1
|
Alekseenko I, Zhukova L, Kondratyeva L, Buzdin A, Chernov I, Sverdlov E. Tumor Cell Communications as Promising Supramolecular Targets for Cancer Chemotherapy: A Possible Strategy. Int J Mol Sci 2024; 25:10454. [PMID: 39408784 PMCID: PMC11476449 DOI: 10.3390/ijms251910454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Fifty-two years have passed since President Nixon launched the "War on Cancer". Despite unparalleled efforts and funds allocated worldwide, the outlined goals were not achieved because cancer treatment approaches such as chemotherapy, radiation therapy, hormonal and targeted therapies have not fully met the expectations. Based on the recent literature, a new direction in cancer therapy can be proposed which targets connections between cancer cells and their microenvironment by chemical means. Cancer-stromal synapses such as immunological synapses between cancer and immune cells provide an attractive target for this approach. Such synapses form ligand-receptor clusters on the interface of the interacting cells. They share a common property of involving intercellular clusters of spatially proximate and cooperatively acting proteins. Synapses provide the space for the focused intercellular signaling molecules exchange. Thus, the disassembly of cancer-stromal synapses may potentially cause the collapse of various tumors. Additionally, the clustered arrangement of synapse components offers opportunities to enhance treatment safety and precision by using targeted crosslinking chemical agents which may inactivate cancer synapses even in reduced concentrations. Furthermore, attaching a cleavable cell-permeable toxic agent(s) to a crosslinker may further enhance the anti-cancer effect of such therapeutics. The highlighted approach promises to be universal, relatively simple and cost-efficient. We also hope that, unlike chemotherapeutic and immune drugs that interact with a single target, by using supramolecular large clusters that include many different components as a target, the emergence of a resistance characteristic of chemo- and immunotherapy is extremely unlikely.
Collapse
Affiliation(s)
- Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Lyudmila Zhukova
- Department of Oncology, SBIH “Moscow Clinical Scientific and Practical Center Named After A.S. Loginov” DHM, 111123 Moscow, Russia;
| | - Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119992 Moscow, Russia
- Oncobox LLC, 121205 Moscow, Russia
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
| | - Eugene Sverdlov
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| |
Collapse
|
2
|
Pichler V, Dalkilic L, Shoaib G, Shapira T, Rankine-Wilson L, Boudehen YM, Chao JD, Sexton D, Prieto M, Quon BS, Tocheva EI, Kremer L, Hsiao W, Av-Gay Y. The diversity of clinical Mycobacterium abscessus isolates in morphology, glycopeptidolipids and infection rates in a macrophage model. J Med Microbiol 2024; 73. [PMID: 39158416 DOI: 10.1099/jmm.0.001869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
Introduction. Mycobacterium abscessus (MABS) is a pathogenic bacterium that can cause severe lung infections, particularly in individuals with cystic fibrosis. MABS colonies can exhibit either a smooth (S) or rough (R) morphotype, influenced by the presence or absence of glycopeptidolipids (GPLs) on their surface, respectively. Despite the clinical significance of these morphotypes, the relationship between GPL levels, morphotype and the pathogenesis of MABS infections remains poorly understood.Gap statement. The mechanisms and implications of GPL production and morphotypes in clinical MABS infections are unclear. There is a gap in understanding their correlation with infectivity and pathogenicity, particularly in patients with underlying lung disease.Aim. This study aimed to investigate the correlation between MABS morphology, GPL and infectivity by analysing strains from cystic fibrosis patients' sputum samples.Methodology. MABS was isolated from patient sputum samples and categorized by morphotype, GPL profile and replication rate in macrophages. A high-content ex vivo infection model using THP-1 cells assessed the infectivity of both clinical and laboratory strains.Results. Our findings revealed that around 50 % of isolates displayed mixed morphologies. GPL analysis confirmed a consistent relationship between GPL content and morphotype that was only found in smooth isolates. Across morphotype groups, no differences were observed in vitro, yet clinical R strains were observed to replicate at higher levels in the THP-1 infection model. Moreover, the proportion of infected macrophages was notably higher among clinical R strains compared to their S counterparts at 72 h post-infection. Clinical variants also infected THP-1 cells at significantly higher rates compared to laboratory strains, highlighting the limited translatability of lab strain infection data to clinical contexts.Conclusion. Our study confirmed the general correlation between morphotype and GPL levels in smooth strains yet unveiled more variability within morphotype groups than previously recognized, particularly during intracellular infection. As the R morphotype is the highest clinical concern, these findings contribute to the expanding knowledge base surrounding MABS infections, offering insights that can steer diagnostic methodologies and treatment approaches.
Collapse
Affiliation(s)
- Virginia Pichler
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- INSERM, IRIM, 34293 Montpellier, France
| | - Lara Dalkilic
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Ghazaleh Shoaib
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Tirosh Shapira
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Leah Rankine-Wilson
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | - Joseph D Chao
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Danielle Sexton
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Miguel Prieto
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Bradley S Quon
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Elitza I Tocheva
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | - William Hsiao
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Yossef Av-Gay
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
3
|
Ebrahim T, Ebrahim AS, Kandouz M. Diversity of Intercellular Communication Modes: A Cancer Biology Perspective. Cells 2024; 13:495. [PMID: 38534339 PMCID: PMC10969453 DOI: 10.3390/cells13060495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
From the moment a cell is on the path to malignant transformation, its interaction with other cells from the microenvironment becomes altered. The flow of molecular information is at the heart of the cellular and systemic fate in tumors, and various processes participate in conveying key molecular information from or to certain cancer cells. For instance, the loss of tight junction molecules is part of the signal sent to cancer cells so that they are no longer bound to the primary tumors and are thus free to travel and metastasize. Upon the targeting of a single cell by a therapeutic drug, gap junctions are able to communicate death information to by-standing cells. The discovery of the importance of novel modes of cell-cell communication such as different types of extracellular vesicles or tunneling nanotubes is changing the way scientists look at these processes. However, are they all actively involved in different contexts at the same time or are they recruited to fulfill specific tasks? What does the multiplicity of modes mean for the overall progression of the disease? Here, we extend an open invitation to think about the overall significance of these questions, rather than engage in an elusive attempt at a systematic repertory of the mechanisms at play.
Collapse
Affiliation(s)
- Thanzeela Ebrahim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48202, USA
| |
Collapse
|
4
|
Matejka N, Amarlou A, Neubauer J, Rudigkeit S, Reindl J. High-Resolution Microscopic Characterization of Tunneling Nanotubes in Living U87 MG and LN229 Glioblastoma Cells. Cells 2024; 13:464. [PMID: 38474428 PMCID: PMC10931022 DOI: 10.3390/cells13050464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
Tunneling nanotubes (TNTs) are fine, nanometer-sized membrane connections between distant cells that provide an efficient communication tool for cellular organization. TNTs are thought to play a critical role in cellular behavior, particularly in cancer cells. The treatment of aggressive cancers such as glioblastoma remains challenging due to their high potential for developing therapy resistance, high infiltration rates, uncontrolled cell growth, and other aggressive features. A better understanding of the cellular organization via cellular communication through TNTs could help to find new therapeutic approaches. In this study, we investigate the properties of TNTs in two glioblastoma cell lines, U87 MG and LN229, including measurements of their diameter by high-resolution live-cell stimulated emission depletion (STED) microscopy and an analysis of their length, morphology, lifetime, and formation by live-cell confocal microscopy. In addition, we discuss how these fine compounds can ideally be studied microscopically. In particular, we show which membrane-labeling method is suitable for studying TNTs in glioblastoma cells and demonstrate that live-cell studies should be preferred to explore the role of TNTs in cellular behavior. Our observations on TNT formation in glioblastoma cells suggest that TNTs could be involved in cell migration and serve as guidance.
Collapse
Affiliation(s)
- Nicole Matejka
- Institute for Applied Physics and Measurement Technology, University of the Bundeswehr Munich, 85577 Neubiberg, Germany; (A.A.); (J.N.); (S.R.); (J.R.)
| | | | | | | | | |
Collapse
|
5
|
Budinger D, Baker V, Heneka MT. Tunneling Nanotubes in the Brain. Results Probl Cell Differ 2024; 73:203-227. [PMID: 39242381 DOI: 10.1007/978-3-031-62036-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Tunneling nanotubes (TNTs) have emerged as intriguing structures facilitating intercellular communications across diverse cell types, which are integral to several biological processes, as well as participating in various disease progression. This review provides an in-depth analysis of TNTs, elucidating their structural characteristics and functional roles, with a particular focus on their significance within the brain environment and their implications in neurological and neurodegenerative disorders. We explore the interplay between TNTs and neurological diseases, offering potential mechanistic insights into disease progression, while also highlighting their potential as viable therapeutic targets. Additionally, we address the significant challenges associated with studying TNTs, from technical limitations to their investigation in complex biological systems. By addressing some of these challenges, this review aims to pave the way for further exploration into TNTs, establishing them as a central focus in advancing our understanding of neurodegenerative disorders.
Collapse
Affiliation(s)
- Dimitri Budinger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Vivian Baker
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Belval, Luxembourg.
| |
Collapse
|
6
|
Wang YF, Shen ZF, Xiang FY, Wang H, Zhang P, Zhang Q. The direct transfer approach for transcellular drug delivery. Drug Deliv 2023; 30:2288799. [PMID: 38037327 PMCID: PMC10987047 DOI: 10.1080/10717544.2023.2288799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/12/2023] [Indexed: 12/02/2023] Open
Abstract
A promising paradigm for drug administration that has garnered increasing attention in recent years is the direct transfer (DT) of nanoparticles for transcellular drug delivery. DT requires direct cell-cell contact and facilitates unidirectional and bidirectional matter exchange between neighboring cells. Consequently, DT enables fast and deep penetration of drugs into the targeted tissues. This comprehensive review discusses the direct transfer concept, which can be delineated into the following three distinct modalities: membrane contact-direct transfer, gap junction-mediated direct transfer (GJ-DT), and tunneling nanotubes-mediated direct transfer (TNTs-DT). Further, the intercellular structures for each modality of direct transfer and their respective merits and demerits are summarized. The review also discusses the recent progress on the drugs or drug delivery systems that could activate DT.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Graduate Department, Bengbu Medical College, Bengbu, Anhui, China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ze-Fan Shen
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Fang-yue Xiang
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Heng Wang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Pu Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi Zhang
- Graduate Department, Bengbu Medical College, Bengbu, Anhui, China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Sáenz-de-Santa-María I, Henderson JM, Pepe A, Zurzolo C. Identification and Characterization of Tunneling Nanotubes for Intercellular Trafficking. Curr Protoc 2023; 3:e939. [PMID: 37994667 DOI: 10.1002/cpz1.939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Tunneling nanotubes (TNTs) are thin membranous channels providing a direct cytoplasmic connection between remote cells. They are commonly observed in different cell cultures and increasing evidence supports their role in intercellular communication, and pathogen and amyloid protein transfer. However, the study of TNTs presents several pitfalls (e.g., difficulty in preserving such delicate structures, possible confusion with other protrusions, structural and functional heterogeneity, etc.) and therefore requires thoroughly designed approaches. The methods described in this protocol represent a guideline for the characterization of TNTs (or TNT-like structures) in cell culture. Specifically, optimized protocols to (1) identify TNTs and the cytoskeletal elements present inside them; (2) evaluate TNT frequency in cell culture; (3) unambiguously distinguish them from other cellular connections or protrusions; (4) monitor their formation in living cells; (5) characterize TNTs by a micropatterning approach; and (6) investigate TNT ultrastructure by cryo-EM are provided. Finally, this article describes how to assess TNT-mediated cell-to-cell transfer of cellular components, which is a fundamental criterion for identifying functional TNTs. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Identification of tunneling nanotubes Alternate Protocol 1: Identifying the cytoskeletal elements present in tunneling nanotubes Alternate Protocol 2: Distinguishing tunneling nanotubes from intercellular bridges formed during cell division Basic Protocol 2: Deciphering tunneling nanotube formation and lifetime by live fluorescent microscopy Alternate Protocol 3: Deciphering tunneling nanotube formation using a live-compatible dye Basic Protocol 3: Assessing tunneling nanotubes functionality in intercellular transfer Alternate Protocol 4: Flow cytometry approach to quantify the rate of vesicle or mitochondria transfer Support Protocol: Controls to support TNT-mediated transfer Basic Protocol 4: Studies of tunneling nanotubes by cell micropatterning Basic Protocol 5: Characterization of the ultrastructure of tunneling nanotubes by cryo-EM.
Collapse
Affiliation(s)
- Inés Sáenz-de-Santa-María
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, CNRS UMR3691, Paris, France
| | - J Michael Henderson
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Anna Pepe
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et Infection, Institut Pasteur, CNRS UMR3691, Paris, France
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Dagar S, Subramaniam S. Tunneling Nanotube: An Enticing Cell-Cell Communication in the Nervous System. BIOLOGY 2023; 12:1288. [PMID: 37886998 PMCID: PMC10604474 DOI: 10.3390/biology12101288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
The field of neuroscience is rapidly progressing, continuously uncovering new insights and discoveries. Among the areas that have shown immense potential in research, tunneling nanotubes (TNTs) have emerged as a promising subject of study. These minute structures act as conduits for the transfer of cellular materials between cells, representing a mechanism of communication that holds great significance. In particular, the interplay facilitated by TNTs among various cell types within the brain, including neurons, astrocytes, oligodendrocytes, glial cells, and microglia, can be essential for the normal development and optimal functioning of this complex organ. The involvement of TNTs in neurodegenerative disorders, such as Alzheimer's disease, Huntington's disease, and Parkinson's disease, has attracted significant attention. These disorders are characterized by the progressive degeneration of neurons and the subsequent decline in brain function. Studies have predicted that TNTs likely play critical roles in the propagation and spread of pathological factors, contributing to the advancement of these diseases. Thus, there is a growing interest in understanding the precise functions and mechanisms of TNTs within the nervous system. This review article, based on our recent work on Rhes-mediated TNTs, aims to explore the functions of TNTs within the brain and investigate their implications for neurodegenerative diseases. Using the knowledge gained from studying TNTs could offer novel opportunities for designing targeted treatments that can stop the progression of neurodegenerative disorders.
Collapse
Affiliation(s)
- Sunayana Dagar
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Srinivasa Subramaniam
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
- The Scripps Research Institute, La Jolla, CA 92037, USA
- Norman Fixel Institute for Neurological Diseases, 130 Scripps Way, C323, Jupiter, FL 33458, USA
| |
Collapse
|
9
|
Chakraborty R, Nonaka T, Hasegawa M, Zurzolo C. Tunnelling nanotubes between neuronal and microglial cells allow bi-directional transfer of α-Synuclein and mitochondria. Cell Death Dis 2023; 14:329. [PMID: 37202391 DOI: 10.1038/s41419-023-05835-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/20/2023]
Abstract
Tunnelling Nanotubes (TNTs) facilitate contact-mediated intercellular communication over long distances. Material transfer via TNTs can range from ions and intracellular organelles to protein aggregates and pathogens. Prion-like toxic protein aggregates accumulating in several neurodegenerative pathologies, such as Alzheimer's, Parkinson's, and Huntington's diseases, have been shown to spread via TNTs not only between neurons, but also between neurons-astrocytes, and neurons-pericytes, indicating the importance of TNTs in mediating neuron-glia interactions. TNT-like structures were also reported between microglia, however, their roles in neuron-microglia interaction remain elusive. In this work, we quantitatively characterise microglial TNTs and their cytoskeletal composition, and demonstrate that TNTs form between human neuronal and microglial cells. We show that α-Synuclein (α-Syn) aggregates increase the global TNT-mediated connectivity between cells, along with the number of TNT connections per cell pair. Homotypic TNTs formed between microglial cells, and heterotypic TNTs between neuronal and microglial cells are furthermore shown to be functional, allowing movement of both α-Syn and mitochondria. Quantitative analysis shows that α-Syn aggregates are transferred predominantly from neuronal to microglial cells, possibly as a mechanism to relieve the burden of accumulated aggregates. By contrast, microglia transfer mitochondria preferably to α-Syn burdened neuronal cells over the healthy ones, likely as a potential rescue mechanism. Besides describing novel TNT-mediated communication between neuronal and microglial cells, this work allows us to better understand the cellular mechanisms of spreading neurodegenerative diseases, shedding light on the role of microglia.
Collapse
Affiliation(s)
- Ranabir Chakraborty
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, Paris, France
| | - Takashi Nonaka
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hasegawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris, France.
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
10
|
Girolamo F, Lim YP, Virgintino D, Stonestreet BS, Chen XF. Inter-Alpha Inhibitor Proteins Modify the Microvasculature after Exposure to Hypoxia-Ischemia and Hypoxia in Neonatal Rats. Int J Mol Sci 2023; 24:6743. [PMID: 37047713 PMCID: PMC10094872 DOI: 10.3390/ijms24076743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Microvasculature develops during early brain development. Hypoxia-ischemia (HI) and hypoxia (H) predispose to brain injury in neonates. Inter-alpha inhibitor proteins (IAIPs) attenuate injury to the neonatal brain after exposure to HI. However, the effects of IAIPs on the brain microvasculature after exposure to HI have not been examined in neonates. Postnatal day-7 rats were exposed to sham treatment or right carotid artery ligation and 8% oxygen for 90 min. HI comprises hypoxia (H) and ischemia to the right hemisphere (HI-right) and hypoxia to the whole body, including the left hemisphere (H-left). Human IAIPs (hIAIPs, 30 mg/kg) or placebo were injected immediately, 24 and 48 h after HI/H. The brains were analyzed 72 h after HI/H to determine the effects of hIAIPs on the microvasculature by laminin immunohistochemistry and calculation of (1) the percentage area stained by laminin, (2) cumulative microvessel length, and (3) density of tunneling nanotubes (TNTs), which are sensitive indicators of the earliest phases of neo-vascularization/collateralization. hIAIPs mainly affected the percent of the laminin-stained area after HI/H, cumulative vessel length after H but not HI, and TNT density in females but not males. hIAIPs modify the effects of HI/H on the microvasculature after brain injury in neonatal rats and exhibit sex-related differential effects. Our findings suggest that treatment with hIAIPs after exposure to H and HI in neonatal rats affects the laminin content of the vessel basal lamina and angiogenic responses in a sex-related fashion.
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Daniela Virgintino
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi F. Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
11
|
Chakraborty R, Belian S, Zurzolo C. Hijacking intercellular trafficking for the spread of protein aggregates in neurodegenerative diseases: a focus on tunneling nanotubes (TNTs). EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:27-43. [PMID: 39698299 PMCID: PMC11648486 DOI: 10.20517/evcna.2023.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 12/20/2024]
Abstract
Over the years, the influence of secretory mechanisms on intercellular communication has been extensively studied. In the central nervous system (CNS), both trans-synaptic (neurotransmitter-based) and long-distance (extracellular vesicles-based) communications regulate activities and homeostasis. In less than a couple of decades, however, there has been a major paradigm shift in our understanding of intercellular communication. Increasing evidence suggests that besides secretory mechanisms (via extracellular vesicles), several cells are capable of establishing long-distance communication routes referred to as Tunneling Nanotubes (TNTs). TNTs are membranous bridges classically supported by F-Actin filaments, allowing for the exchange of different types of intracellular components between the connected cells, ranging from ions and organelles to pathogens and toxic protein aggregates. The roles of TNTs in pathological spreading of several neurodegenerative conditions such as Prion diseases, Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD) have been well established. However, the fragile nature of these structures and lack of specific biomarkers raised some skepticism regarding their existence. In this review, we will first place TNTs within the spectrum of intercellular communication mechanisms before discussing their known and hypothesized biological relevance in vitro and in vivo in physiological and neurodegenerative contexts. Finally, we discuss the challenges and promising prospects in the field of TNT studies.
Collapse
Affiliation(s)
- Ranabir Chakraborty
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris F-75015, France
- Université Paris Saclay, Gif-sur-Yvette, Paris 91190, France
- Authors contributed equally
| | - Sevan Belian
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris F-75015, France
- Université Paris Saclay, Gif-sur-Yvette, Paris 91190, France
- Authors contributed equally
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris F-75015, France
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Jahnke R, Matthiesen S, Zaeck LM, Finke S, Knittler MR. Chlamydia trachomatis Cell-to-Cell Spread through Tunneling Nanotubes. Microbiol Spectr 2022; 10:e0281722. [PMID: 36219107 PMCID: PMC9769577 DOI: 10.1128/spectrum.02817-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/16/2022] [Indexed: 01/09/2023] Open
Abstract
Tunneling nanotubes (TNTs) are transient cellular connections that consist of dynamic membrane protrusions. They play an important role in cell-to-cell communication and mediate the intercellular exchanges of molecules and organelles. TNTs can form between different cell types and may contribute to the spread of pathogens by serving as cytoplasmic corridors. We demonstrate that Chlamydia (C.) trachomatis-infected human embryonic kidney (HEK) 293 cells and other cells form TNT-like structures through which reticulate bodies (RBs) pass into uninfected cells. Observed TNTs have a life span of 1 to 5 h and contain microtubules, which are essential for chlamydial transfer. They can bridge distances of up to 50 μm between connecting neighboring cells. Consistent with the biological role for TNTs, we show that C. trachomatis spread also occurs under conditions in which the extracellular route of chlamydial entry into host cells is blocked. Based on our findings, we propose that TNTs play a critical role in the direct, cell-to-cell transmission of chlamydia. IMPORTANCE Intracellular bacterial pathogens often undergo a life cycle in which they parasitize infected host cells in membranous vacuoles. Two pathways have been described by which chlamydia can exit infected host cells: lytic cell destruction or exit via extrusion formation. Whether direct, cell-to-cell contact may also play a role in the spread of infection is unknown. Tunneling nanotubes (TNTs) interconnect the cytoplasm of adjacent cells to mediate efficient communication and the exchange of material between them. We used Chlamydia trachomatis and immortalized cells to analyze whether TNTs mediate bacterial transmission from an infected donor to uninfected acceptor cells. We show that chlamydia-infected cells build TNTs through which the intracellular reticulate bodies (RBs) of the chlamydia can pass into uninfected neighboring cells. Our study contributes to the understanding of the function of TNTs in the cell-to-cell transmission of intracellular pathogens and provides new insights into the strategies by which chlamydia spreads among multicellular tissues.
Collapse
Affiliation(s)
- Rico Jahnke
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald, Germany
| | - Svea Matthiesen
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald, Germany
| | - Luca M. Zaeck
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald, Germany
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald, Germany
| | - Michael R. Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald, Germany
| |
Collapse
|
13
|
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
|
14
|
Chelladurai R, Debnath K, Jana NR, Basu JK. Spontaneous formation and growth kinetics of lipid nanotubules induced by passive nanoparticles. SOFT MATTER 2022; 18:7082-7090. [PMID: 36043324 DOI: 10.1039/d2sm00900e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Lipid nanotubules (LNTs) are conduits that form on the membranes of cells and organelles, and they are ubiquitous in all forms of life from archaea and bacteria to plants and mammals. The formation, shape and dynamics of these LNTs are critical for cellular functions, supporting the transport of myriad cellular cargoes as well as communication within and between cells, and they are also widely believed to be responsible for exploitation of host cells by pathogens for the spread of infection and diseases. In vitro kinetic control of LNT formation can considerably enhance the scope of utilization of these structures for disease control and therapy. Here we report a new paradigm for spontaneous lipid nanotubulation, capturing the dynamical regimes of growth, stabilization and retraction of the tubes through the binding of synthetic nanoparticles on supported lipid bilayers (SLBs). The tubulation is determined by the spontaneous binding-unbinding of nanoparticles on the LNTs. The presented methodology could be used to rectify malfunctioning cellular tubules or to prevent the pathogenic spread of diseases through inhibition of cell-to-cell nanotubule formation.
Collapse
Affiliation(s)
| | - Koushik Debnath
- Indian Association for the Cultivation of Science, Kolkata, India
| | - Nikhil R Jana
- Indian Association for the Cultivation of Science, Kolkata, India
| | | |
Collapse
|
15
|
Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin Sci (Lond) 2022; 136:493-520. [PMID: 35415751 PMCID: PMC9008595 DOI: 10.1042/cs20210625] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/25/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Albuminuria is the hallmark of both primary and secondary proteinuric glomerulopathies, including focal segmental glomerulosclerosis (FSGS), obesity-related nephropathy, and diabetic nephropathy (DN). Moreover, albuminuria is an important feature of all chronic kidney diseases (CKDs). Podocytes play a key role in maintaining the permselectivity of the glomerular filtration barrier (GFB) and injury of the podocyte, leading to foot process (FP) effacement and podocyte loss, the unifying underlying mechanism of proteinuric glomerulopathies. The metabolic insult of hyperglycemia is of paramount importance in the pathogenesis of DN, while insults leading to podocyte damage are poorly defined in other proteinuric glomerulopathies. However, shared mechanisms of podocyte damage have been identified. Herein, we will review the role of haemodynamic and oxidative stress, inflammation, lipotoxicity, endocannabinoid (EC) hypertone, and both mitochondrial and autophagic dysfunction in the pathogenesis of the podocyte damage, focussing particularly on their role in the pathogenesis of DN. Gaining a better insight into the mechanisms of podocyte injury may provide novel targets for treatment. Moreover, novel strategies for boosting podocyte repair may open the way to podocyte regenerative medicine.
Collapse
|
16
|
Fiorentino J, Scialdone A. The role of cell geometry and cell-cell communication in gradient sensing. PLoS Comput Biol 2022; 18:e1009552. [PMID: 35286298 PMCID: PMC8963572 DOI: 10.1371/journal.pcbi.1009552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/29/2022] [Accepted: 02/17/2022] [Indexed: 11/19/2022] Open
Abstract
Cells can measure shallow gradients of external signals to initiate and accomplish a migration or a morphogenetic process. Recently, starting from mathematical models like the local-excitation global-inhibition (LEGI) model and with the support of empirical evidence, it has been proposed that cellular communication improves the measurement of an external gradient. However, the mathematical models that have been used have over-simplified geometries (e.g., they are uni-dimensional) or assumptions about cellular communication, which limit the possibility to analyze the gradient sensing ability of more complex cellular systems. Here, we generalize the existing models to study the effects on gradient sensing of cell number, geometry and of long- versus short-range cellular communication in 2D systems representing epithelial tissues. We find that increasing the cell number can be detrimental for gradient sensing when the communication is weak and limited to nearest neighbour cells, while it is beneficial when there is long-range communication. We also find that, with long-range communication, the gradient sensing ability improves for tissues with more disordered geometries; on the other hand, an ordered structure with mostly hexagonal cells is advantageous with nearest neighbour communication. Our results considerably extend the current models of gradient sensing by epithelial tissues, making a step further toward predicting the mechanism of communication and its putative mediator in many biological processes.
Collapse
Affiliation(s)
- Jonathan Fiorentino
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München; München, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München; Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München; Neuherberg, Germany
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München; München, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München; Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München; Neuherberg, Germany
| |
Collapse
|
17
|
Hunley C, Marucho M. Electrical Propagation of Condensed and Diffuse Ions Along Actin Filaments. J Comput Neurosci 2022; 50:91-107. [PMID: 34392446 PMCID: PMC8818025 DOI: 10.1007/s10827-021-00795-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 02/03/2023]
Abstract
In this article, we elucidate the roles of divalent ion condensation and highly polarized immobile water molecules on the propagation of ionic calcium waves along actin filaments. We introduced a novel electrical triple layer model and used a non-linear Debye-Huckel theory with a non-linear, dissipative, electrical transmission line model to characterize the physicochemical properties of each monomer in the filament. This characterization is carried out in terms of an electric circuit model containing monomeric flow resistances and ionic capacitances in both the condensed and diffuse layers. We considered resting and excited states of a neuron using representative mono and divalent electrolyte mixtures. Additionally, we used 0.05V and 0.15V voltage inputs to study ionic waves along actin filaments in voltage clamp experiments. Our results reveal that the physicochemical properties characterizing the condensed and diffuse layers lead to different electrical conductive mediums depending on the ionic species and the neuron state. This region specific propagation mechanism provides a more realistic avenue of delivery by way of cytoskeleton filaments for larger charged cationic species. A new direct path for transporting divalent ions might be crucial for many electrical processes found in localized neuron elements such as at mitochondria and dendritic spines.
Collapse
Affiliation(s)
- Christian Hunley
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, 78249-5003, TX, USA
| | - Marcelo Marucho
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, 78249-5003, TX, USA.
| |
Collapse
|
18
|
Mezzasalma SA, Grassi L, Grassi M. Physical and chemical properties of carbon nanotubes in view of mechanistic neuroscience investigations. Some outlook from condensed matter, materials science and physical chemistry. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112480. [PMID: 34857266 DOI: 10.1016/j.msec.2021.112480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/08/2021] [Accepted: 10/07/2021] [Indexed: 01/17/2023]
Abstract
The open border between non-living and living matter, suggested by increasingly emerging fields of nanoscience interfaced to biological systems, requires a detailed knowledge of nanomaterials properties. An account of the wide spectrum of phenomena, belonging to physical chemistry of interfaces, materials science, solid state physics at the nanoscale and bioelectrochemistry, thus is acquainted for a comprehensive application of carbon nanotubes interphased with neuron cells. This review points out a number of conceptual tools to further address the ongoing advances in coupling neuronal networks with (carbon) nanotube meshworks, and to deepen the basic issues that govern a biological cell or tissue interacting with a nanomaterial. Emphasis is given here to the properties and roles of carbon nanotube systems at relevant spatiotemporal scales of individual molecules, junctions and molecular layers, as well as to the point of view of a condensed matter or materials scientist. Carbon nanotube interactions with blood-brain barrier, drug delivery, biocompatibility and functionalization issues are also regarded.
Collapse
Affiliation(s)
- Stefano A Mezzasalma
- Ruder Bošković Institute, Materials Physics Division, Bijeniška cesta 54, 10000 Zagreb, Croatia; Lund Institute for advanced Neutron and X-ray Science (LINXS), Lund University, IDEON Building, Delta 5, Scheelevägen 19, 223 70 Lund, Sweden.
| | - Lucia Grassi
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy.
| |
Collapse
|
19
|
Concomitant and decoupled effects of cigarette smoke and SCAL1 upregulation on oncogenic phenotypes and ROS detoxification in lung adenocarcinoma cells. Sci Rep 2021; 11:18345. [PMID: 34526564 PMCID: PMC8443756 DOI: 10.1038/s41598-021-97869-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide, with smoking as its primary predisposing factor. Although carcinogens in cigarettes are known to cause oncogenic DNA alterations, analyses of patient cohorts revealed heterogeneous genetic aberrations with no clear driver mutations. The contribution of noncoding RNAs (ncRNAs) in the pathogenesis of lung cancer has since been demonstrated. Their dysregulation has been linked to cancer initiation and progression. A novel long noncoding RNA (lncRNA) called smoke and cancer-associated lncRNA 1 (SCAL1) was recently found upregulated in smoke-exposed adenocarcinomic alveolar epithelial cells. The present study characterized the phenotypic consequences of SCAL1 overexpression and knockdown using A549 cells as model system, with or without prior exposure to cigarette smoke extract (CSE). Increase in SCAL1 levels either by CSE treatment or SCAL1 overexpression led to increased cell migration, extensive cytoskeletal remodeling, and resistance to apoptosis. Further, SCAL1 levels were negatively correlated with intracellular levels of reactive oxygen species (ROS). In contrast, SCAL1 knockdown showed converse results for these assays. These results confirm the oncogenic function of SCAL1 and its role as a CSE-activated lncRNA that mediates ROS detoxification in A549 cells, thereby allowing them to develop resistance to and survive smoke-induced toxicity.
Collapse
|
20
|
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
|
21
|
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
|
22
|
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: 52] [Impact Index Per Article: 13.0] [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
|
23
|
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
|
24
|
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: 85] [Impact Index Per Article: 21.3] [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
|
25
|
Wang XT, Sun H, Chen NH, Yuan YH. Tunneling nanotubes: A novel pharmacological target for neurodegenerative diseases? Pharmacol Res 2021; 170:105541. [PMID: 33711434 DOI: 10.1016/j.phrs.2021.105541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/06/2021] [Accepted: 03/07/2021] [Indexed: 12/25/2022]
Abstract
Diversiform ways of intercellular communication are vital links in maintaining homeostasis and disseminating physiological states. Among intercellular bridges, tunneling nanotubes (TNTs) discovered in 2004 were recognized as potential pharmacology targets related to the pathogenesis of common or infrequent neurodegenerative disorders. The neurotoxic aggregates in neurodegenerative diseases including scrapie prion protein (PrPSc), mutant tau protein, amyloid-beta (Aβ) protein, alpha-synuclein (α-syn) as well as mutant Huntington (mHTT) protein could promote TNT formation via certain physiological mechanisms, in turn, mediating the intercellular transmission of neurotoxicity. In this review, we described in detail the skeleton, the formation, the physicochemical properties, and the functions of TNTs, while paying particular attention to the key role of TNTs in the transport of pathological proteins during neurodegeneration.
Collapse
Affiliation(s)
- Xiao-Tong Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Hua Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Castonguay AM, Gravel C, Lévesque M. Treating Parkinson's Disease with Antibodies: Previous Studies and Future Directions. JOURNAL OF PARKINSONS DISEASE 2021; 11:71-92. [PMID: 33104039 PMCID: PMC7990466 DOI: 10.3233/jpd-202221] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder mainly characterized by the degeneration of dopaminergic neurons in the substantia nigra. Degenerating neurons contain abnormal aggregates called Lewy bodies, that are predominantly composed of the misfolded and/or mutated alpha-synuclein protein. Post-translational modifications, cellular stress, inflammation and gene mutations are thought to trigger its pathological misfolding and aggregation. With alpha-synuclein pathology being strongly associated with dopaminergic neuronal toxicity, strategies aimed to reduce its burden are expected to be beneficial in slowing disease progression. Moreover, multiple sources of evidence suggest a cell-to-cell transmission of pathological alpha-synuclein in a prion-like manner. Therefore, antibodies targeting extra- or intracellular alpha-synuclein could be efficient in limiting the aggregation and transmission. Several active and passive immunization strategies have been explored to target alpha-synuclein. Here, we summarize immunotherapeutic approaches that were tested in pre-clinical or clinical studies in the last two decades in an attempt to treat Parkinson's disease.
Collapse
Affiliation(s)
- Anne-Marie Castonguay
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, QC, Canada
| | - Claude Gravel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, QC, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, QC, Canada
| |
Collapse
|
29
|
Intercellular Transfer of Mitochondria between Senescent Cells through Cytoskeleton-Supported Intercellular Bridges Requires mTOR and CDC42 Signalling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6697861. [PMID: 34373767 PMCID: PMC8349290 DOI: 10.1155/2021/6697861] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/31/2021] [Accepted: 06/19/2021] [Indexed: 12/23/2022]
Abstract
Cellular senescence is a state of irreversible cell proliferation arrest induced by various stressors including telomere attrition, DNA damage, and oncogene induction. While beneficial as an acute response to stress, the accumulation of senescent cells with increasing age is thought to contribute adversely to the development of cancer and a number of other age-related diseases, including neurodegenerative diseases for which there are currently no effective disease-modifying therapies. Non-cell-autonomous effects of senescent cells have been suggested to arise through the SASP, a wide variety of proinflammatory cytokines, chemokines, and exosomes secreted by senescent cells. Here, we report an additional means of cell communication utilised by senescent cells via large numbers of membrane-bound intercellular bridges-or tunnelling nanotubes (TNTs)-containing the cytoskeletal components actin and tubulin, which form direct physical connections between cells. We observe the presence of mitochondria in these TNTs and show organelle transfer through the TNTs to adjacent cells. While transport of individual mitochondria along single TNTs appears by time-lapse studies to be unidirectional, we show by differentially labelled co-culture experiments that organelle transfer through TNTs can occur between different cells of equivalent cell age, but that senescent cells, rather than proliferating cells, appear to be predominant mitochondrial donors. Using small molecule inhibitors, we demonstrate that senescent cell TNTs are dependent on signalling through the mTOR pathway, which we further show is mediated at least in part through the downstream actin-cytoskeleton regulatory factor CDC42. These findings have significant implications for the development of senomodifying therapies, as they highlight the need to account for local direct cell-cell contacts as well as the SASP in order to treat cancer and diseases of ageing in which senescence is a key factor.
Collapse
|
30
|
Auguste M, Balbi T, Ciacci C, Canesi L. Conservation of Cell Communication Systems in Invertebrate Host-Defence Mechanisms: Possible Role in Immunity and Disease. BIOLOGY 2020; 9:E234. [PMID: 32824821 PMCID: PMC7464772 DOI: 10.3390/biology9080234] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Innate immunity is continuously revealing multiple and highly conserved host-defence mechanisms. Studies on mammalian immunocytes are showing different communication systems that may play a role in coordinating innate immune responses also in invertebrates. Extracellular traps (ETs) are an immune response by which cells release net-like material, including DNA, histones and proteins. ETs are thought to immobilise and kill microorganisms, but are also involved in inflammation and autoimmune disease. Immune cells are also known to communicate through extracellular vesicles secreted in the extracellular environment or exosomes, which can carry a variety of different signalling molecules. Tunnelling nanotubes (TNTs) represent a direct cell-to-cell communication over a long distance, that allow for bi- or uni-directional transfer of cellular components between cells. Their functional role in a number of physio-pathological processes, including immune responses and pathogen transfer, has been underlined. Although ETs, exosomes, and TNTs have been described in invertebrate species, their possible role in immune responses is not fully understood. In this work, available data on these communication systems are summarised, in an attempt to provide basic information for further studies on their relevance in invertebrate immunity and disease.
Collapse
Affiliation(s)
- Manon Auguste
- Department of Earth Environment and Life Sciences (DISTAV), University of Genoa, 16136 Genoa, Italy; (M.A.); (T.B.)
| | - Teresa Balbi
- Department of Earth Environment and Life Sciences (DISTAV), University of Genoa, 16136 Genoa, Italy; (M.A.); (T.B.)
| | - Caterina Ciacci
- Department of Biomolecular Sciences (DIBS), University “Carlo Bo” of Urbino, 61029 Urbino, Italy;
| | - Laura Canesi
- Department of Earth Environment and Life Sciences (DISTAV), University of Genoa, 16136 Genoa, Italy; (M.A.); (T.B.)
| |
Collapse
|
31
|
Singhi D, Srivastava P. Role of Bacterial Cytoskeleton and Other Apparatuses in Cell Communication. Front Mol Biosci 2020; 7:158. [PMID: 32766280 PMCID: PMC7378377 DOI: 10.3389/fmolb.2020.00158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/24/2020] [Indexed: 11/13/2022] Open
Abstract
The bacterial cytoskeleton is crucial for sensing the external environment and plays a major role in cell to cell communication. There are several other apparatuses such as conjugation tubes, membrane vesicles, and nanotubes used by bacterial cells for communication. The present review article describes the various bacterial cytoskeletal proteins and other apparatuses, the physical structures they form and their role in sensing environmental stress. The implications of this cellular communication in pathogenicity are discussed.
Collapse
Affiliation(s)
| | - Preeti Srivastava
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
32
|
Affiliation(s)
- Kate E Keller
- Department of Ophthalmology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
33
|
Franchi M, Piperigkou Z, Riti E, Masola V, Onisto M, Karamanos NK. Long filopodia and tunneling nanotubes define new phenotypes of breast cancer cells in 3D cultures. Matrix Biol Plus 2020; 6-7:100026. [PMID: 33543024 PMCID: PMC7852320 DOI: 10.1016/j.mbplus.2020.100026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/20/2020] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer cell invasion into the surrounding extracellular matrix (ECM) takes place when cell-cell junctions are disrupted upon epithelial-to-mesenchymal transition (EMT). Both cancer cell-stroma and cell-cell crosstalk are essential to support the continuous tumor invasion. Cancer cells release microvesicles and exosomes containing bioactive molecules and signal peptides, which are recruited by neighboring cells or carried to distant sites, thus supporting intercellular communication and cargo transfer. Besides this indirect communication mode, cancer cells can develop cytoplasmic intercellular protrusions or tunneling nanotubes (TNTs), which allow the direct communication and molecular exchange between connected distinct cells. Using scanning electron microscopy (SEM) we show for the first time that MDA-MB-231 (high metastatic potential) and shERβ MDA-MB-231 (low metastatic potential) breast cancer cells cultured on fibronectin and collagen type I or 17β-estradiol (E2) develop TNTs and very long flexible filopodia. Interestingly, the less aggressive shERβ MDA-MB-231 cells treated with E2 in 3D collagen matrix showed the highest development of TNTs and filopodia. TNTs were often associated to adhering exosomes and microvesicles surfing from one cell to another, but no filopodia exhibited vesicle-like cytoplasmic structures on their surface. Moreover, E2 affected the expression of matrix macromolecules and cell effectors mostly in the presence of ERβ. Our novel data highlights the significance of matrix substrates and the presence of E2 and ERβ in the formation of cellular protrusion and the production of surface structures, defining novel phenotypes that unravel nodal reports for breast cancer progression.
Collapse
Key Words
- 3D, three dimensional
- Breast cancer
- CAFs, cancer-associated fibroblasts
- E2, 17β-estradiol
- ECM, extracellular matrix
- EMT, epithelial-to-mesenchymal transition
- ER, estrogen receptor
- Estrogen receptor beta
- FGF, fibroblast growth factor
- FIB-SEM, focused-ion beam scanning electron microscopy
- Filopodia
- HGF, hepatocyte growth factor
- Intercellular communication
- MMPs, matrix metalloproteinases
- SEM, scanning electron microscope
- Scanning electron microscopy
- TGFβ, transforming growth factor beta
- TNTs, tunneling nanotubes
- Tunneling nanotubes
- miRNAs, microRNAs
Collapse
Affiliation(s)
- Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Eirini Riti
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Valentina Masola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
34
|
Tunneling Nanotubes and the Eye: Intercellular Communication and Implications for Ocular Health and Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7246785. [PMID: 32352005 PMCID: PMC7171654 DOI: 10.1155/2020/7246785] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
Cellular communication is an essential process for the development and maintenance of all tissues including the eye. Recently, a new method of cellular communication has been described, which relies on formation of tubules, called tunneling nanotubes (TNTs). These structures connect the cytoplasm of adjacent cells and allow the direct transport of cellular cargo between cells without the need for secretion into the extracellular milieu. TNTs may be an important mechanism for signaling between cells that reside long distances from each other or for cells in aqueous environments, where diffusion-based signaling is challenging. Given the wide range of cargoes transported, such as lysosomes, endosomes, mitochondria, viruses, and miRNAs, TNTs may play a role in normal homeostatic processes in the eye as well as function in ocular disease. This review will describe TNT cellular communication in ocular cell cultures and the mammalian eye in vivo, the role of TNTs in mitochondrial transport with an emphasis on mitochondrial eye diseases, and molecules involved in TNT biogenesis and their function in eyes, and finally, we will describe TNT formation in inflammation, cancer, and stem cells, focusing on pathological processes of particular interest to vision scientists.
Collapse
|
35
|
Alimohamadi H, Ovryn B, Rangamani P. Modeling membrane nanotube morphology: the role of heterogeneity in composition and material properties. Sci Rep 2020; 10:2527. [PMID: 32054874 PMCID: PMC7018976 DOI: 10.1038/s41598-020-59221-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 01/27/2020] [Indexed: 01/14/2023] Open
Abstract
Membrane nanotubes are dynamic structures that may connect cells over long distances. Nanotubes are typically thin cylindrical tubes, but they may occasionally have a beaded architecture along the tube. In this paper, we study the role of membrane mechanics in governing the architecture of these tubes and show that the formation of bead-like structures along the nanotubes can result from local heterogeneities in the membrane either due to protein aggregation or due to membrane composition. We present numerical results that predict how membrane properties, protein density, and local tension compete to create a phase space that governs the morphology of a nanotube. We also find that there exists a discontinuity in the energy that impedes two beads from fusing. These results suggest that the membrane-protein interaction, membrane composition, and membrane tension closely govern the tube radius, number of beads, and the bead morphology.
Collapse
Affiliation(s)
- Haleh Alimohamadi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, 92093, USA
| | - Ben Ovryn
- Department of Physics, New York Institute of Technology, New York, NY, 11568, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, 92093, USA.
| |
Collapse
|
36
|
Sun YY, Bradley JM, Keller KE. Phenotypic and Functional Alterations in Tunneling Nanotubes Formed by Glaucomatous Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2020; 60:4583-4595. [PMID: 31675075 PMCID: PMC6827425 DOI: 10.1167/iovs.19-28084] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Trabecular meshwork (TM) cells detect and coordinate responses to intraocular pressure (IOP) in the eye. TM cells become dysfunctional in glaucoma where IOP is often elevated. Recently, we showed that normal TM (NTM) cells communicate by forming tubular connections called tunneling nanotubes (TNTs). Here, we investigated TNTs in glaucomatous TM (GTM) cells. Methods Primary GTM and NTM cells were established from cadaver eyes. Transfer of Vybrant DiO and DiD-labeled vesicles via TNT connections was measured. Imaris software measured the number and length of cell protrusions from immunofluorescent confocal images. Live-cell imaging of the actin cytoskeleton was performed. The distribution of myosin-X, a regulator of TNTs/filopodia, was investigated in TM cells and tissue. Results GTM cells contained significantly more transferred fluorescent vesicles than NTM cells (49.6% vs. 35%). Although NTM cells had more protrusions at the cell surface than GTM cells (7.61 vs. 4.65 protrusions/cell), GTM protrusions were significantly longer (12.1 μm vs. 9.76 μm). Live-cell imaging demonstrated that the GTM actin cytoskeleton was less dynamic, and vesicle transfer between cells was significantly slower than NTM cells. Furthermore, rearrangement of the actin cortex adjacent to the TNT may influence TNT formation. Myosin-X immunostaining was punctate and disorganized in GTM cells and tissue compared to age-matched NTM controls. Conclusions Together, our data demonstrate that GTM cells have phenotypic and functional differences in their TNTs. Significantly slower vesicle transfer via TNTs in GTM cells may delay the timely propagation of cellular signals when pressures become elevated in glaucoma.
Collapse
Affiliation(s)
- Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - John M Bradley
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
37
|
Korenkova O, Pepe A, Zurzolo C. Fine intercellular connections in development: TNTs, cytonemes, or intercellular bridges? Cell Stress 2020; 4:30-43. [PMID: 32043076 PMCID: PMC6997949 DOI: 10.15698/cst2020.02.212] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Intercellular communication is a fundamental property of multicellular organisms, necessary for their adequate responses to changing environment. Tunneling nanotubes (TNTs) represent a novel means of intercellular communication being a long cell-to-cell conduit. TNTs are actively formed under a broad range of stresses and are also proposed to exist under physiological conditions. Development is a physiological condition of particular interest, as it requires fine coordination. Here we discuss whether protrusions shown to exist during embryonic development of different species could be TNTs or if they represent other types of cell structure, like cytonemes or intercellular bridges, that are suggested to play an important role in development.
Collapse
Affiliation(s)
- Olga Korenkova
- Unit of Membrane Traffic and Pathogenesis, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France.,Université Paris-Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Anna Pepe
- Unit of Membrane Traffic and Pathogenesis, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France
| | - Chiara Zurzolo
- Unit of Membrane Traffic and Pathogenesis, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
38
|
Whitehead J, Zhang J, Harvestine JN, Kothambawala A, Liu GY, Leach JK. Tunneling nanotubes mediate the expression of senescence markers in mesenchymal stem/stromal cell spheroids. Stem Cells 2020; 38:80-89. [PMID: 31298767 PMCID: PMC6954984 DOI: 10.1002/stem.3056] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2019] [Accepted: 06/25/2019] [Indexed: 02/06/2023]
Abstract
The therapeutic potential of mesenchymal stem/stromal cells (MSCs) is limited by acquired senescence following prolonged culture expansion and high-passage numbers. However, the degree of cell senescence is dynamic, and cell-cell communication is critical to promote cell survival. MSC spheroids exhibit improved viability compared with monodispersed cells, and actin-rich tunneling nanotubes (TNTs) may mediate cell survival and other functions through the exchange of cytoplasmic components. Building upon our previous demonstration of TNTs bridging MSCs within these cell aggregates, we hypothesized that TNTs would influence the expression of senescence markers in MSC spheroids. We confirmed the existence of functional TNTs in MSC spheroids formed from low-passage, high-passage, and mixtures of low- and high-passage cells using scanning electron microscopy, confocal microscopy, and flow cytometry. The contribution of TNTs toward the expression of senescence markers was investigated by blocking TNT formation with cytochalasin D (CytoD), an inhibitor of actin polymerization. CytoD-treated spheroids exhibited decreases in cytosol transfer. Compared with spheroids formed solely of high-passage MSCs, the addition of low-passage MSCs reduced p16 expression, a known genetic marker of senescence. We observed a significant increase in p16 expression in high-passage cells when TNT formation was inhibited, establishing the importance of TNTs in MSC spheroids. These data confirm the restorative role of TNTs within MSC spheroids formed with low- and high-passage cells and represent an exciting approach to use higher-passage cells in cell-based therapies.
Collapse
Affiliation(s)
- Jacklyn Whitehead
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Jiali Zhang
- Department of Chemistry, University of California, Davis, CA 95616
| | - Jenna N. Harvestine
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Alefia Kothambawala
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Gang-yu Liu
- Department of Chemistry, University of California, Davis, CA 95616
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817
| |
Collapse
|
39
|
Atkinson SP. A preview of selected articles. Stem Cells 2020. [DOI: 10.1002/stem.3138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
40
|
Matejka N, Reindl J. Perspectives of cellular communication through tunneling nanotubes in cancer cells and the connection to radiation effects. Radiat Oncol 2019; 14:218. [PMID: 31796110 PMCID: PMC6889217 DOI: 10.1186/s13014-019-1416-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Direct cell-to-cell communication is crucial for the survival of cells in stressful situations such as during or after radiation exposure. This communication can lead to non-targeted effects, where non-treated or non-infected cells show effects induced by signal transduction from non-healthy cells or vice versa. In the last 15 years, tunneling nanotubes (TNTs) were identified as membrane connections between cells which facilitate the transfer of several cargoes and signals. TNTs were identified in various cell types and serve as promoter of treatment resistance e.g. in chemotherapy treatment of cancer. Here, we discuss our current understanding of how to differentiate tunneling nanotubes from other direct cellular connections and their role in the stress reaction of cellular networks. We also provide a perspective on how the capability of cells to form such networks is related to the ability to surpass stress and how this can be used to study radioresistance of cancer cells.
Collapse
Affiliation(s)
- Nicole Matejka
- Institut für angewandte Physik und Messtechnik, Universität der Bundeswehr München, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Judith Reindl
- Institut für angewandte Physik und Messtechnik, Universität der Bundeswehr München, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| |
Collapse
|
41
|
Vargas JY, Loria F, Wu Y, Córdova G, Nonaka T, Bellow S, Syan S, Hasegawa M, van Woerden GM, Trollet C, Zurzolo C. The Wnt/Ca 2+ pathway is involved in interneuronal communication mediated by tunneling nanotubes. EMBO J 2019; 38:e101230. [PMID: 31625188 PMCID: PMC6885744 DOI: 10.15252/embj.2018101230] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 01/15/2023] Open
Abstract
Tunneling nanotubes (TNTs) are actin-based transient tubular connections that allow direct communication between distant cells. TNTs play an important role in several physiological (development, immunity, and tissue regeneration) and pathological (cancer, neurodegeneration, and pathogens transmission) processes. Here, we report that the Wnt/Ca2+ pathway, an intracellular cascade that is involved in actin cytoskeleton remodeling, has a role in TNT formation and TNT-mediated transfer of cargoes. Specifically, we found that Ca2+ /calmodulin-dependent protein kinase II (CaMKII), a transducer of the Wnt/Ca2+ pathway, regulates TNTs in a neuronal cell line and in primary neurons. We identified the β isoform of CaMKII as a key molecule in modulating TNT formation and transfer, showing that this depends on the actin-binding activity of the protein. Finally, we found that the transfer of vesicles and aggregated α-synuclein between primary neurons can be regulated by the activation of the Wnt/Ca2+ pathway. Our findings suggest that Wnt/Ca2+ pathway could be a novel promising target for therapies designed to impair TNT-mediated propagation of pathogens.
Collapse
Affiliation(s)
- Jessica Y Vargas
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Frida Loria
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
- Present address:
Centro de Biología Molecular Severo Ochoa (CSIC‐UAM)Departamento de Biología MolecularUniversidad Autónoma de MadridMadridSpain
| | - Yuan‐Ju Wu
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Gonzalo Córdova
- Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieCentre de Recherche en MyologieUMRS974Sorbonne UniversitéParisFrance
| | - Takashi Nonaka
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | | | - Sylvie Syan
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Masato Hasegawa
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Geeske M van Woerden
- Department of NeuroscienceErasmus Medical CenterRotterdamThe Netherlands
- ENCORE Expertise Center for Neurodevelopmental DisordersErasmus Medical CenterRotterdamThe Netherlands
| | - Capucine Trollet
- Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieCentre de Recherche en MyologieUMRS974Sorbonne UniversitéParisFrance
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| |
Collapse
|
42
|
Chabanon M, Rangamani P. Geometric coupling of helicoidal ramps and curvature-inducing proteins in organelle membranes. J R Soc Interface 2019; 16:20190354. [PMID: 31480932 DOI: 10.1098/rsif.2019.0354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cellular membranes display an incredibly diverse range of shapes, both in the plasma membrane and at membrane bound organelles. These morphologies are intricately related to cellular functions, enabling and regulating fundamental membrane processes. However, the biophysical mechanisms at the origin of these complex geometries are not fully understood from the standpoint of membrane-protein coupling. In this study, we focused on a minimal model of helicoidal ramps representative of specialized endoplasmic reticulum compartments. Given a helicoidal membrane geometry, we asked what is the distribution of spontaneous curvature required to maintain this shape at mechanical equilibrium? Based on the Helfrich energy of elastic membranes with spontaneous curvature, we derived the shape equation for minimal surfaces, and applied it to helicoids. We showed the existence of switches in the sign of the spontaneous curvature associated with geometric variations of the membrane structures. Furthermore, for a prescribed gradient of spontaneous curvature along the exterior boundaries, we identified configurations of the helicoidal ramps that are confined between two infinitely large energy barriers. Overall our results suggest possible mechanisms for geometric control of helicoidal ramps in membrane organelles based on curvature-inducing proteins.
Collapse
Affiliation(s)
- Morgan Chabanon
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla CA, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla CA, USA
| |
Collapse
|
43
|
Stress-induced tunneling nanotubes support treatment adaptation in prostate cancer. Sci Rep 2019; 9:7826. [PMID: 31127190 PMCID: PMC6534589 DOI: 10.1038/s41598-019-44346-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
Tunneling nanotubes (TNTs) are actin-based membranous structures bridging distant cells for intercellular communication. We define roles for TNTs in stress adaptation and treatment resistance in prostate cancer (PCa). Androgen receptor (AR) blockade and metabolic stress induce TNTs, but not in normal prostatic epithelial or osteoblast cells. Co-culture assays reveal enhanced TNT formation between stressed and unstressed PCa cells as well as from stressed PCa to osteoblasts. Stress-induced chaperones clusterin and YB-1 localize within TNTs, are transported bi-directionally via TNTs and facilitate TNT formation in PI3K/AKT and Eps8-dependent manner. AR variants, induced by AR antagonism to mediate resistance to AR pathway inhibition, also enhance TNT production and rescue loss of clusterin- or YB-1-repressed TNT formation. TNT disruption sensitizes PCa to treatment-induced cell death. These data define a mechanistic network involving stress induction of chaperone and AR variants, PI3K/AKT signaling, actin remodeling and TNT-mediated intercellular communication that confer stress adaptative cell survival.
Collapse
|
44
|
Effect of tolytoxin on tunneling nanotube formation and function. Sci Rep 2019; 9:5741. [PMID: 30952909 PMCID: PMC6450976 DOI: 10.1038/s41598-019-42161-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 03/22/2019] [Indexed: 12/20/2022] Open
Abstract
Tunneling nanotubes (TNTs) are actin-containing membrane protrusions that play an essential role in long-range intercellular communication. They are involved in development of various diseases by allowing transfer of pathogens or protein aggregates as well as organelles such as mitochondria. Increase in TNT formation has been linked to many pathological conditions. Here we show that nM concentrations of tolytoxin, a cyanobacterial macrolide that targets actin by inhibition of its polymerization, significantly decrease the number of TNT-connected cells, as well as transfer of mitochondria and α-synuclein fibrils in two different cell lines of neuronal (SH-SY5Y) and epithelial (SW13) origin. As the cytoskeleton of the tested cell remain preserved, this macrolide could serve as a valuable tool for future therapies against diseases propagated by TNTs.
Collapse
|
45
|
Zhang J, Zhang J, Zhao L, Xin Y, Liu S, Cui W. Differential roles of microtubules in the two formation stages of membrane nanotubes between human mesenchymal stem cells and neonatal mouse cardiomyocytes. Biochem Biophys Res Commun 2019; 512:441-447. [PMID: 30904163 DOI: 10.1016/j.bbrc.2019.03.075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 01/27/2023]
Abstract
Membrane nanotubes (MNTs) are a kind of novel way for communication between two distant cells. It was recently shown that MNTs can be formed between distressed cardiomyocytes (CMs) and mesenchymal stem cells (MSCs). As a cytoskeleton-containing structure, the role of microtubules in MNTs is not fully understood. Here, we investigated this question. By membrane dye staining, we found that the numbers of MNTs between human MSCs (hMSCs) and distressed neonatal mouse CMs (NMCMs) increased gradually from 3 to 16 h and remained constant from 16 to 30 h, which were identified as active formation stage (the 1st stage, ≤16 h in coculture), and mature and stable stage (the 2nd stage, >16 h in coculture), respectively. In the 1st stage, more MNTs originated from hMSCs, whereas more MNTs originated from NMCMs in the 2nd stage. The formation of MNTs was affected when microtubules were disrupted by nocodazole in the 1st stage, but not in the 2nd stage. MNTs became shorter and thinner when microtubules were disrupted in the 2nd stage. Immunofluorescence staining and flow cytometry showed that mitochondria in hMSCs were transported into distressed NMCMs, which was suppressed by nocodazole in the 2nd stage. Tunnel staining showed that hypoxia/reoxygenation-induced apoptosis of NMCMs only in the 2nd stage could be rescued by direct, but not indirect, coculture with hMSCs. This rescue function was weakened when the mitochondrial functions of cocultured hMSCs were disrupted by EtBr or microtubules in cocultures were disrupted by nocodazole. All these results suggested that there are two stages for MNT formation, and microtubules played differential roles in the two stages: During the 1st stage, microtubules were required for MNT formation, whereas during the 2nd stage, microtubules were related to the morphological features of MNTs and played a key role in anti-apoptosis of MNTs by mitochondrial transfer.
Collapse
Affiliation(s)
- Jianghui Zhang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Jing Zhang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Limin Zhao
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Yi Xin
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Sa Liu
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Wei Cui
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| |
Collapse
|
46
|
A novel Microproteomic Approach Using Laser Capture Microdissection to Study Cellular Protrusions. Int J Mol Sci 2019; 20:ijms20051172. [PMID: 30866487 PMCID: PMC6429397 DOI: 10.3390/ijms20051172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 12/28/2022] Open
Abstract
Cell–cell communication is vital to multicellular organisms, and distinct types of cellular protrusions play critical roles during development, cell signaling, and the spreading of pathogens and cancer. The differences in the structure and protein composition of these different types of protrusions and their specific functions have not been elucidated due to the lack of a method for their specific isolation and analysis. In this paper, we described, for the first time, a method to specifically isolate distinct protrusion subtypes, based on their morphological structures or fluorescent markers, using laser capture microdissection (LCM). Combined with a unique fixation and protein extraction protocol, we pushed the limits of microproteomics and demonstrate that proteins from LCM-isolated protrusions can successfully and reproducibly be identified by mass spectrometry using ultra-high field Orbitrap technologies. Our method confirmed that different types of protrusions have distinct proteomes and it promises to advance the characterization and the understanding of these unique structures to shed light on their possible role in health and disease.
Collapse
|
47
|
Daniels DR. Transport of solid bodies along tubular membrane tethers. PLoS One 2019; 14:e0210259. [PMID: 30650122 PMCID: PMC6334941 DOI: 10.1371/journal.pone.0210259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 12/19/2018] [Indexed: 11/18/2022] Open
Abstract
We study the crucial role of membrane fluctuations in maintaining a narrow gap between a fluid membrane tube and an enclosed solid particle. Solvent flows can occur in this gap, hence giving rise to a finite particle mobility along the tube. While our study has relevance for how cells are able to transport large organelles or other cargo along connecting membrane tubes, known as tunneling nanotubes, our calculations are also framed so that they can be tested by a specific in vitro experiment: A tubular membrane tether can be pulled from a membrane reservoir, such as an aspirated Giant Unilamellar Vesicle (GUV), e.g. using a conjugated bead that binds to the membrane and is held in a laser trap. We compute the subsequent mobility of colloidal particles trapped in the tube, focusing on the case when the particle is large compared to the equilibrium tube radius. We predict that the particle mobility should scale as ∼ σ−2/3, with σ the membrane tension.
Collapse
Affiliation(s)
- D. R. Daniels
- College of Engineering, Swansea University, Bay Campus, Swansea, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Reindl J, Shevtsov M, Dollinger G, Stangl S, Multhoff G. Membrane Hsp70-supported cell-to-cell connections via tunneling nanotubes revealed by live-cell STED nanoscopy. Cell Stress Chaperones 2019; 24:213-221. [PMID: 30632067 PMCID: PMC6363613 DOI: 10.1007/s12192-018-00958-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/19/2018] [Accepted: 12/03/2018] [Indexed: 01/19/2023] Open
Abstract
Heat shock protein Hsp70 (Hsp70) is found on the cell surface of a large variety of human and mouse tumor cell types including U87, GL261 glioblastoma, and 4T1 mammary carcinoma cells. We studied the role of membrane-bound Hsp70 (mHsp70) in the formation of cell-to-cell connections via tunneling nanotubes (TNTs) using live-cell STED nanoscopy. This technique allows the visualization of microstructures in the 100-nm range in the living cells. We could show that the presence of tumor-derived mHsp70 in TNTs with a diameter ranging from 120 to 140 nm predominantly originates from cholesterol-rich-microdomains containing the lipid compound globoyltriaosylceramide (Gb3). Under non-stress conditions, Hsp70 and Gb3 are structurally clustered in the membrane of TNTs of tumor cells that showed tumor type specific variations in the amount of cell-to-cell connection networks. Furthermore depletion of cholesterol and ionizing radiation as a stress factor results in a complete loss of Hsp70-containing TNTs.
Collapse
Affiliation(s)
- Judith Reindl
- Universität der Bundeswehr München, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Maxim Shevtsov
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technischen Universität München (TUM), Ismaningerstrasse 22, 81675 Munich, Germany
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky ave. 4, St. Petersburg, 194064 Russia
- Pavlov First Saint Petersburg State Medical University, L’va Tolstogo str. 6/8, St. Petersburg, 197022 Russia
- Polenov Russian Scientific Research Institute of Neurosurgery, Mayakovskogo str. 12, St. Petersburg, 191104 Russia
| | - Günther Dollinger
- Universität der Bundeswehr München, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| | - Stefan Stangl
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technischen Universität München (TUM), Ismaningerstrasse 22, 81675 Munich, Germany
| | - Gabriele Multhoff
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technischen Universität München (TUM), Ismaningerstrasse 22, 81675 Munich, Germany
| |
Collapse
|
49
|
Jash E, Prasad P, Kumar N, Sharma T, Goldman A, Sehrawat S. Perspective on nanochannels as cellular mediators in different disease conditions. Cell Commun Signal 2018; 16:76. [PMID: 30409198 PMCID: PMC6222982 DOI: 10.1186/s12964-018-0281-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/08/2018] [Indexed: 01/05/2023] Open
Abstract
Tunnelling nanotubes (TNTs), also known as membrane nanochannels, are actin-based structures that facilitate cytoplasmic connections for rapid intercellular transfer of signals, organelles and membrane components. These dynamic TNTs can form de novo in animal cells and establish complex intercellular networks between distant cells up to 150 μm apart. Within the last decade, TNTs have been discovered in different cell types including tumor cells, macrophages, monocytes, endothelial cells and T cells. It has also been further elucidated that these nanotubes play a vital role in diseased conditions such as cancer, where TNT formation occurs at a higher pace and is used for rapid intercellular modulation of chemo-resistance. Viruses such as HIV, HSV and prions also hijack the existing TNT connections between host cells for rapid transmission and evasion of the host immune responses. The following review aims to describe the heterogeneity of TNTs, their role in different tissues and disease conditions in order to enhance our understanding on how these nanotubes can be used as a target for therapies.
Collapse
Affiliation(s)
- Eshna Jash
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Peeyush Prasad
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Naveen Kumar
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Taruna Sharma
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Aaron Goldman
- Mitra Biotech, Integrative Immuno-Oncology Center, Woburn, MA, 01801, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA. .,Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| | - Seema Sehrawat
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India. .,Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
50
|
Charles-Orszag A, Tsai FC, Bonazzi D, Manriquez V, Sachse M, Mallet A, Salles A, Melican K, Staneva R, Bertin A, Millien C, Goussard S, Lafaye P, Shorte S, Piel M, Krijnse-Locker J, Brochard-Wyart F, Bassereau P, Duménil G. Adhesion to nanofibers drives cell membrane remodeling through one-dimensional wetting. Nat Commun 2018; 9:4450. [PMID: 30361638 PMCID: PMC6202395 DOI: 10.1038/s41467-018-06948-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/19/2018] [Indexed: 01/22/2023] Open
Abstract
The shape of cellular membranes is highly regulated by a set of conserved mechanisms that can be manipulated by bacterial pathogens to infect cells. Remodeling of the plasma membrane of endothelial cells by the bacterium Neisseria meningitidis is thought to be essential during the blood phase of meningococcal infection, but the underlying mechanisms are unclear. Here we show that plasma membrane remodeling occurs independently of F-actin, along meningococcal type IV pili fibers, by a physical mechanism that we term 'one-dimensional' membrane wetting. We provide a theoretical model that describes the physical basis of one-dimensional wetting and show that this mechanism occurs in model membranes interacting with nanofibers, and in human cells interacting with extracellular matrix meshworks. We propose one-dimensional wetting as a new general principle driving the interaction of cells with their environment at the nanoscale that is diverted by meningococci during infection.
Collapse
Affiliation(s)
- Arthur Charles-Orszag
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, 75006, France
| | - Feng-Ching Tsai
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | - Daria Bonazzi
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France
| | - Valeria Manriquez
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, 75006, France
| | | | | | | | - Keira Melican
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.,Department of Neuroscience, Swedish Medical Nanoscience Center, Karolinska Institutet, Solna, 171 77, Sweden
| | - Ralitza Staneva
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, 75005, France
| | - Aurélie Bertin
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | | | - Sylvie Goussard
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France
| | - Pierre Lafaye
- Antibody Engineering, Institut Pasteur, Paris, 75015, France
| | | | - Matthieu Piel
- Systems Biology of Cell Polarity and Cell Division, Institut Pierre-Gilles De Gennes, Paris, 75005, France.,Institut Curie, Paris, 75005, France
| | | | - Françoise Brochard-Wyart
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | - Guillaume Duménil
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.
| |
Collapse
|