1
|
Vanherle S, Loix M, Miron VE, Hendriks JJA, Bogie JFJ. Lipid metabolism, remodelling and intercellular transfer in the CNS. Nat Rev Neurosci 2025; 26:214-231. [PMID: 39972160 DOI: 10.1038/s41583-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Lipid metabolism encompasses the catabolism and anabolism of lipids, and is fundamental for the maintenance of cellular homeostasis, particularly within the lipid-rich CNS. Increasing evidence further underscores the importance of lipid remodelling and transfer within and between glial cells and neurons as key orchestrators of CNS lipid homeostasis. In this Review, we summarize and discuss the complex landscape of processes involved in lipid metabolism, remodelling and intercellular transfer in the CNS. Highlighted are key pathways, including those mediating lipid (and lipid droplet) biogenesis and breakdown, lipid oxidation and phospholipid metabolism, as well as cell-cell lipid transfer mediated via lipoproteins, extracellular vesicles and tunnelling nanotubes. We further explore how the dysregulation of these pathways contributes to the onset and progression of neurodegenerative diseases, and examine the homeostatic and pathogenic impacts of environment, diet and lifestyle on CNS lipid metabolism.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Veronique E Miron
- Keenan Research Centre for Biomedical Science and Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.
- University MS Centre, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
2
|
Korenkova O, Liu S, Prlesi I, Pepe A, Albadri S, Del Bene F, Zurzolo C. Tunneling nanotubes enable intercellular transfer in zebrafish embryos. Dev Cell 2025; 60:524-534.e3. [PMID: 39541978 DOI: 10.1016/j.devcel.2024.10.016] [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: 09/12/2023] [Revised: 06/25/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Tunneling nanotubes (TNTs) are thin intercellular connections that facilitate the transport of diverse cargoes, ranging from ions to organelles. While TNT studies have predominantly been conducted in cell cultures, the existence of open-ended TNTs within live organisms remains unverified. Despite the observation of intercellular connections during embryonic development across various species, their functional role in facilitating material transfer between connected cells has not been confirmed. In this study, we performed mosaic labeling of gastrula cells in zebrafish embryos to demonstrate the coexistence of TNT-like structures alongside other cellular protrusions. These embryonic TNT-like connections exhibited a morphology similar to that of TNTs described in cell culture, appeared to have similar formation mechanisms, and could be induced by Eps8 overexpression and CK666 treatment. Most notably, we demonstrated their capability to transfer both soluble cargoes and organelles, thus confirming their open-endedness. This study demonstrates the existence of functional, open-ended TNTs in a living embryo.
Collapse
Affiliation(s)
- Olga Korenkova
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, 75015 Paris, France; Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Shiyu Liu
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, 75015 Paris, France; Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Inès Prlesi
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, 75015 Paris, France
| | - Anna Pepe
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, 75015 Paris, France
| | - Shahad Albadri
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, 75015 Paris, France.
| |
Collapse
|
3
|
Nakano T, Irie K, Matsuo K, Mishima K, Nakamura Y. Molecular and cellular mechanisms of mitochondria transfer in models of central nervous system disease. J Cereb Blood Flow Metab 2024:271678X241300223. [PMID: 39539186 PMCID: PMC11565516 DOI: 10.1177/0271678x241300223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/01/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
In the central nervous system (CNS), neuronal function and dysfunction are critically dependent on mitochondrial integrity and activity. In damaged or diseased brains, mitochondrial dysfunction reduces adenosine triphosphate (ATP) levels and impairs ATP-dependent neural firing and neurotransmitter dynamics. Restoring mitochondrial capacity to generate ATP may be fundamental in restoring neuronal function. Recent studies in animals and humans have demonstrated that endogenous mitochondria may be released into the extracellular environment and transported or exchanged between cells in the CNS. Under pathological conditions in the CNS, intercellular mitochondria transfer contributes to new classes of signaling and multifunctional cellular activities, thereby triggering deleterious effects or promoting beneficial responses. Therefore, to take full advantage of the beneficial effects of mitochondria, it may be useful to transplant healthy and viable mitochondria into damaged tissues. In this review, we describe recent findings on the mechanisms of mitochondria transfer and provide an overview of experimental methodologies, including tissue sourcing, mitochondrial isolation, storage, and modification, aimed at optimizing mitochondria transplantation therapy for CNS disorders. Additionally, we examine the clinical relevance and potential strategies for the therapeutic application of mitochondria transplantation.
Collapse
Affiliation(s)
- Takafumi Nakano
- Department of Oncology and Infectious Disease Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
- Department of Physiology and Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Keiichi Irie
- Department of Physiology and Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Koichi Matsuo
- Department of Oncology and Infectious Disease Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kenichi Mishima
- Department of Physiology and Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Yoshihiko Nakamura
- Department of Emergency and Critical Care Medicine, Fukuoka University Hospital, Fukuoka, Japan
| |
Collapse
|
4
|
Kotarba S, Kozłowska M, Scios M, Saramowicz K, Barczuk J, Granek Z, Siwecka N, Wiese W, Golberg M, Galita G, Sychowski G, Majsterek I, Rozpędek-Kamińska W. Potential Mechanisms of Tunneling Nanotube Formation and Their Role in Pathology Spread in Alzheimer's Disease and Other Proteinopathies. Int J Mol Sci 2024; 25:10797. [PMID: 39409126 PMCID: PMC11477428 DOI: 10.3390/ijms251910797] [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: 09/10/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia worldwide. The etiopathogenesis of this disease remains unknown. Currently, several hypotheses attempt to explain its cause, with the most well-studied being the cholinergic, beta-amyloid (Aβ), and Tau hypotheses. Lately, there has been increasing interest in the role of immunological factors and other proteins such as alpha-synuclein (α-syn) and transactive response DNA-binding protein of 43 kDa (TDP-43). Recent studies emphasize the role of tunneling nanotubes (TNTs) in the spread of pathological proteins within the brains of AD patients. TNTs are small membrane protrusions composed of F-actin that connect non-adjacent cells. Conditions such as pathogen infections, oxidative stress, inflammation, and misfolded protein accumulation lead to the formation of TNTs. These structures have been shown to transport pathological proteins such as Aβ, Tau, α-syn, and TDP-43 between central nervous system (CNS) cells, as confirmed by in vitro studies. Besides their role in spreading pathology, TNTs may also have protective functions. Neurons burdened with α-syn can transfer protein aggregates to glial cells and receive healthy mitochondria, thereby reducing cellular stress associated with α-syn accumulation. Current AD treatments focus on alleviating symptoms, and clinical trials with Aβ-lowering drugs have proven ineffective. Therefore, intensifying research on TNTs could bring scientists closer to a better understanding of AD and the development of effective therapies.
Collapse
Affiliation(s)
- Szymon Kotarba
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Marta Kozłowska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Małgorzata Scios
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Julia Barczuk
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Zuzanna Granek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Michał Golberg
- Department of Histology and Embryology, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Grzegorz Sychowski
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| |
Collapse
|
5
|
Notario Manzano R, Chaze T, Rubinstein E, Penard E, Matondo M, Zurzolo C, Brou C. Proteomic landscape of tunneling nanotubes reveals CD9 and CD81 tetraspanins as key regulators. eLife 2024; 13:RP99172. [PMID: 39250349 PMCID: PMC11383530 DOI: 10.7554/elife.99172] [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] [Indexed: 09/11/2024] Open
Abstract
Tunneling nanotubes (TNTs) are open actin- and membrane-based channels, connecting remote cells and allowing direct transfer of cellular material (e.g. vesicles, mRNAs, protein aggregates) from the cytoplasm to the cytoplasm. Although they are important especially, in pathological conditions (e.g. cancers, neurodegenerative diseases), their precise composition and their regulation were still poorly described. Here, using a biochemical approach allowing to separate TNTs from cell bodies and from extracellular vesicles and particles (EVPs), we obtained the full composition of TNTs compared to EVPs. We then focused on two major components of our proteomic data, the CD9 and CD81 tetraspanins, and further investigated their specific roles in TNT formation and function. We show that these two tetraspanins have distinct non-redundant functions: CD9 participates in stabilizing TNTs, whereas CD81 expression is required to allow the functional transfer of vesicles in the newly formed TNTs, possibly by regulating docking to or fusion with the opposing cell.
Collapse
Affiliation(s)
- Roberto Notario Manzano
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and Infection, CNRS 18 UMR 3691, Institut Pasteur, Université Paris Cité, Paris, France
- Sorbonne Université, ED394 - Physiologie, Physiopathologie et Thérapeutique, Paris, France
| | - Thibault Chaze
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut Pasteur, Paris, France
| | - Eric Rubinstein
- Centre d'Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, CIMI-Paris, Paris, France
| | - Esthel Penard
- Ultrastructural BioImaging Core Facility (UBI), C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut Pasteur, Paris, France
| | - Chiara Zurzolo
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and Infection, CNRS 18 UMR 3691, Institut Pasteur, Université Paris Cité, Paris, France
| | - Christel Brou
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and Infection, CNRS 18 UMR 3691, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
6
|
Liu E, Zhang Y, Wang JZ. Updates in Alzheimer's disease: from basic research to diagnosis and therapies. Transl Neurodegener 2024; 13:45. [PMID: 39232848 PMCID: PMC11373277 DOI: 10.1186/s40035-024-00432-x] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized pathologically by extracellular deposition of β-amyloid (Aβ) into senile plaques and intracellular accumulation of hyperphosphorylated tau (pTau) as neurofibrillary tangles. Clinically, AD patients show memory deterioration with varying cognitive dysfunctions. The exact molecular mechanisms underlying AD are still not fully understood, and there are no efficient drugs to stop or reverse the disease progression. In this review, we first provide an update on how the risk factors, including APOE variants, infections and inflammation, contribute to AD; how Aβ and tau become abnormally accumulated and how this accumulation plays a role in AD neurodegeneration. Then we summarize the commonly used experimental models, diagnostic and prediction strategies, and advances in periphery biomarkers from high-risk populations for AD. Finally, we introduce current status of development of disease-modifying drugs, including the newly officially approved Aβ vaccines, as well as novel and promising strategies to target the abnormal pTau. Together, this paper was aimed to update AD research progress from fundamental mechanisms to the clinical diagnosis and therapies.
Collapse
Affiliation(s)
- Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yao Zhang
- Department of Endocrine, Liyuan Hospital, Key Laboratory of Ministry of Education for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Jian-Zhi Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
7
|
Nguyen DLB, Okolicsanyi RK, Haupt LM. Heparan sulfate proteoglycans: Mediators of cellular and molecular Alzheimer's disease pathogenic factors via tunnelling nanotubes? Mol Cell Neurosci 2024; 129:103936. [PMID: 38750678 DOI: 10.1016/j.mcn.2024.103936] [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: 03/07/2024] [Revised: 04/14/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Neurological disorders impact around one billion individuals globally (15 % approx.), with significant implications for disability and mortality with their impact in Australia currently amounts to 6.8 million deaths annually. Heparan sulfate proteoglycans (HSPGs) are complex extracellular molecules implicated in promoting Tau fibril formation resulting in Tau tangles, a hallmark of Alzheimer's disease (AD). HSPG-Tau protein interactions contribute to various AD stages via aggregation, toxicity, and clearance, largely via interactions with the glypican 1 and syndecan 3 core proteins. The tunnelling nanotubes (TNTs) pathway is emerging as a facilitator of intercellular molecule transport, including Tau and Amyloid β proteins, across extensive distances. While current TNT-associated evidence primarily stems from cancer models, their role in Tau propagation and its effects on recipient cells remain unclear. This review explores the interplay of TNTs, HSPGs, and AD-related factors and proposes that HSPGs influence TNT formation in neurodegenerative conditions such as AD.
Collapse
Affiliation(s)
- Duy L B Nguyen
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, Queensland 4059, Australia
| | - Rachel K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, Queensland 4059, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia
| | - Larisa M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, Queensland 4059, Australia; Centre for Biomedical Technologies, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD 4059, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia; Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices, Queensland University of Technology (QUT), Australia.
| |
Collapse
|
8
|
Hu J, Wang L, Chen J, Liang Y. Construction of a cell-based aggregation and seeding model for the Tau protein. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1085-1088. [PMID: 38682159 PMCID: PMC11322869 DOI: 10.3724/abbs.2024057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Affiliation(s)
- Jiying Hu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072China
- Office of Core FacilityShenzhen Bay LaboratoryShenzhen518000China
| | - Liqiang Wang
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072China
| | - Jie Chen
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072China
| | - Yi Liang
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072China
| |
Collapse
|
9
|
Zhou C, Huang M, Wang S, Chu S, Zhang Z, Chen N. Tunneling nanotubes: The transport highway for astrocyte-neuron communication in the central nervous system. Brain Res Bull 2024; 209:110921. [PMID: 38447659 DOI: 10.1016/j.brainresbull.2024.110921] [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: 01/02/2024] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Tunneling nanotubes (TNTs) have emerged as pivotal structures for intercellular communication, enabling the transfer of cellular components across distant cells. Their involvement in neurological disorders has attracted considerable scientific interest. This review delineates the functions of TNTs within the central nervous system, examining their role in the transmission of bioenergetic substrates, and signaling molecules, and their multifaceted impact on both physiological and pathological processes, with an emphasis on neurodegenerative diseases. The review highlights the selectivity and specificity of TNTs as dedicated pathways for intercellular cargo delivery, particularly under stress conditions that provoke increased TNT formation. The potential of TNTs as therapeutic targets is explored in depth. We pay particular attention to the interactions between astrocytes and neurons mediated by TNTs, which are fundamental to brain architecture and function. Dysfunctions in these interactions are implicated in the spread of protein aggregates and mitochondrial anomalies, contributing to the pathogenesis of neurodegenerative diseases. The review culminates with a synthesis of the current understanding of TNT biology and identifies research gaps, advocating for intensified exploration into TNTs as a promising therapeutic frontier.
Collapse
Affiliation(s)
- Cuixiang Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; 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
| | - Min Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; 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
| | - Shasha 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; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shifeng Chu
- 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
| | - Zhao Zhang
- 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.
| | - Naihong Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; 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; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
10
|
Giusti V, Kaur G, Giusto E, Civiero L. Brain clearance of protein aggregates: a close-up on astrocytes. Mol Neurodegener 2024; 19:5. [PMID: 38229094 PMCID: PMC10790381 DOI: 10.1186/s13024-024-00703-1] [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: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 01/18/2024] Open
Abstract
Protein misfolding and accumulation defines a prevailing feature of many neurodegenerative disorders, finally resulting in the formation of toxic intra- and extracellular aggregates. Intracellular aggregates can enter the extracellular space and be subsequently transferred among different cell types, thus spreading between connected brain districts.Although microglia perform a predominant role in the removal of extracellular aggregated proteins, mounting evidence suggests that astrocytes actively contribute to the clearing process. However, the molecular mechanisms used by astrocytes to remove misfolded proteins are still largely unknown.Here we first provide a brief overview of the progressive transition from soluble monomers to insoluble fibrils that characterizes amyloid proteins, referring to α-Synuclein and Tau as archetypical examples. We then highlight the mechanisms at the basis of astrocyte-mediated clearance with a focus on their potential ability to recognize, collect, internalize and digest extracellular protein aggregates. Finally, we explore the potential of targeting astrocyte-mediated clearance as a future therapeutic approach for the treatment of neurodegenerative disorders characterized by protein misfolding and accumulation.
Collapse
Affiliation(s)
| | - Gurkirat Kaur
- Department of Biology, University of Padova, Padua, Italy
| | | | - Laura Civiero
- IRCCS San Camillo Hospital, Venice, Italy.
- Department of Biology, University of Padova, Padua, Italy.
| |
Collapse
|
11
|
Khaled M, Al-Jamal H, Tajer L, El-Mir R. Alzheimer's Disease in Lebanon: Exploring Genetic and Environmental Risk Factors-A Comprehensive Review. J Alzheimers Dis 2024; 99:21-40. [PMID: 38640157 DOI: 10.3233/jad-231432] [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: 04/21/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that displays a high prevalence in Lebanon causing a local burden in healthcare and socio-economic sectors. Unfortunately, the lack of prevalence studies and clinical trials in Lebanon minimizes the improvement of AD patient health status. In this review, we include over 155 articles to cover the different aspects of AD ranging from mechanisms to possible treatment and management tools. We highlight some important modifiable and non-modifiable risk factors of the disease including genetics, age, cardiovascular diseases, smoking, etc. Finally, we propose a hypothetical genetic synergy model between APOE4 and TREM2 genes which constitutes a potential early diagnostic tool that helps in reducing the risk of AD based on preventative measures decades before cognitive decline. The studies on AD in Lebanon and the Middle East are scarce. This review points out the importance of genetic mapping in the understanding of disease pathology which is crucial for the emergence of novel diagnostic tools. Hence, we establish a rigid basis for further research to identify the most influential genetic and environmental risk factors for the purpose of using more specific diagnostic tools and possibly adopting a local management protocol.
Collapse
Affiliation(s)
| | - Hadi Al-Jamal
- Faculty of Public Health III, Lebanese University, Tripoli, Lebanon
| | - Layla Tajer
- Faculty of Public Health III, Lebanese University, Tripoli, Lebanon
| | - Reem El-Mir
- Faculty of Public Health III, Lebanese University, Tripoli, Lebanon
| |
Collapse
|
12
|
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
|
13
|
Henderson JM, Ljubojevic N, Belian S, Chaze T, Castaneda D, Battistella A, Giai Gianetto Q, Matondo M, Descroix S, Bassereau P, Zurzolo C. Tunnelling nanotube formation is driven by Eps8/IRSp53-dependent linear actin polymerization. EMBO J 2023; 42:e113761. [PMID: 38009333 PMCID: PMC10711657 DOI: 10.15252/embj.2023113761] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/28/2023] Open
Abstract
Tunnelling nanotubes (TNTs) connect distant cells and mediate cargo transfer for intercellular communication in physiological and pathological contexts. How cells generate these actin-mediated protrusions to span lengths beyond those attainable by canonical filopodia remains unknown. Through a combination of micropatterning, microscopy, and optical tweezer-based approaches, we demonstrate that TNTs formed through the outward extension of actin achieve distances greater than the mean length of filopodia and that branched Arp2/3-dependent pathways attenuate the extent to which actin polymerizes in nanotubes, thus limiting their occurrence. Proteomic analysis using epidermal growth factor receptor kinase substrate 8 (Eps8) as a positive effector of TNTs showed that, upon Arp2/3 inhibition, proteins enhancing filament turnover and depolymerization were reduced and Eps8 instead exhibited heightened interactions with the inverted Bin/Amphiphysin/Rvs (I-BAR) domain protein IRSp53 that provides a direct connection with linear actin polymerases. Our data reveals how common protrusion players (Eps8 and IRSp53) form tunnelling nanotubes, and that when competing pathways overutilizing such proteins and monomeric actin in Arp2/3 networks are inhibited, processes promoting linear actin growth dominate to favour tunnelling nanotube formation.
Collapse
Affiliation(s)
- J Michael Henderson
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and InfectionCNRS UMR 3691, Université de Paris, Institut PasteurParisFrance
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico‐Chimie CurieParisFrance
- Present address:
Department of ChemistryBowdoin CollegeBrunswickMEUSA
| | - Nina Ljubojevic
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and InfectionCNRS UMR 3691, Université de Paris, Institut PasteurParisFrance
- Sorbonne UniversitéParisFrance
| | - Sevan Belian
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and InfectionCNRS UMR 3691, Université de Paris, Institut PasteurParisFrance
- Université Paris‐SaclayGif‐sur‐YvetteFrance
| | - Thibault Chaze
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut PasteurParisFrance
| | - Daryl Castaneda
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and InfectionCNRS UMR 3691, Université de Paris, Institut PasteurParisFrance
- Keele UniversityKeeleUK
| | - Aude Battistella
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico‐Chimie CurieParisFrance
| | - Quentin Giai Gianetto
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut PasteurParisFrance
- Bioinformatics and Biostatistics Hub, Computational Biology DepartmentCNRS USR 3756, Institut PasteurParisFrance
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut PasteurParisFrance
| | - Stéphanie Descroix
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico‐Chimie CurieParisFrance
- Institut Pierre‐Gilles de GennesParisFrance
| | - Patricia Bassereau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico‐Chimie CurieParisFrance
| | - Chiara Zurzolo
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and InfectionCNRS UMR 3691, Université de Paris, Institut PasteurParisFrance
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
14
|
Shulman M, Kong J, O'Gorman J, Ratti E, Rajagovindan R, Viollet L, Huang E, Sharma S, Racine AM, Czerkowicz J, Graham D, Li Y, Hering H, Haeberlein SB. TANGO: a placebo-controlled randomized phase 2 study of efficacy and safety of the anti-tau monoclonal antibody gosuranemab in early Alzheimer's disease. NATURE AGING 2023; 3:1591-1601. [PMID: 38012285 PMCID: PMC10724064 DOI: 10.1038/s43587-023-00523-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/10/2023] [Indexed: 11/29/2023]
Abstract
In Alzheimer's disease, the spread of aberrantly phosphorylated tau is an important criterion in the Braak staging of disease severity and correlates with disease symptomatology. Here, we report the results of TANGO ( NCT03352557 ), a randomized, double-blind, placebo-controlled, parallel-group and multiple-dose long-term trial of gosuranemab-a monoclonal antibody to N-terminal tau-in patients with early Alzheimer's disease. The primary objective was to assess the safety and tolerability of gosuranemab compared to placebo. The secondary objectives were to assess the efficacy of multiple doses of gosuranemab in slowing cognitive and functional impairment (using the Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) scores at week 78) and evaluate the immunogenicity of gosuranemab (using the incidence of anti-gosuranemab antibody responses). Participants were randomized (n = 654); received (n = 650) low-dose (125 mg once every 4 weeks (q4w), n = 58; 375 mg q12w, n = 58), intermediate-dose (600 mg q4w, n = 106) or high-dose (2,000 mg q4w, n = 214) gosuranemab or placebo (q4w, n = 214) intravenously for 78 weeks; and assigned to cerebrospinal fluid (n = 327) and/or tau positron emission tomography (n = 357) biomarker substudies. Gosuranemab had an acceptable safety profile and was generally well tolerated (incidence of serious adverse events: placebo, 12.1%; low dose, 10.3%; intermediate dose, 12.3%; high dose, 11.7%). The incidence of treatment-emergent gosuranemab antibody responses was low at all time points. No significant effects were identified in cognitive and functional tests as no dose resulted in a favorable change from the baseline CDR-SB score at week 78 compared to placebo control (adjusted mean change: placebo, 1.85; low dose, 2.20; intermediate dose, 2.24; high dose, 1.85). At week 76, all doses caused significant (P < 0.0001) reductions in the cerebrospinal fluid levels of unbound N-terminal tau compared to placebo.
Collapse
Affiliation(s)
| | | | | | - Elena Ratti
- Biogen, Cambridge, MA, USA
- Takeda Pharmaceuticals, Cambridge, MA, USA
| | | | - Louis Viollet
- Biogen, Cambridge, MA, USA
- Moderna, Cambridge, MA, USA
| | | | | | - Annie M Racine
- Biogen, Cambridge, MA, USA
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
15
|
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
|
16
|
Wolzak K, Vermunt L, Campo MD, Jorge-Oliva M, van Ziel AM, Li KW, Smit AB, Chen-Ploktkin A, Irwin DJ, Lemstra AW, Pijnenburg Y, van der Flier W, Zetterberg H, Gobom J, Blennow K, Visser PJ, Teunissen CE, Tijms BM, Scheper W. Protein disulfide isomerases as CSF biomarkers for the neuronal response to tau pathology. Alzheimers Dement 2023; 19:3563-3574. [PMID: 36825551 DOI: 10.1002/alz.12978] [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: 08/05/2022] [Revised: 10/06/2021] [Accepted: 01/13/2023] [Indexed: 02/25/2023]
Abstract
INTRODUCTION Cerebrospinal fluid (CSF) biomarkers for specific cellular disease processes are lacking for tauopathies. In this translational study we aimed to identify CSF biomarkers reflecting early tau pathology-associated unfolded protein response (UPR) activation. METHODS We employed mass spectrometry proteomics and targeted immunoanalysis in a combination of biomarker discovery in primary mouse neurons in vitro and validation in patient CSF from two independent large multicentre cohorts (EMIF-AD MBD, n = 310; PRIDE, n = 771). RESULTS First, we identify members of the protein disulfide isomerase (PDI) family in the neuronal UPR-activated secretome and validate secretion upon tau aggregation in vitro. Next, we demonstrate that PDIA1 and PDIA3 levels correlate with total- and phosphorylated-tau levels in CSF. PDIA1 levels are increased in CSF from AD patients compared to controls and patients with tau-unrelated frontotemporal and Lewy body dementia (LBD). HIGHLIGHTS Neuronal unfolded protein response (UPR) activation induces the secretion of protein disulfide isomerases (PDIs) in vitro. PDIA1 is secreted upon tau aggregation in neurons in vitro. PDIA1 and PDIA3 levels correlate with total and phosphorylated tau levels in CSF. PDIA1 levels are increased in CSF from Alzheimer's disease (AD) patients compared to controls. PDIA1 levels are not increased in CSF from tau-unrelated frontotemporal dementia (FTD) and Lewy body dementia (LBD) patients.
Collapse
Affiliation(s)
- Kimberly Wolzak
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Functional Genomics Section, Department of Human Genetics, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Lisa Vermunt
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
| | - Marta Del Campo
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo- CEU, CEU Universities, Madrid, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Marta Jorge-Oliva
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Anna Maria van Ziel
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Functional Genomics Section, Department of Human Genetics, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Ka Wan Li
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - August B Smit
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Alice Chen-Ploktkin
- Department of Neurology, Perelman school of medicine, University of Pennsylvania, Philadelphia, USA
| | - David J Irwin
- Department of Neurology, Perelman school of medicine, University of Pennsylvania, Philadelphia, USA
- Penn Frontotemporal Degeneration Center, Perelman school of medicine, University of Pennsylvania, Philadelphia, USA
| | - Afina W Lemstra
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
| | - Yolande Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
| | - Wiesje van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Pieter Jelle Visser
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
| | - Betty M Tijms
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
| | - Wiep Scheper
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Functional Genomics Section, Department of Human Genetics, Amsterdam University Medical Centers (UMC) location Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| |
Collapse
|
17
|
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: 60] [Impact Index Per Article: 30.0] [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
|
18
|
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
|
19
|
I F. The unique neuropathological vulnerability of the human brain to aging. Ageing Res Rev 2023; 87:101916. [PMID: 36990284 DOI: 10.1016/j.arr.2023.101916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD)-related neurofibrillary tangles (NFT), argyrophilic grain disease (AGD), aging-related tau astrogliopathy (ARTAG), limbic predominant TDP-43 proteinopathy (LATE), and amygdala-predominant Lewy body disease (LBD) are proteinopathies that, together with hippocampal sclerosis, progressively appear in the elderly affecting from 50% to 99% of individuals aged 80 years, depending on the disease. These disorders usually converge on the same subject and associate with additive cognitive impairment. Abnormal Tau, TDP-43, and α-synuclein pathologies progress following a pattern consistent with an active cell-to-cell transmission and abnormal protein processing in the host cell. However, cell vulnerability and transmission pathways are specific for each disorder, albeit abnormal proteins may co-localize in particular neurons. All these alterations are unique or highly prevalent in humans. They all affect, at first, the archicortex and paleocortex to extend at later stages to the neocortex and other regions of the telencephalon. These observations show that the phylogenetically oldest areas of the human cerebral cortex and amygdala are not designed to cope with the lifespan of actual humans. New strategies aimed at reducing the functional overload of the human telencephalon, including optimization of dream repair mechanisms and implementation of artificial circuit devices to surrogate specific brain functions, appear promising.
Collapse
Affiliation(s)
- Ferrer I
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain; Biomedical Research Network of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
20
|
The central role of tau in Alzheimer’s disease: From neurofibrillary tangle maturation to the induction of cell death. Brain Res Bull 2022; 190:204-217. [DOI: 10.1016/j.brainresbull.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
|
21
|
Polanco JC, Götz J. Exosomal and vesicle-free tau seeds-propagation and convergence in endolysosomal permeabilization. FEBS J 2022; 289:6891-6907. [PMID: 34092031 DOI: 10.1111/febs.16055] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/19/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023]
Abstract
In Alzheimer's disease (AD), β-amyloid peptides aggregate to form amyloid plaques, and the microtubule-associated protein tau forms neurofibrillary tangles. However, severity and duration of AD correlate with the stereotypical emergence of tau tangles throughout the brain, suggestive of a gradual region-to-region spreading of pathological tau. The current notion in the field is that misfolded tau seeds propagate transsynaptically and corrupt the proper folding of soluble tau in recipient neurons. This is supported by accumulating evidence showing that in AD, functional connectivity and not proximity predicts the spreading of tau pathology. Tau seeds can be found in two flavors, vesicle-free, that is, naked as in oligomers and fibrils, or encapsulated by membranes of secreted vesicles known as exosomes. Both types of seeds have been shown to propagate between interconnected neurons. Here, we describe potential ways of how their propagation can be controlled in several subcellular compartments by manipulating mechanisms affecting production, neuron-to-neuron transmission, internalization, endosomal escape, and autophagy. We emphasize that although vesicle-free tau seeds and exosomes differ, they share the ability to trigger endolysosomal permeabilization. Such a mechanistic convergence in endolysosomal permeabilization presents itself as a unique opportunity to target both types of tau seeding. We discuss the cellular response to endolysosomal damage that might be key to control permeabilization, and the significant overlap in the seeding mechanism of proteopathic agents other than tau, which suggests that targeting the endolysosomal pathway could pave the way toward developing broad-spectrum treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan Carlos Polanco
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Li L, Jiang Y, Wu G, Mahaman YAR, Ke D, Wang Q, Zhang B, Wang JZ, Li HL, Liu R, Wang X. Phosphorylation of Truncated Tau Promotes Abnormal Native Tau Pathology and Neurodegeneration. Mol Neurobiol 2022; 59:6183-6199. [PMID: 35896773 DOI: 10.1007/s12035-022-02972-7] [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: 04/14/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022]
Abstract
Abnormal posttranslational modifications of tau play important roles in mediating neurodegeneration in tauopathies including Alzheimer's disease. Both phosphorylation and truncation are implicated in the pathogenesis of tauopathies. However, whether phosphorylation aggravates truncated tau-induced pathology and neurodegeneration remains elusive. Here, we construct different tau fragments cleaved by delta secretase, with either phosphorylation or non-phosphorylation mimic mutations, and evaluate the contributions of phosphorylation to truncated tau-induced pathological and behavioral alterations in vitro and in vivo through biochemical methods including detergent insoluble tau extraction, western blot, immunofluorescence, flow cytometry, and behavior tests. Our results show that the self-aggregation of phospho-truncated tau is significantly influenced by the domain it contains. N-terminal inhibits, proline-rich domain promotes, and C-terminus have no impact on phospho-truncated tau aggregation. Phosphorylation of truncated tau1-368, which contains the microtubule-binding repeat domain and the proline-rich domain, induces endogenous tau phosphorylation and aggregation. In vivo, phospho-tau1-368 but not non-phospho-tau1-368 leads to a decrease in body weight of C57BL/6 J mice. Intriguingly, although tau1-368-induced anxiety behavior in C57BL/6 J mice is phosphorylation-independent, the recognition memory of mice is impaired by phospho-tau1-368, but not by non-phospho-tau1-368. Immunofluorescence staining shows that overexpressing phospho-tau1-368 results in neuronal loss and gliosis in the hippocampus, while the transmission of tau1-368 is phosphorylation-independent as revealed by the flow cytometry results in vitro and immunofluorescence staining in vivo. Our findings indicate that phosphorylation of truncated tau significantly fosters endogenous tau pathology and neurodegeneration.
Collapse
Affiliation(s)
- Longfei Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanli Jiang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Wu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yacoubou Abdoul Razak Mahaman
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bin Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, JS, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Hong-Lian Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, JS, China.
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
23
|
Hromadkova L, Siddiqi MK, Liu H, Safar JG. Populations of Tau Conformers Drive Prion-like Strain Effects in Alzheimer's Disease and Related Dementias. Cells 2022; 11:2997. [PMID: 36230957 PMCID: PMC9562632 DOI: 10.3390/cells11192997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Recent findings of diverse populations of prion-like conformers of misfolded tau protein expand the prion concept to Alzheimer's disease (AD) and monogenic frontotemporal lobar degeneration (FTLD)-MAPT P301L, and suggest that distinct strains of misfolded proteins drive the phenotypes and progression rates in many neurodegenerative diseases. Notable progress in the previous decades has generated many lines of proof arguing that yeast, fungal, and mammalian prions determine heritable as well as infectious traits. The extraordinary phenotypic diversity of human prion diseases arises from structurally distinct prion strains that target, at different progression speeds, variable brain structures and cells. Although human prion research presents beneficial lessons and methods to study the mechanism of strain diversity of protein-only pathogens, the fundamental molecular mechanism by which tau conformers are formed and replicate in diverse tauopathies is still poorly understood. In this review, we summarize up to date advances in identification of diverse tau conformers through biophysical and cellular experimental paradigms, and the impact of heterogeneity of pathological tau strains on personalized structure- and strain-specific therapeutic approaches in major tauopathies.
Collapse
Affiliation(s)
- Lenka Hromadkova
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - He Liu
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jiri G. Safar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
24
|
Donnelly KM, Coleman CM, Fuller ML, Reed VL, Smerina D, Tomlinson DS, Pearce MMP. Hunting for the cause: Evidence for prion-like mechanisms in Huntington’s disease. Front Neurosci 2022; 16:946822. [PMID: 36090278 PMCID: PMC9448931 DOI: 10.3389/fnins.2022.946822] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
The hypothesis that pathogenic protein aggregates associated with neurodegenerative diseases spread from cell-to-cell in the brain in a manner akin to infectious prions has gained substantial momentum due to an explosion of research in the past 10–15 years. Here, we review current evidence supporting the existence of prion-like mechanisms in Huntington’s disease (HD), an autosomal dominant neurodegenerative disease caused by expansion of a CAG repeat tract in exon 1 of the huntingtin (HTT) gene. We summarize information gained from human studies and in vivo and in vitro models of HD that strongly support prion-like features of the mutant HTT (mHTT) protein, including potential involvement of molecular features of mHTT seeds, synaptic structures and connectivity, endocytic and exocytic mechanisms, tunneling nanotubes, and nonneuronal cells in mHTT propagation in the brain. We discuss mechanisms by which mHTT aggregate spreading and neurotoxicity could be causally linked and the potential benefits of targeting prion-like mechanisms in the search for new disease-modifying therapies for HD and other fatal neurodegenerative diseases.
Collapse
Affiliation(s)
- Kirby M. Donnelly
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
| | - Cevannah M. Coleman
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
| | - Madison L. Fuller
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
| | - Victoria L. Reed
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
| | - Dayna Smerina
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
| | - David S. Tomlinson
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
| | - Margaret M. Panning Pearce
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA, United States
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
- *Correspondence: Margaret M. Panning Pearce,
| |
Collapse
|
25
|
Lagalwar S. Mechanisms of tunneling nanotube-based propagation of neurodegenerative disease proteins. Front Mol Neurosci 2022; 15:957067. [PMID: 35909452 PMCID: PMC9336677 DOI: 10.3389/fnmol.2022.957067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Tunneling nanotubes (TNTs), intercellular connections enriched with F-actin, were first identified as a viable means of cellular communication and organelle transport in animal cells at the early part of this century. Within the last 10 years, these microscopic and highly dynamic protrusions have been implicated in neurodegenerative disease propagation and pathogenesis. A host of aggregation-prone protein inclusions, including those containing alpha-synuclein, tau, prions and others, hijack this communication mechanism in both neurons and astrocytes. The exact cellular mechanisms underlying TNT-based propagation remain largely unknown, however, common practices can be identified. First, selective expression of the aggregation-prone form of proteins increases TNT density; next, endo-lysosomal pathways appear to support the loading and unloading of protein onto the TNT; and finally, TNT assembly results in the spontaneous formation of aggregation-prone protein inclusions in “acceptor” cells, indicating that TNTs are involved in not only the transport of inclusions but also in the seeding of new inclusions in naïve cells. These observations have implications for the spreading of neurodegenerative disease in the central nervous system and the consequent progression of symptoms. Here, I will summarize the empirical evidence of TNT-based aggregation-prone protein propagation to date, and propose an inclusive model of aggregate inclusion propagation along TNTs.
Collapse
|
26
|
Turos-Korgul L, Kolba MD, Chroscicki P, Zieminska A, Piwocka K. Tunneling Nanotubes Facilitate Intercellular Protein Transfer and Cell Networks Function. Front Cell Dev Biol 2022; 10:915117. [PMID: 35903550 PMCID: PMC9314668 DOI: 10.3389/fcell.2022.915117] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022] Open
Abstract
The past decade witnessed a huge interest in the communication machinery called tunneling nanotubes (TNTs) which is a novel, contact-dependent type of intercellular protein transfer (IPT). As the IPT phenomenon plays a particular role in the cross-talk between cells, including cancer cells as well as in the immune and nervous systems, it therefore participates in remodeling of the cellular networks. The following review focuses on the placing the role of tunneling nanotube-mediated protein transfer between distant cells. Firstly, we describe different screening methods used to study IPT including tunneling nanotubes. Further, we present various examples of TNT-mediated protein transfer in the immune system, cancer microenvironment and in the nervous system, with particular attention to the methods used to verify the transfer of individual proteins.
Collapse
|
27
|
Simonsen A, Wollert T. Don't forget to be picky – selective autophagy of protein aggregates in neurodegenerative diseases. Curr Opin Cell Biol 2022; 75:102064. [DOI: 10.1016/j.ceb.2022.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/12/2022] [Accepted: 01/22/2022] [Indexed: 12/16/2022]
|
28
|
Effects of oligomer toxicity, fibril toxicity and fibril spreading in synucleinopathies. Cell Mol Life Sci 2022; 79:174. [PMID: 35244787 PMCID: PMC8897347 DOI: 10.1007/s00018-022-04166-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 12/18/2022]
Abstract
Protein misfolding is a general hallmark of protein deposition diseases, such as Alzheimer’s disease or Parkinson’s disease, in which different types of aggregated species (oligomers, protofibrils and fibrils) are generated by the cells. Despite widespread interest, the relationship between oligomers and fibrils in the aggregation process and spreading remains elusive. A large variety of experimental evidences supported the idea that soluble oligomeric species of different proteins might be more toxic than the larger fibrillar forms. Furthermore, the lack of correlation between the presence of the typical pathological inclusions and disease sustained this debate. However, recent data show that the β-sheet core of the α-Synuclein (αSyn) fibrils is unable to establish persistent interactions with the lipid bilayers, but they can release oligomeric species responsible for an immediate dysfunction of the recipient neurons. Reversibly, such oligomeric species could also contribute to pathogenesis via neuron-to-neuron spreading by their direct cell-to-cell transfer or by generating new fibrils, following their neuronal uptake. In this Review, we discuss the various mechanisms of cellular dysfunction caused by αSyn, including oligomer toxicity, fibril toxicity and fibril spreading.
Collapse
|
29
|
Yoshida S, Hasegawa T. Deciphering the prion-like behavior of pathogenic protein aggregates in neurodegenerative diseases. Neurochem Int 2022; 155:105307. [PMID: 35181393 DOI: 10.1016/j.neuint.2022.105307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are hitherto classified based on their core clinical features, the anatomical distribution of neurodegeneration, and the cell populations mainly affected. On the other hand, the wealth of neuropathological, genetic, molecular and biochemical studies have identified the existence of distinct insoluble protein aggregates in the affected brain regions. These findings have spread the use of a collective term, proteinopathy, for neurodegenerative disorders with particular type of structurally altered protein accumulation. Particularly, a recent breakthrough in this field came with the discovery that these protein aggregates can transfer from one cell to another, thereby converting normal proteins to potentially toxic, misfolded species in a prion-like manner. In this review, we focus specifically on the molecular and cellular basis that underlies the seeding activity and transcellular spreading phenomenon of neurodegeneration-related protein aggregates, and discuss how these events contribute to the disease progression.
Collapse
Affiliation(s)
- Shun Yoshida
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan; Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa, Yamagata, 992-1202, Japan
| | - Takafumi Hasegawa
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan.
| |
Collapse
|
30
|
Gómez O, Perini-Villanueva G, Yuste A, Rodríguez-Navarro JA, Poch E, Bejarano E. Autophagy and Glycative Stress: A Bittersweet Relationship in Neurodegeneration. Front Cell Dev Biol 2022; 9:790479. [PMID: 35004686 PMCID: PMC8733682 DOI: 10.3389/fcell.2021.790479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/06/2021] [Indexed: 12/27/2022] Open
Abstract
Autophagy is a fine-tuned proteolytic pathway that moves dysfunctional/aged cellular components into the lysosomal compartment for degradation. Over the last 3 decades, global research has provided evidence for the protective role of autophagy in different brain cell components. Autophagic capacities decline with age, which contributes to the accumulation of obsolete/damaged organelles and proteins and, ultimately, leads to cellular aging in brain tissues. It is thus well-accepted that autophagy plays an essential role in brain homeostasis, and malfunction of this catabolic system is associated with major neurodegenerative disorders. Autophagy function can be modulated by different types of stress, including glycative stress. Glycative stress is defined as a cellular status with abnormal and accelerated accumulation of advanced glycation end products (AGEs). It occurs in hyperglycemic states, both through the consumption of high-sugar diets or under metabolic conditions such as diabetes. In recent years, glycative stress has gained attention for its adverse impact on brain pathology. This is because glycative stress stimulates insoluble, proteinaceous aggregation that is linked to the malfunction of different neuropathological proteins. Despite the emergence of new literature suggesting that autophagy plays a major role in fighting glycation-derived damage by removing cytosolic AGEs, excessive glycative stress might also negatively impact autophagic function. In this mini-review, we provide insight on the status of present knowledge regarding the role of autophagy in brain physiology and pathophysiology, with an emphasis on the cytoprotective role of autophagic function to ameliorate the adverse effects of glycation-derived damage in neurons, glia, and neuron-glia interactions.
Collapse
Affiliation(s)
- Olga Gómez
- School of Health Sciences and Veterinary School, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Giuliana Perini-Villanueva
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Andrea Yuste
- School of Health Sciences and Veterinary School, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | | | - Enric Poch
- School of Health Sciences and Veterinary School, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Eloy Bejarano
- School of Health Sciences and Veterinary School, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| |
Collapse
|
31
|
Abstract
The CAG/CAA expansion encoding polyQ huntingtin (mutant huntingtin [mHTT]) causes Huntington's disease (HD), which is characterized by atrophy and loss of striatal medium spiny neurons (MSNs), which are preceded by neuropathological alterations in the cortex. Previous studies have shown that mHTT can spread in the brain, but the mechanisms involved in the stereotyped degeneration and dysfunction of the neurons from the striatum to the cortex remain unclear. In this study, we found that the mHTT expression initially restricted in the striatum later spread to the cortical regions in mouse brains. Such transmission was diminished in mice that lacked the striatal-enriched protein Ras-homolog enriched in the striatum (Rhes). Rhes restricted to MSNs was also found in the cortical layers of the brain, indicating a new transmission route for the Rhes protein to the brain. Mechanistically, Rhes promotes such transmission via a direct cell-to-cell contact mediated by tunneling nanotubes (TNTs), the membranous protrusions that enable the transfer of mHTT, Rhes, and other vesicular cargoes. These transmission patterns suggest that Rhes and mHTT are likely co-transported in the brain using TNT-like cell-to-cell contacts. On the basis of these new results, a perspective is presented in this review: Rhes may ignite the mHTT transmission from the striatum that may coincide with HD onset and disease progression through an anatomically connected striato-cortical retrograde route.
Collapse
|
32
|
Chidambaram H, Das R, Chinnathambi S. G-Protein coupled Purinergic P2Y12 receptor interacts and internalizes TauRD-mediated by membrane-associated actin cytoskeleton remodelling in microglia. Eur J Cell Biol 2022; 101:151201. [DOI: 10.1016/j.ejcb.2022.151201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
|
33
|
Huang H, Toker N, Burr E, Okoro J, Moog M, Hearing C, Lagalwar S. Intercellular Propagation and Aggregate Seeding of Mutant Ataxin-1. J Mol Neurosci 2021; 72:708-718. [PMID: 34826062 PMCID: PMC8986690 DOI: 10.1007/s12031-021-01944-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/06/2021] [Indexed: 01/07/2023]
Abstract
Intercellular propagation of aggregated protein inclusions along actin-based tunneling nanotubes (TNTs) has been reported as a means of pathogenic spread in Alzheimer’s, Parkinson’s, and Huntington’s diseases. Propagation of oligomeric-structured polyglutamine-expanded ataxin-1 (Atxn1[154Q]) has been reported in the cerebellum of a Spinocerebellar ataxia type 1 (SCA1) knock-in mouse to correlate with disease propagation. In this study, we investigated whether a physiologically relevant polyglutamine-expanded ATXN1 protein (ATXN1[82Q]) could propagate intercellularly. Using a cerebellar-derived live cell model, we observed ATXN1 aggregates form in the nucleus, subsequently form in the cytoplasm, and finally, propagate to neighboring cells along actin-based intercellular connections. Additionally, we observed the facilitation of aggregate-resistant proteins into aggregates given the presence of aggregation-prone proteins within cells. Taken together, our results support a pathogenic role of intercellular propagation of polyglutamine-expanded ATXN1 inclusions.
Collapse
Affiliation(s)
- Haoyang Huang
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Nicholas Toker
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Eliza Burr
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Jeff Okoro
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Maia Moog
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Casey Hearing
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Sarita Lagalwar
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA.
| |
Collapse
|
34
|
Imbimbo BP, Ippati S, Watling M, Balducci C. A critical appraisal of tau-targeting therapies for primary and secondary tauopathies. Alzheimers Dement 2021; 18:1008-1037. [PMID: 34533272 DOI: 10.1002/alz.12453] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Primary tauopathies are neurological disorders in which tau protein deposition is the predominant pathological feature. Alzheimer's disease is a secondary tauopathy with tau forming hyperphosphorylated insoluble aggregates. Tau pathology can propagate from region to region in the brain, while alterations in tau processing may impair tau physiological functions. METHODS We reviewed literature on tau biology and anti-tau drugs using PubMed, meeting abstracts, and ClnicalTrials.gov. RESULTS The past 15 years have seen >30 drugs interfering with tau aggregation, processing, and accumulation reaching the clinic. Initial results with tau aggregation inhibitors and anti-tau monoclonal antibodies have not shown clinical efficacy. DISCUSSION The reasons for these clinical failures are unclear but could be linked to the clearing of physiological forms of tau by non-specific drugs. Research is now concentrating efforts on developing reliable translational animal models and selective compounds targeting specific tau epitopes, neurotoxic tau aggregates, and post-translational tau modifications.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milan, Italy
| |
Collapse
|
35
|
The 3.0 Cell Communication: New Insights in the Usefulness of Tunneling Nanotubes for Glioblastoma Treatment. Cancers (Basel) 2021; 13:cancers13164001. [PMID: 34439156 PMCID: PMC8392307 DOI: 10.3390/cancers13164001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/05/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Communication between cells helps tumors acquire resistance to chemotherapy and makes the struggle against cancer more challenging. Tunneling nanotubes (TNTs) are long channels able to connect both nearby and distant cells, contributing to a more malignant phenotype. This finding might be useful in designing novel strategies of drug delivery exploiting these systems of connection. This would be particularly important to reach tumor niches, where glioblastoma stem cells proliferate and provoke immune escape, thereby increasing metastatic potential and tumor recurrence a few months after surgical resection of the primary mass. Along with the direct inhibition of TNT formation, TNT analysis, and targeting strategies might be useful in providing innovative tools for the treatment of this tumor. Abstract Glioblastoma (GBM) is a particularly challenging brain tumor characterized by a heterogeneous, complex, and multicellular microenvironment, which represents a strategic network for treatment escape. Furthermore, the presence of GBM stem cells (GSCs) seems to contribute to GBM recurrence after surgery, and chemo- and/or radiotherapy. In this context, intercellular communication modalities play key roles in driving GBM therapy resistance. The presence of tunneling nanotubes (TNTs), long membranous open-ended channels connecting distant cells, has been observed in several types of cancer, where they emerge to steer a more malignant phenotype. Here, we discuss the current knowledge about the formation of TNTs between different cellular types in the GBM microenvironment and their potential role in tumor progression and recurrence. Particularly, we highlight two prospective strategies targeting TNTs as possible therapeutics: (i) the inhibition of TNT formation and (ii) a boost in drug delivery between cells through these channels. The latter may require future studies to design drug delivery systems that are exchangeable through TNTs, thus allowing for access to distant tumor niches that are involved in tumor immune escape, maintenance of GSC plasticity, and increases in metastatic potential.
Collapse
|
36
|
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
|
37
|
Zurzolo C. Tunneling nanotubes: Reshaping connectivity. Curr Opin Cell Biol 2021; 71:139-147. [PMID: 33866130 DOI: 10.1016/j.ceb.2021.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/22/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022]
Abstract
Tunneling nanotubes (TNTs), open membranous channels between connected cells, represent a novel direct way of communication between distant cells for the diffusion of various cellular material, including survival or death signals, genetic material, organelles, and pathogens. Their discovery prompted us to review our understanding of many physiological and pathological processes involving cellular communication but also allowed us to discover new mechanisms of communication at a distance. While this has enriched the field, it has also generated some confusion, as different TNT-like protrusions have been described, and it is not clear whether they have the same structure-function. Most studies have been based on low-resolution imaging methods, and one of the major problems is the inconsistency in demonstrating the capacity of these various connections to transfer material between cells belonging to different populations. This brief review examines the fundamental properties of TNTs. In adult tissues, TNTs are stimulated by different diseases, stresses, and inflammatory signals. 'Moreover', based on the similarity of the processes of development of synaptic spines and TNT formation, we argue that TNTs in the brain predate synaptic transmission, being instrumental in the orchestration of the immature neuronal circuit.
Collapse
Affiliation(s)
- Chiara Zurzolo
- Membrane Traffic and Pathogenesis, Institut Pasteur, UMR3691 CNRS, 75015, Paris, France.
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Opportunities and Challenges in Tunneling Nanotubes Research: How Far from Clinical Application? Int J Mol Sci 2021; 22:ijms22052306. [PMID: 33669068 PMCID: PMC7956326 DOI: 10.3390/ijms22052306] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/21/2021] [Accepted: 02/21/2021] [Indexed: 02/08/2023] Open
Abstract
Tunneling nanotubes (TNTs) are recognized long membrane nanotubes connecting distance cells. In the last decade, growing evidence has shown that these subcellular structures mediate the specific transfer of cellular materials, pathogens, and electrical signals between cells. As intercellular bridges, they play a unique role in embryonic development, collective cell migration, injured cell recovery, cancer treatment resistance, and pathogen propagation. Although TNTs have been considered as potential drug targets for treatment, there is still a long way to go to translate the research findings into clinical practice. Herein, we emphasize the heterogeneous nature of TNTs by systemically summarizing the current knowledge on their morphology, structure, and biogenesis in different types of cells. Furthermore, we address the communication efficiency and biological outcomes of TNT-dependent transport related to diseases. Finally, we discuss the opportunities and challenges of TNTs as an exciting therapeutic approach by focusing on the development of efficient and safe drugs targeting TNTs.
Collapse
|
41
|
Chastagner P, Loria F, Vargas JY, Tois J, I Diamond M, Okafo G, Brou C, Zurzolo C. Fate and propagation of endogenously formed Tau aggregates in neuronal cells. EMBO Mol Med 2020; 12:e12025. [PMID: 33179866 PMCID: PMC7721367 DOI: 10.15252/emmm.202012025] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Tau accumulation in the form of neurofibrillary tangles in the brain is a hallmark of tauopathies such as Alzheimer's disease (AD). Tau aggregates accumulate in brain regions in a defined spatiotemporal pattern and may induce the aggregation of native Tau in a prion-like manner. However, the underlying mechanisms of cell-to-cell spreading of Tau pathology are unknown and could involve encapsulation within exosomes, trans-synaptic passage, and tunneling nanotubes (TNTs). We have established a neuronal cell model to monitor both internalization of externally added fibrils, synthetic (K18) or Tau from AD brain extracts, and real-time conversion of microtubule-binding domain of Tau fused to a fluorescent marker into aggregates. We found that these endogenously formed deposits colabel with ubiquitin and p62 but are not recruited to macroautophagosomes, eventually escaping clearance. Furthermore, endogenous K18-seeded Tau aggregates spread to neighboring cells where they seed new deposits. Transfer of Tau aggregates depends on direct cell contact, and they are found inside TNTs connecting neuronal cells. We further demonstrate that contact-dependent transfer occurs in primary neurons and between neurons and astrocytes in organotypic cultures.
Collapse
Affiliation(s)
| | - Frida Loria
- Unité de Trafic Membranaire et PathogenèseInstitut PasteurParisFrance
- Present address:
Laboratorio de Apoyo a la InvestigaciónHospital Universitario Fundación AlcorcónMadridSpain
| | - Jessica Y Vargas
- Unité de Trafic Membranaire et PathogenèseInstitut PasteurParisFrance
| | - Josh Tois
- Unité de Trafic Membranaire et PathogenèseInstitut PasteurParisFrance
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative DiseasesPeter O'Donnell Jr. Brain InstituteUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | | | - Christel Brou
- Unité de Trafic Membranaire et PathogenèseInstitut PasteurParisFrance
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et PathogenèseInstitut PasteurParisFrance
| |
Collapse
|