilization of concepts developed in applied nanofluidics for the interpretation of biological systems.
Collapse
Affiliation(s)
- Winfried S Peters
- School of Biological Sciences, Washington State University, Pullman, WA, 99164, USA; Department of Biology, Purdue University Fort Wayne, Fort Wayne, IN, 46805, USA.
| | - Kaare H Jensen
- Department of Physics, Technical University of Denmark, DK-2800 Kgs., Lyngby, Denmark.
| | - Howard A Stone
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ, 08544, USA.
| | - Michael Knoblauch
- School of Biological Sciences, Washington State University, Pullman, WA, 99164, USA.
| |
Collapse
|
99
|
Pinnell JR, Cui M, Tieu K. Exosomes in Parkinson disease. J Neurochem 2021; 157:413-428. [PMID: 33372290 DOI: 10.1111/jnc.15288] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022]
Abstract
Parkinson disease (PD) is a prevalent neurodegenerative disease, in which the formation of misfolded and aggregated α-synuclein is a key neuropathological hallmark. Recent studies reveal that extracellular vesicles such as exosomes present a potential mechanism for propagation of pathological α-synuclein throughout the brain. The ability of exosomes to transport proteins and genetic material between cells, including mRNA and microRNAs which have been implicated in PD pathology, provides critical insights as to how exosomes may contribute to pathological progression in PD. Advances have also been made in the investigation of exosomes as potential tools for the modulation of Parkinson's pathology; their detection extracellularly may facilitate their use as biomarkers, while their small size could be utilised as vectors for the delivery of therapeutics. The aim of this review was to highlight our current knowledge of the role of exosomes in PD and potential clinical application.
Collapse
Affiliation(s)
- Jennifer R Pinnell
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA.,Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, UK
| | - Mei Cui
- Department of Neurology, Huashan hospital, Fudan University, Shanghai, China
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA.,Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| |
Collapse
|
100
|
Dagar S, Pushpa K, Pathak D, Samaddar S, Saxena A, Banerjee S, Mylavarapu SVS. Nucleolin regulates 14-3-3ζ mRNA and promotes cofilin phosphorylation to induce tunneling nanotube formation. FASEB J 2021; 35:e21199. [PMID: 33222276 DOI: 10.1096/fj.202001152r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/19/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2023]
Abstract
Tunneling nanotubes (TNTs) mediate intercellular communication between animal cells in health and disease, but the mechanisms of their biogenesis and function are poorly understood. Here we report that the RNA-binding protein (RBP) nucleolin, which interacts with the known TNT-inducing protein MSec, is essential for TNT formation in mammalian cells. Nucleolin, through its RNA-binding domains (RBDs), binds to and maintains the cytosolic levels of 14-3-3ζ mRNA, and is, therefore, required for TNT formation. A specific region of the 3'-untranslated region (UTR) of the 14-3-3ζ mRNA is likely to be involved in its regulation by nucleolin. Functional complementation experiments suggest that nucleolin and 14-3-3ζ form a linear signaling axis that promotes the phosphorylation and inactivation of the F-actin depolymerization factor cofilin to induce TNT formation. MSec also similarly inactivates cofilin, but potentiates TNT formation independent of the nucleolin-14-3-3ζ axis, despite biochemically interacting with both proteins. We show that 14-3-3ζ and nucleolin are required for the formation of TNTs between primary mouse neurons and astrocytes and in multiple other mammalian cell types. We also report that the Caenorhabditis elegans orthologs of 14-3-3ζ and MSec regulate the size and architecture of the TNT-like cellular protrusions of the distal tip cell (DTC), the germline stem cell niche in the gonad. Our study demonstrates a novel and potentially conserved mRNA-guided mechanism of TNT formation through the maintenance of cellular 14-3-3ζ mRNA levels by the RBP nucleolin.
Collapse
Affiliation(s)
- Sunayana Dagar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
- Affiliated to the Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | - Kumari Pushpa
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| | - Diksha Pathak
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| | | | - Anjana Saxena
- Biology Department, Brooklyn College AND Biology and Biochemistry Programs, Graduate Center, CUNY, New York, NY, USA
| | | | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
- Affiliated to the Kalinga Institute of Industrial Technology, Bhubaneswar, India
| |
Collapse
|