1
|
Palese F, Rakotobe M, Zurzolo C. Transforming the concept of connectivity: unveiling tunneling nanotube biology and their roles in brain development and neurodegeneration. Physiol Rev 2025; 105:1823-1865. [PMID: 40067081 DOI: 10.1152/physrev.00023.2024] [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: 07/09/2024] [Revised: 09/17/2024] [Accepted: 02/03/2025] [Indexed: 05/08/2025] Open
Abstract
Tunneling nanotubes (TNTs) are thin tubular membrane protrusions that connect distant cells, generating a complex cellular network. Over the past few decades, research on TNTs has provided important insights into their biology, including structural composition, formation mechanisms, modulators, and functionality. It has been discovered that TNTs allow cytoplasmic continuity between connected cells, facilitating fast intercellular communication via both passive and active exchange of materials. These features are pivotal in the nervous system, where rapid processing of inputs is physiologically required. TNTs have been implicated in the progression of neurodegenerative diseases and cancer in various in vitro models, and TNT-like structures have also been observed in the developing brain and in vivo. This highlights their significant role in pathophysiological processes. In this comprehensive review we aim to provide an extensive overview of TNTs, starting from key structural features and mechanisms of formation and describing the main experimental techniques used to detect these structures both in vitro and in vivo. We focus primarily on the nervous system, where the discovery of TNTs could prompt a reconsideration of the brain functioning as individual units (the neuronal theory of Cajal) versus neurons being physically connected, as Golgi believed. We illustrate the involvement of TNTs in brain development and neurodegenerative states and highlight the limitations and future research needs in this field.
Collapse
Affiliation(s)
- Francesca Palese
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris, France
| | - Malalaniaina Rakotobe
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris, France
| | - 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
|
2
|
Van Alstyne M, Pratt J, Parker R. Diverse influences on tau aggregation and implications for disease progression. Genes Dev 2025; 39:555-581. [PMID: 40113250 PMCID: PMC12047666 DOI: 10.1101/gad.352551.124] [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: 03/22/2025]
Abstract
Tau is an intrinsically disordered protein that accumulates in fibrillar aggregates in neurodegenerative diseases. The misfolding of tau can be understood as an equilibrium between different states and their propensity to form higher-order fibers, which is affected by several factors. First, modulation of the biochemical state of tau due to ionic conditions, post-translational modifications, cofactors, and interacting molecules or assemblies can affect the formation and structure of tau fibrils. Second, cellular processes impact tau aggregation through modulating stability, clearance, disaggregation, and transport. Third, through interactions with glial cells, the neuronal microenvironment can affect intraneuronal conditions with impacts on tau fibrilization and toxicity. Importantly, tau fibrils propagate through the brain via a "prion-like" manner, contributing to disease progression. This review highlights the biochemical and cellular pathways that modulate tau aggregation and discusses implications for pathobiology and tau-directed therapeutic approaches.
Collapse
Affiliation(s)
- Meaghan Van Alstyne
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
| | - James Pratt
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA
| | - Roy Parker
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA;
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
| |
Collapse
|
3
|
Landis MK, Kunze A. Intra-axonal Nanomagnetic Forces Differentially Impact hTau40 Transport Dynamics in Primary Cortical and Hippocampal Neurons. ACS NANO 2025; 19:7884-7897. [PMID: 39963892 DOI: 10.1021/acsnano.4c14767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
A crucial aspect of neural engineering is the ability to manipulate proteins that are substantially involved in axonal outgrowth and maintenance. Previous work in this field has shown that applying low-magnitude (piconewton) forces to early stage neurons can result in altered distributions of critical structural proteins, such as the microtubule-associated protein Tau. Uncovering the mechanisms of Tau redistribution could provide a tool for manipulating dysregulated forms of the protein. This study examined how the transport of Tau responded to intra-axonal nanomagnetic forces (NMFs) in primary cortical and hippocampal neurons. High magnification, live cell fluorescent imaging was employed to visualize the transport of both full-length human Tau (hTau40) and amine-terminated, starch-coated fluorescent magnetic nanoparticles (afMNPs) to observe how these cell-internal forces could impact the transport of hTau40 within the axon. Here, we found that afMNPs acted by pulling on hTau40 puncta under NMF application, especially within cortical cells, where afMNPs were more likely to be found within the axon. Forces greater than 1 pN enabled differentiated transport speeds and displacement of hTau40 based on relative force direction. This data indicates that NMF can be utilized to engineer hTau40 transport, even in cells at later stages of maturation.
Collapse
Affiliation(s)
- Mackenna K Landis
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
- Montana Nanotechnology Facility, Montana State University, Bozeman, Montana 59717, United States
- Optical Technology Center, Montana State University, Bozeman, Montana 59717, United States
| |
Collapse
|
4
|
Gong Z, Wu T, Zhao Y, Guo J, Zhang Y, Li B, Li Y. Intercellular Tunneling Nanotubes as Natural Biophotonic Conveyors. ACS NANO 2025; 19:1036-1043. [PMID: 39630614 DOI: 10.1021/acsnano.4c12681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Tunneling nanotubes (TNTs), submicrometer membranous channels that bridge and connect distant cells, play a pivotal role in intercellular communication. Organelle transfer within TNTs is crucial in regulating cell growth, signal transmission, and disease progression. However, precise control over individual organelle transport within TNTs remains elusive. In this study, we introduce an optical technique that harnesses TNTs as biophotonic conveyors for the directional transport of individual organelles between cells. By utilizing near-infrared light propagating along the TNTs, optical forces were exerted on the organelles, enabling their active transport in a predetermined direction and at a controlled velocity. As a potential application, TNT conveyors were employed to inhibit mitochondrial hijacking from immune cells to cancer cells, thereby activating immune cells and suppressing cancer cell growth. Furthermore, neural modulation was achieved by transporting mitochondria and neurotransmitter-containing vesicles between neurons via TNT conveyors and axonal conveyors, respectively. This study presents a robust and precise approach to immune activation and neural regulation through the manipulation of organelle transfer at the subcellular level.
Collapse
Affiliation(s)
- Zhiyong Gong
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou 511443, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Tianli Wu
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou 511443, China
| | - Yanan Zhao
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou 511443, China
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yao Zhang
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou 511443, China
- Key Laboratory of Optoelectronic Information and Sensing Technologies of Guangdong Higher Education Institutes, Jinan University, Guangzhou 510632, China
| | - Baojun Li
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou 511443, China
| | - Yuchao Li
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou 511443, China
| |
Collapse
|
5
|
Wei Y, Du X, Guo H, Han J, Liu M. Mitochondrial dysfunction and Alzheimer's disease: pathogenesis of mitochondrial transfer. Front Aging Neurosci 2024; 16:1517965. [PMID: 39741520 PMCID: PMC11685155 DOI: 10.3389/fnagi.2024.1517965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, mitochondrial transfer has emerged as a universal phenomenon intertwined with various systemic physiological and pathological processes. Alzheimer's disease (AD) is a multifactorial disease, with mitochondrial dysfunction at its core. Although numerous studies have found evidence of mitochondrial transfer in AD models, the precise mechanisms remain unclear. Recent studies have revealed the dynamic transfer of mitochondria in Alzheimer's disease, not only between nerve cells and glial cells, but also between nerve cells and glial cells. In this review, we explore the pathways and mechanisms of mitochondrial transfer in Alzheimer's disease and how these transfer activities contribute to disease progression.
Collapse
Affiliation(s)
- Yun Wei
- *Correspondence: Yun Wei, ; Meixia Liu,
| | | | | | | | - Meixia Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Rakotobe M, Zurzolo C. [Tunneling nanotubes (TNTs): An essential yet overlooked modality of inter-cellular communication]. Med Sci (Paris) 2024; 40:829-836. [PMID: 39656980 DOI: 10.1051/medsci/2024152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
Abstract
Tunneling nanotubes (TNTs) are open membranous protrusions that allow direct communication between distant cells. Recent research has revealed their significant biological roles, prompting a reassessment of many physiological and pathological processes, especially in the nervous system where TNT properties could play a key physiological role. TNT-like connections have been observed in the developing brain and are implicated in neurodegenerative diseases, brain cancers, as well as in other diseases, underscoring their importance in pathophysiological events. This review covers the key features of TNTs, including their structural properties, formation mechanisms, and detection challenges. We also explore their functions, focusing on the nervous system. The discovery of TNTs may lead to a reconsideration of brain function as a physically connected neuronal network, as proposed by Golgi, complementing Cajal's theory of neurons as separate entities.
Collapse
Affiliation(s)
- Malalaniaina Rakotobe
- Trafic membranaire et pathogénèse, Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Paris, France
| | - Chiara Zurzolo
- Trafic membranaire et pathogénèse, Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Paris, France
| |
Collapse
|
7
|
Salem S, Alpaugh M, Saint-Pierre M, Alves-Martins-Borba FN, Cerquera-Cleves C, Lemieux M, Ngonza-Nito SB, De Koninck P, Melki R, Cicchetti F. Treatment with Tau fibrils impact Huntington's disease-related phenotypes in cell and mouse models. Neurobiol Dis 2024; 202:106696. [PMID: 39389154 DOI: 10.1016/j.nbd.2024.106696] [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: 06/19/2024] [Revised: 09/13/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
There is now compelling evidence for the presence of pathological forms of Tau in tissues of both patients and animal models of Huntington's disease (HD). While the root cause of this illness is a mutation within the huntingtin gene, a number of studies now suggest that HD could also be considered a secondary tauopathy. However, the contributory role of Tau in the pathogenesis and pathophysiology of this condition, as well as its implications in cellular toxicity and consequent behavioral impairments are largely unknown. We therefore performed intracerebral stereotaxic injections of recombinant human Tau monomers and fibrils into the knock-in zQ175 mouse model of HD. Tau fibrils induced cognitive and anxiety-like phenotypes predominantly in zQ175 mice and increased the number and size of insoluble mutant huntingtin (mHTT) aggregates in the brains of treated animals. To better understand the putative mechanisms through which Tau could initiate and/or contribute to pathology, we incubated StHdh striatal cells, an in vitro model of HD, with the different Tau forms and evaluated the effects on cell functionality and heat shock proteins Hsp70 and Hsp90. Calcium imaging experiments showed functional impairments of HD StHdh cells following treatment with Tau fibrils, as well as significant changes to the levels of both heat shock proteins which were found trapped within mHTT aggregates. The accumulation of Hsp70 and 90 within aggregates was also present in mouse tissue which suggests that alteration of molecular chaperone-dependent protein quality control may influence aggregation, implicating proteostasis in the mHTT-Tau interplay.
Collapse
Affiliation(s)
- Shireen Salem
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| | - Melanie Alpaugh
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Martine Saint-Pierre
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Flavia Natale Alves-Martins-Borba
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Catalina Cerquera-Cleves
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Mado Lemieux
- CERVO Brain Research Center, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Soki Bradel Ngonza-Nito
- Labortory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Paul De Koninck
- CERVO Brain Research Center, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Ronald Melki
- Labortory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Francesca Cicchetti
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
8
|
Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol 2024; 25:845-864. [PMID: 39014245 DOI: 10.1038/s41580-024-00753-9] [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: 06/10/2024] [Indexed: 07/18/2024]
Abstract
Tau protein is involved in various cellular processes, including having a canonical role in binding and stabilization of microtubules in neurons. Tauopathies are neurodegenerative diseases marked by the abnormal accumulation of tau protein aggregates in neurons, as seen, for example, in conditions such as frontotemporal dementia and Alzheimer disease. Mutations in tau coding regions or that disrupt tau mRNA splicing, tau post-translational modifications and cellular stress factors (such as oxidative stress and inflammation) increase the tendency of tau to aggregate and interfere with its clearance. Pathological tau is strongly implicated in the progression of neurodegenerative diseases, and the propagation of tau aggregates is associated with disease severity. Recent technological advancements, including cryo-electron microscopy and disease models derived from human induced pluripotent stem cells, have increased our understanding of tau-related pathology in neurodegenerative conditions. Substantial progress has been made in deciphering tau aggregate structures and the molecular mechanisms that underlie protein aggregation and toxicity. In this Review, we discuss recent insights into the diverse cellular functions of tau and the pathology of tau inclusions and explore the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sarah A Naguib
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
9
|
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
|
10
|
Scheiblich H, Eikens F, Wischhof L, Opitz S, Jüngling K, Cserép C, Schmidt SV, Lambertz J, Bellande T, Pósfai B, Geck C, Spitzer J, Odainic A, Castro-Gomez S, Schwartz S, Boussaad I, Krüger R, Glaab E, Di Monte DA, Bano D, Dénes Á, Latz E, Melki R, Pape HC, Heneka MT. Microglia rescue neurons from aggregate-induced neuronal dysfunction and death through tunneling nanotubes. Neuron 2024; 112:3106-3125.e8. [PMID: 39059388 DOI: 10.1016/j.neuron.2024.06.029] [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: 10/26/2023] [Revised: 02/12/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024]
Abstract
Microglia are crucial for maintaining brain health and neuron function. Here, we report that microglia establish connections with neurons using tunneling nanotubes (TNTs) in both physiological and pathological conditions. These TNTs facilitate the rapid exchange of organelles, vesicles, and proteins. In neurodegenerative diseases like Parkinson's and Alzheimer's disease, toxic aggregates of alpha-synuclein (α-syn) and tau accumulate within neurons. Our research demonstrates that microglia use TNTs to extract neurons from these aggregates, restoring neuronal health. Additionally, microglia share their healthy mitochondria with burdened neurons, reducing oxidative stress and normalizing gene expression. Disrupting mitochondrial function with antimycin A before TNT formation eliminates this neuroprotection. Moreover, co-culturing neurons with microglia and promoting TNT formation rescues suppressed neuronal activity caused by α-syn or tau aggregates. Notably, TNT-mediated aggregate transfer is compromised in microglia carrying Lrrk22(Gly2019Ser) or Trem2(T66M) and (R47H) mutations, suggesting a role in the pathology of these gene variants in neurodegenerative diseases.
Collapse
Affiliation(s)
- Hannah Scheiblich
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn, Germany; Max-Planck-Institute for Biology of Ageing, Cologne, Germany
| | - Frederik Eikens
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn, Germany; Max-Planck-Institute for Biology of Ageing, Cologne, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases, Bonn, Germany; Max-Planck-Institute for Biology of Ageing, Cologne, Germany
| | - Sabine Opitz
- Institute of Neuropathology, University of Bonn, Bonn, Germany
| | - Kay Jüngling
- Institute of Physiology I, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Csaba Cserép
- Institute of Experimental Medicine, Budapest, Hungary
| | - Susanne V Schmidt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | | | - Tracy Bellande
- Institut François Jacob, CEA and Laboratory of Neurodegenerative Diseases, Fontenay-aux-Roses, France
| | - Balázs Pósfai
- Institute of Experimental Medicine, Budapest, Hungary
| | - Charlotte Geck
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Jasper Spitzer
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Alexandru Odainic
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection & Immunity, University of Melbourne, Melbourne, VIC, Australia
| | | | | | - Ibrahim Boussaad
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | | | - Daniele Bano
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Ádám Dénes
- Institute of Experimental Medicine, Budapest, Hungary
| | - Eike Latz
- German Center for Neurodegenerative Diseases, Bonn, Germany; Institute of innate immunity, University Hospital Bonn, Bonn, Germany
| | - Ronald Melki
- Institut François Jacob, CEA and Laboratory of Neurodegenerative Diseases, Fontenay-aux-Roses, France
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases, Bonn, Germany; Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg; Institute of innate immunity, University Hospital Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts, Medical School, Worcester, MA, USA.
| |
Collapse
|
11
|
Raghavan A, Kashyap R, Sreedevi P, Jos S, Chatterjee S, Alex A, D’Souza MN, Giridharan M, Muddashetty R, Manjithaya R, Padavattan S, Nath S. Astroglia proliferate upon the biogenesis of tunneling nanotubes via α-synuclein dependent transient nuclear translocation of focal adhesion kinase. iScience 2024; 27:110565. [PMID: 39184442 PMCID: PMC11342280 DOI: 10.1016/j.isci.2024.110565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/13/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Astroglia play crucial neuroprotective roles by internalizing pathogenic aggregates and facilitating their degradation. Here, we show that α-SYN protofibril-induced organelle toxicities and reactive oxygen species (ROS) cause premature cellular senescence in astrocytes and astrocyte-derived cancer cells, resulting in a transient increase in the biogenesis of tunneling nanotubes (TNTs). TNT-biogenesis and TNT-mediated cell-to-cell transfer lead to clearance of α-SYN-induced organelle toxicities, reduction in cellular ROS levels, and reversal of cellular senescence. Enhanced cell proliferation is seen in the post-recovered cells after recovering from α-SYN-induced organelle toxicities. Further, we show that α-SYN-induced senescence promotes the transient localization of focal adhesion kinase (FAK) in the nucleus. FAK-mediated regulation of Rho-associated kinases plays a significant role in the biogenesis of TNTs and their subsequent proliferation. Our study emphasizes that TNT biogenesis has a potential role in the clearance of α-SYN-induced cellular toxicities, the consequences of which cause enhanced proliferation in the post-recovered astroglia cells.
Collapse
Affiliation(s)
- Abinaya Raghavan
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, India
| | - Rachana Kashyap
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, India
| | - P. Sreedevi
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Sneha Jos
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Suchana Chatterjee
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, India
| | - Ann Alex
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, India
| | | | - Mridhula Giridharan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ravi Muddashetty
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bengaluru, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Sivaraman Padavattan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Sangeeta Nath
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
12
|
Lee E, Park H, Kim S. Transcellular transmission and molecular heterogeneity of aggregation-prone proteins in neurodegenerative diseases. Mol Cells 2024; 47:100089. [PMID: 38971320 PMCID: PMC11286998 DOI: 10.1016/j.mocell.2024.100089] [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: 05/14/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024] Open
Abstract
The accumulation of aggregation-prone proteins in a specific neuronal population is a common feature of neurodegenerative diseases, which is correlated with the development of pathological lesions in diseased brains. The formation and progression of pathological protein aggregates in susceptible neurons induce cellular dysfunction, resulting in progressive degeneration. Moreover, recent evidence supports the notion that the cell-to-cell transmission of pathological protein aggregates may be involved in the onset and progression of many neurodegenerative diseases. Indeed, several studies have identified different pathological aggregate strains. Although how these different aggregate strains form remains unclear, a variety of biomolecular compositions or cross-seeding events promoted by the presence of other protein aggregates in the cellular environment may affect the formation of different strains of pathological aggregates, which in turn can influence complex pathologies in diseased brains. In this review, we summarize the recent results regarding cell-to-cell transmission and the molecular heterogeneity of pathological aggregate strains, raising key questions for future research directions.
Collapse
Affiliation(s)
- Eunmin Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Korea
| | - Hyeonwoo Park
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Korea
| | - Sangjune Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Korea.
| |
Collapse
|
13
|
Chu D, Yang X, Wang J, Zhou Y, Gu JH, Miao J, Wu F, Liu F. Tau truncation in the pathogenesis of Alzheimer's disease: a narrative review. Neural Regen Res 2024; 19:1221-1232. [PMID: 37905868 PMCID: PMC11467920 DOI: 10.4103/1673-5374.385853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Alzheimer's disease is characterized by two major neuropathological hallmarks-the extracellular β-amyloid plaques and intracellular neurofibrillary tangles consisting of aggregated and hyperphosphorylated Tau protein. Recent studies suggest that dysregulation of the microtubule-associated protein Tau, especially specific proteolysis, could be a driving force for Alzheimer's disease neurodegeneration. Tau physiologically promotes the assembly and stabilization of microtubules, whereas specific truncated fragments are sufficient to induce abnormal hyperphosphorylation and aggregate into toxic oligomers, resulting in them gaining prion-like characteristics. In addition, Tau truncations cause extensive impairments to neural and glial cell functions and animal cognition and behavior in a fragment-dependent manner. This review summarizes over 60 proteolytic cleavage sites and their corresponding truncated fragments, investigates the role of specific truncations in physiological and pathological states of Alzheimer's disease, and summarizes the latest applications of strategies targeting Tau fragments in the diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xingyue Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Jin Miao
- Laboratory of Animal Center, Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
14
|
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
|
15
|
Rose K, Jepson T, Shukla S, Maya-Romero A, Kampmann M, Xu K, Hurley JH. Tau fibrils induce nanoscale membrane damage and nucleate cytosolic tau at lysosomes. Proc Natl Acad Sci U S A 2024; 121:e2315690121. [PMID: 38781206 PMCID: PMC11145263 DOI: 10.1073/pnas.2315690121] [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: 09/09/2023] [Accepted: 04/08/2024] [Indexed: 05/25/2024] Open
Abstract
The prion-like spread of protein aggregates is a leading hypothesis for the propagation of neurofibrillary lesions in the brain, including the spread of tau inclusions associated with Alzheimer's disease. The mechanisms of cellular uptake of tau seeds and subsequent nucleated polymerization of cytosolic tau are major questions in the field, and the potential for coupling between the entry and nucleation mechanisms has been little explored. We found that in primary astrocytes and neurons, endocytosis of tau seeds leads to their accumulation in lysosomes. This in turn leads to lysosomal swelling, deacidification, and recruitment of ESCRT proteins, but not Galectin-3, to the lysosomal membrane. These observations are consistent with nanoscale damage of the lysosomal membrane. Live cell imaging and STORM superresolution microscopy further show that the nucleation of cytosolic tau occurs primarily at the lysosome membrane under these conditions. These data suggest that tau seeds escape from lysosomes via nanoscale damage rather than wholesale rupture and that nucleation of cytosolic tau commences as soon as tau fibril ends emerge from the lysosomal membrane.
Collapse
Affiliation(s)
- Kevin Rose
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
| | - Tyler Jepson
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Graduate Group in Biophysics, University of California, Berkeley, CA94720
| | - Sankalp Shukla
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
| | - Alex Maya-Romero
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA94158
| | - Ke Xu
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Graduate Group in Biophysics, University of California, Berkeley, CA94720
- Department of Chemistry, University of California, Berkeley, CA94720
| | - James H. Hurley
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Graduate Group in Biophysics, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA94720
| |
Collapse
|
16
|
Chen CY, Tseng YH, Ward JP. A mathematical model on the propagation of tau pathology in neurodegenerative diseases. J Math Biol 2024; 89:4. [PMID: 38750128 DOI: 10.1007/s00285-024-02101-z] [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: 09/28/2023] [Revised: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 06/28/2024]
Abstract
A system of partial differential equations is developed to study the spreading of tau pathology in the brain for Alzheimer's and other neurodegenerative diseases. Two cases are considered with one assuming intracellular diffusion through synaptic activities or the nanotubes that connect the adjacent cells. The other, in addition to intracellular spreading, takes into account of the secretion of the tau species which are able to diffuse, move with the interstitial fluid flow and subsequently taken up by the surrounding cells providing an alternative pathway for disease spreading. Cross membrane transport of the tau species are considered enabling us to examine the role of extracellular clearance of tau protein on the disease status. Bifurcation analysis is carried out for the steady states of the spatially homogeneous system yielding the results that fast cross-membrane transport combined with effective extracellular clearance is key to maintain the brain's healthy status. Numerical simulations of the first case exhibit solutions of travelling wave form describing the gradual outward spreading of the pathology; whereas the second case shows faster spreading with the buildup of neurofibrillary tangles quickly elevated throughout. Our investigation thus indicates that the gradual progression of the intracellular spreading case is more consistent with the clinical observations of the development of Alzheimer's disease.
Collapse
Affiliation(s)
- C Y Chen
- Department of Applied Mathematics, National University of Kaohsiung, Kaohsiung, Taiwan.
| | - Y H Tseng
- Department of Applied Mathematics, National University of Kaohsiung, Kaohsiung, Taiwan
| | - J P Ward
- Department of Mathematical Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
17
|
Zheng H, Sun H, Cai Q, Tai HC. The Enigma of Tau Protein Aggregation: Mechanistic Insights and Future Challenges. Int J Mol Sci 2024; 25:4969. [PMID: 38732197 PMCID: PMC11084794 DOI: 10.3390/ijms25094969] [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: 03/30/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Tau protein misfolding and aggregation are pathological hallmarks of Alzheimer's disease and over twenty neurodegenerative disorders. However, the molecular mechanisms of tau aggregation in vivo remain incompletely understood. There are two types of tau aggregates in the brain: soluble aggregates (oligomers and protofibrils) and insoluble filaments (fibrils). Compared to filamentous aggregates, soluble aggregates are more toxic and exhibit prion-like transmission, providing seeds for templated misfolding. Curiously, in its native state, tau is a highly soluble, heat-stable protein that does not form fibrils by itself, not even when hyperphosphorylated. In vitro studies have found that negatively charged molecules such as heparin, RNA, or arachidonic acid are generally required to induce tau aggregation. Two recent breakthroughs have provided new insights into tau aggregation mechanisms. First, as an intrinsically disordered protein, tau is found to undergo liquid-liquid phase separation (LLPS) both in vitro and inside cells. Second, cryo-electron microscopy has revealed diverse fibrillar tau conformations associated with different neurodegenerative disorders. Nonetheless, only the fibrillar core is structurally resolved, and the remainder of the protein appears as a "fuzzy coat". From this review, it appears that further studies are required (1) to clarify the role of LLPS in tau aggregation; (2) to unveil the structural features of soluble tau aggregates; (3) to understand the involvement of fuzzy coat regions in oligomer and fibril formation.
Collapse
Affiliation(s)
| | | | | | - Hwan-Ching Tai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
18
|
Doering S, McCullough A, Gordon BA, Chen CD, McKay N, Hobbs D, Keefe S, Flores S, Scott J, Smith H, Jarman S, Jackson K, Hornbeck RC, Ances BM, Xiong C, Aschenbrenner AJ, Hassenstab J, Cruchaga C, Daniels A, Bateman RJ, Morris JC, Benzinger TLS. Deconstructing pathological tau by biological process in early stages of Alzheimer disease: a method for quantifying tau spatial spread in neuroimaging. EBioMedicine 2024; 103:105080. [PMID: 38552342 PMCID: PMC10995809 DOI: 10.1016/j.ebiom.2024.105080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Neuroimaging studies often quantify tau burden in standardized brain regions to assess Alzheimer disease (AD) progression. However, this method ignores another key biological process in which tau spreads to additional brain regions. We have developed a metric for calculating the extent tau pathology has spread throughout the brain and evaluate the relationship between this metric and tau burden across early stages of AD. METHODS 445 cross-sectional participants (aged ≥ 50) who had MRI, amyloid PET, tau PET, and clinical testing were separated into disease-stage groups based on amyloid positivity and cognitive status (older cognitively normal control, preclinical AD, and symptomatic AD). Tau burden and tau spatial spread were calculated for all participants. FINDINGS We found both tau metrics significantly elevated across increasing disease stages (p < 0.0001) and as a function of increasing amyloid burden for participants with preclinical (p < 0.0001, p = 0.0056) and symptomatic (p = 0.010, p = 0.0021) AD. An interaction was found between tau burden and tau spatial spread when predicting amyloid burden (p = 0.00013). Analyses of slope between tau metrics demonstrated more spread than burden in preclinical AD (β = 0.59), but then tau burden elevated relative to spread (β = 0.42) once participants had symptomatic AD, when the tau metrics became highly correlated (R = 0.83). INTERPRETATION Tau burden and tau spatial spread are both strong biomarkers for early AD but provide unique information, particularly at the preclinical stage. Tau spatial spread may demonstrate earlier changes than tau burden which could have broad impact in clinical trial design. FUNDING This research was supported by the Knight Alzheimer Disease Research Center (Knight ADRC, NIH grants P30AG066444, P01AG026276, P01AG003991), Dominantly Inherited Alzheimer Network (DIAN, NIH grants U01AG042791, U19AG03243808, R01AG052550-01A1, R01AG05255003), and the Barnes-Jewish Hospital Foundation Willman Scholar Fund.
Collapse
Affiliation(s)
- Stephanie Doering
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Nicole McKay
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Diana Hobbs
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Sarah Keefe
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jalen Scott
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Hunter Smith
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Stephen Jarman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Kelley Jackson
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Beau M Ances
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Chengjie Xiong
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Jason Hassenstab
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Alisha Daniels
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
19
|
Martinez-Feduchi P, Jin P, Yao B. Epigenetic modifications of DNA and RNA in Alzheimer's disease. Front Mol Neurosci 2024; 17:1398026. [PMID: 38726308 PMCID: PMC11079283 DOI: 10.3389/fnmol.2024.1398026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder and the most common form of dementia. There are two main types of AD: familial and sporadic. Familial AD is linked to mutations in amyloid precursor protein (APP), presenilin-1 (PSEN1), and presenilin-2 (PSEN2). On the other hand, sporadic AD is the more common form of the disease and has genetic, epigenetic, and environmental components that influence disease onset and progression. Investigating the epigenetic mechanisms associated with AD is essential for increasing understanding of pathology and identifying biomarkers for diagnosis and treatment. Chemical covalent modifications on DNA and RNA can epigenetically regulate gene expression at transcriptional and post-transcriptional levels and play protective or pathological roles in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
20
|
Li W, Li JY. Overlaps and divergences between tauopathies and synucleinopathies: a duet of neurodegeneration. Transl Neurodegener 2024; 13:16. [PMID: 38528629 DOI: 10.1186/s40035-024-00407-y] [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: 11/07/2023] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Proteinopathy, defined as the abnormal accumulation of proteins that eventually leads to cell death, is one of the most significant pathological features of neurodegenerative diseases. Tauopathies, represented by Alzheimer's disease (AD), and synucleinopathies, represented by Parkinson's disease (PD), show similarities in multiple aspects. AD manifests extrapyramidal symptoms while dementia is also a major sign of advanced PD. We and other researchers have sequentially shown the cross-seeding phenomenon of α-synuclein (α-syn) and tau, reinforcing pathologies between synucleinopathies and tauopathies. The highly overlapping clinical and pathological features imply shared pathogenic mechanisms between the two groups of disease. The diagnostic and therapeutic strategies seemingly appropriate for one distinct neurodegenerative disease may also apply to a broader spectrum. Therefore, a clear understanding of the overlaps and divergences between tauopathy and synucleinopathy is critical for unraveling the nature of the complicated associations among neurodegenerative diseases. In this review, we discuss the shared and diverse characteristics of tauopathies and synucleinopathies from aspects of genetic causes, clinical manifestations, pathological progression and potential common therapeutic approaches targeting the pathology, in the aim to provide a timely update for setting the scheme of disease classification and provide novel insights into the therapeutic development for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wen Li
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, China Medical University, Shenyang, 110122, China
| | - Jia-Yi Li
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, China Medical University, Shenyang, 110122, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, BMC A10, 22184, Lund, Sweden.
| |
Collapse
|
21
|
Xu H, Qiu Q, Hu P, Hoxha K, Jang E, O'Reilly M, Kim C, He Z, Marotta N, Changolkar L, Zhang B, Wu H, Schellenberg GD, Kraemer B, Luk KC, Lee EB, Trojanowski JQ, Brunden KR, Lee VMY. MSUT2 regulates tau spreading via adenosinergic signaling mediated ASAP1 pathway in neurons. Acta Neuropathol 2024; 147:55. [PMID: 38472475 PMCID: PMC10933148 DOI: 10.1007/s00401-024-02703-3] [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: 12/06/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Inclusions comprised of microtubule-associated protein tau (tau) are implicated in a group of neurodegenerative diseases, collectively known as tauopathies, that include Alzheimer's disease (AD). The spreading of misfolded tau "seeds" along neuronal networks is thought to play a crucial role in the progression of tau pathology. Consequently, restricting the release or uptake of tau seeds may inhibit the spread of tau pathology and potentially halt the advancement of the disease. Previous studies have demonstrated that the Mammalian Suppressor of Tauopathy 2 (MSUT2), an RNA binding protein, modulates tau pathogenesis in a transgenic mouse model. In this study, we investigated the impact of MSUT2 on tau pathogenesis using tau seeding models. Our findings indicate that the loss of MSUT2 mitigates human tau seed-induced pathology in neuron cultures and mouse models. In addition, MSUT2 regulates many gene transcripts, including the Adenosine Receptor 1 (A1AR), and we show that down regulation or inhibition of A1AR modulates the activity of the "ArfGAP with SH3 Domain, Ankyrin Repeat, and PH Domain 1 protein" (ASAP1), thereby influencing the internalization of pathogenic tau seeds into neurons resulting in reduction of tau pathology.
Collapse
Affiliation(s)
- Hong Xu
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Qi Qiu
- Department of Genetics, Penn Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peng Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education), Shanghai Ocean University, Shanghai, China
| | - Kevt'her Hoxha
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elliot Jang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mia O'Reilly
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Kim
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhuohao He
- Interdisciplinary Research Center On Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Nicholas Marotta
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bin Zhang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, Penn Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Kraemer
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt R Brunden
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [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: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
23
|
Needs HI, Glover E, Pereira GC, Witt A, Hübner W, Dodding MP, Henley JM, Collinson I. Rescue of mitochondrial import failure by intercellular organellar transfer. Nat Commun 2024; 15:988. [PMID: 38307874 PMCID: PMC10837123 DOI: 10.1038/s41467-024-45283-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
Mitochondria are the powerhouses of eukaryotic cells, composed mostly of nuclear-encoded proteins imported from the cytosol. Thus, problems with the import machinery will disrupt their regenerative capacity and the cell's energy supplies - particularly troublesome for energy-demanding cells of nervous tissue and muscle. Unsurprisingly then, import breakdown is implicated in disease. Here, we explore the consequences of import failure in mammalian cells; wherein, blocking the import machinery impacts mitochondrial ultra-structure and dynamics, but, surprisingly, does not affect import. Our data are consistent with a response involving intercellular mitochondrial transport via tunnelling nanotubes to import healthy mitochondria and jettison those with blocked import sites. These observations support the existence of a widespread mechanism for the rescue of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hope I Needs
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Emily Glover
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Gonçalo C Pereira
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
- Nanna Therapeutics, Merrifield Centre, Rosemary Lane, Cambridge, CB1 3LQ, UK
| | - Alina Witt
- Fakultät für Physik, Universität Bielefeld, Bielefeld, Postfach 100131 D-33501, Germany
| | - Wolfgang Hübner
- Fakultät für Physik, Universität Bielefeld, Bielefeld, Postfach 100131 D-33501, Germany
| | - Mark P Dodding
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Jeremy M Henley
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.
| | - Ian Collinson
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
24
|
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
|
25
|
Bartak M, Bąska P, Chodkowski M, Tymińska B, Bańbura MW, Cymerys J. Neurons cytoskeletal architecture remodeling during the replication cycle of mouse coronavirus MHV-JHM: a morphological in vitro study. BMC Vet Res 2024; 20:18. [PMID: 38195523 PMCID: PMC10775625 DOI: 10.1186/s12917-023-03813-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/10/2023] [Indexed: 01/11/2024] Open
Abstract
Nowadays, the population is still struggling with a post-COVID19 syndrome known as long COVID, including a broad spectrum of neurological problems. There is an urgent need for a better understanding and exploration of the mechanisms of coronavirus neurotropism. For this purpose, the neurotropic strain of mouse hepatitis virus (MHV-JHM) originating from the beta-coronavirus genus, the same as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been used. The role of the cytoskeleton during virus replication in neurons in vitro was determined to understand the mechanisms of MHV-JHM neuroinfection. We have described for the first time the changes of actin filaments during MHV-JHM infection. We also observed productive replication of MHV-JHM in neurons during 168 h p.i. and syncytial cytopathic effect. We discovered that the MHV-JHM strain modulated neuronal cytoskeleton during infection, which were manifested by: (i) condensation of actin filaments in the cortical layer of the cytoplasm, (ii) formation of microtubule cisternae structures containing viral antigen targeting viral replication site (iii) formation of tunneling nanotubes used by MHV-JHM for intercellular transport. Additionally, we demonstrated that the use of cytoskeletal inhibitors have reduced virus replication in neurons, especially noscapine and nocodazole, the microtubule shortening factors.
Collapse
Affiliation(s)
- Michalina Bartak
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland.
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8 St., Warsaw, 02-786, Poland
| | - Marcin Chodkowski
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Kozielska 4 St., Warsaw, 01-063, Poland
| | - Beata Tymińska
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland
| | - Marcin W Bańbura
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland
| | - Joanna Cymerys
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland.
| |
Collapse
|
26
|
Valdebenito S, Ajasin D, Valerdi K, Liu YR, Rao S, Eugenin EA. Mechanisms of Intracellular Communication in Cancer and Pathogen Spreading. Results Probl Cell Differ 2024; 73:301-326. [PMID: 39242384 DOI: 10.1007/978-3-031-62036-2_13] [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
Cell-to-cell interactions are essential for proper development, homeostasis, and complex syncytia/organ formation and function. Intercellular communication are mediated by multiple mechanisms including soluble mediators, adhesion molecules and specific mechanisms of cell to cell communication such as Gap junctions (GJ), tunneling nanotubes (TNT), and exosomes. Only recently, has been discovered that TNTs and exosomes enable the exchange of large signaling molecules, RNA, viral products, antigens, and organelles opening new avenues of research and therapeutic approaches. The focus of this review is to summarize these recent findings in physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - David Ajasin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Karl Valerdi
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | | | - Samvrit Rao
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, USA
| | - Eliseo A Eugenin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA.
| |
Collapse
|
27
|
Szabó-Meleg E. Intercellular Highways in Transport Processes. Results Probl Cell Differ 2024; 73:173-201. [PMID: 39242380 DOI: 10.1007/978-3-031-62036-2_9] [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: 09/09/2024]
Abstract
Communication among cells is vital in multicellular organisms. Various structures and mechanisms have evolved over time to achieve the intricate flow of material and information during this process. One such way of communication is through tunnelling membrane nanotubes (TNTs), which were initially described in 2004. These TNTs are membrane-bounded actin-rich cellular extensions, facilitating direct communication between distant cells. They exhibit remarkable diversity in terms of structure, morphology, and function, in which cytoskeletal proteins play an essential role. Biologically, TNTs play a crucial role in transporting membrane components, cell organelles, and nucleic acids, and they also present opportunities for the efficient transmission of bacteria and viruses, furthermore, may contribute to the dissemination of misfolded proteins in certain neurodegenerative diseases. Convincing results of studies conducted both in vitro and in vivo indicate that TNTs play roles in various biomedical processes, including cell differentiation, tissue regeneration, neurodegenerative diseases, immune response and function, as well as tumorigenesis.
Collapse
Affiliation(s)
- Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary.
| |
Collapse
|
28
|
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
|
29
|
Davidson R, Krider RI, Borsellino P, Noorda K, Alhwayek G, Vida TA. Untangling Tau: Molecular Insights into Neuroinflammation, Pathophysiology, and Emerging Immunotherapies. Curr Issues Mol Biol 2023; 45:8816-8839. [PMID: 37998730 PMCID: PMC10670294 DOI: 10.3390/cimb45110553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
Neuroinflammation, a core pathological feature observed in several neurodegenerative diseases, including Alzheimer's disease (AD), is rapidly gaining attention as a target in understanding the molecular underpinnings of these disorders. Glial cells, endothelial cells, peripheral immune cells, and astrocytes produce a variety of pro-inflammatory mediators that exacerbate the disease progression. Additionally, microglial cells play a complex role in AD, facilitating the clearance of pathological amyloid-beta peptide (Aβ) plaques and aggregates of the tau protein. Tau proteins, traditionally associated with microtubule stabilization, have come under intense scrutiny for their perturbed roles in neurodegenerative conditions. In this narrative review, we focus on recent advances from molecular insights that have revealed aberrant tau post-translational modifications, such as phosphorylation and acetylation, serving as pathological hallmarks. These modifications also trigger the activation of CNS-resident immune cells, such as microglia and astrocytes substantially contributing to neuroinflammation. This intricate relationship between tau pathologies and neuroinflammation fosters a cascading impact on neural pathophysiology. Furthermore, understanding the molecular mechanisms underpinning tau's influence on neuroinflammation presents a frontier for the development of innovative immunotherapies. Neurodegenerative diseases have been relatively intractable to conventional pharmacology using small molecules. We further comprehensively document the many alternative approaches using immunotherapy targeting tau pathological epitopes and structures with a wide array of antibodies. Clinical trials are discussed using these therapeutic approaches, which have both promising and disappointing outcomes. Future directions for tau immunotherapies may include combining treatments with Aβ immunotherapy, which may result in more significant clinical outcomes for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (R.D.); (R.I.K.); (P.B.); (K.N.); (G.A.)
| |
Collapse
|
30
|
Schneeweis A, Pak DTS. Wherefore Art Tau? Functional importance of site-specific tau phosphorylation in diverse subcellular domains. Int J Biochem Cell Biol 2023; 164:106475. [PMID: 37778693 DOI: 10.1016/j.biocel.2023.106475] [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: 11/07/2022] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023]
Abstract
Tau has canonically been considered as an axonal protein, but studies have observed tau localization in other subcellular domains of neurons. This relocated tau has been identified in both physiological and pathological conditions, and it is often labeled mislocalized. Furthermore, these forms of tau are referred to as "hyperphosphorylated" without specifying the phosphosites involved. On the contrary, we speculate that tau may have multiple physiological functions in various locations regulated via specific phosphorylation sites, although this picture is obscured by a lack of comprehensive phosphosite analysis. Here, we examine findings in the literature on the subcellular location of tau and potential roles tau has in those regions. We intentionally focus on the site-specific phosphorylated patterns involved in governing these properties, which are not well elucidated. To facilitate understanding of these events, we have begun establishing a comprehensive map of tau phosphorylation signatures. Such efforts may clarify tau's diverse physiological functions beyond the axon as well as promote development of novel therapeutic strategies directed against distinct tau subpopulations.
Collapse
Affiliation(s)
- Amanda Schneeweis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
31
|
Hedna R, DiMaio A, Robin M, Allegro D, Tatoni M, Peyrot V, Barbier P, Kovacic H, Breuzard G. 2-Aminothiazole-Flavonoid Hybrid Derivatives Binding to Tau Protein and Responsible for Antitumor Activity in Glioblastoma. Int J Mol Sci 2023; 24:15050. [PMID: 37894731 PMCID: PMC10606064 DOI: 10.3390/ijms242015050] [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: 09/12/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Tau protein has been described for several decades as a promoter of tubulin assembly into microtubules. Dysregulation or alterations in Tau expression have been related to various brain cancers, including the highly aggressive and lethal brain tumor glioblastoma multiform (GBM). In this respect, Tau holds significant promise as a target for the development of novel therapies. Here, we examined the structure-activity relationship of a new series of seventeen 2-aminothiazole-fused to flavonoid hybrid compounds (TZF) on Tau binding, Tau fibrillation, and cellular effects on Tau-expressing cancer cells. By spectrofluorometric approach, we found that two compounds, 2 and 9, demonstrated high affinity for Tau and exhibited a strong propensity to inhibit Tau fibrillation. Then, the biological activity of these compounds was evaluated on several Tau-expressing cells derived from glioblastoma. The two lead compounds displayed a high anti-metabolic activity on cells related to an increased fission of the mitochondria network. Moreover, we showed that both compounds induced microtubule bundling within newly formed neurite-like protrusions, as well as with defection of cell migration. Taken together, our results provide a strong experimental basis to develop new potent molecules targeting Tau-expressing cancer cells, such as GBM.
Collapse
Affiliation(s)
- Rayane Hedna
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| | - Attilio DiMaio
- Faculté de Pharmacie, Institut Méditerranéen de Biodiversité et Ecologie Marine et Continentale (IMBE), UMR 7263, CNRS, IRD 237, Aix-Marseille Université, 13005 Marseille, France; (A.D.); (M.R.)
| | - Maxime Robin
- Faculté de Pharmacie, Institut Méditerranéen de Biodiversité et Ecologie Marine et Continentale (IMBE), UMR 7263, CNRS, IRD 237, Aix-Marseille Université, 13005 Marseille, France; (A.D.); (M.R.)
| | - Diane Allegro
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| | - Mario Tatoni
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| | - Vincent Peyrot
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| | - Pascale Barbier
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (R.H.); (D.A.); (M.T.); (V.P.); (P.B.); (H.K.)
| |
Collapse
|
32
|
Hu J, Sha W, Yuan S, Wu J, Huang Y. Aggregation, Transmission, and Toxicity of the Microtubule-Associated Protein Tau: A Complex Comprehension. Int J Mol Sci 2023; 24:15023. [PMID: 37834471 PMCID: PMC10573976 DOI: 10.3390/ijms241915023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The microtubule-associated protein tau is an intrinsically disordered protein containing a few short and transient secondary structures. Tau physiologically associates with microtubules (MTs) for its stabilization and detaches from MTs to regulate its dynamics. Under pathological conditions, tau is abnormally modified, detaches from MTs, and forms protein aggregates in neuronal and glial cells. Tau protein aggregates can be found in a number of devastating neurodegenerative diseases known as "tauopathies", such as Alzheimer's disease (AD), frontotemporal dementia (FTD), corticobasal degeneration (CBD), etc. However, it is still unclear how the tau protein is compacted into ordered protein aggregates, and the toxicity of the aggregates is still debated. Fortunately, there has been considerable progress in the study of tau in recent years, particularly in the understanding of the intercellular transmission of pathological tau species, the structure of tau aggregates, and the conformational change events in the tau polymerization process. In this review, we summarize the concepts of tau protein aggregation and discuss the views on tau protein transmission and toxicity.
Collapse
Affiliation(s)
- Jiaxin Hu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
| | - Wenchi Sha
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
| | - Shuangshuang Yuan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
| | - Jiarui Wu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yunpeng Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
33
|
Dagar S, Subramaniam S. Tunneling Nanotube: An Enticing Cell-Cell Communication in the Nervous System. BIOLOGY 2023; 12:1288. [PMID: 37886998 PMCID: PMC10604474 DOI: 10.3390/biology12101288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
The field of neuroscience is rapidly progressing, continuously uncovering new insights and discoveries. Among the areas that have shown immense potential in research, tunneling nanotubes (TNTs) have emerged as a promising subject of study. These minute structures act as conduits for the transfer of cellular materials between cells, representing a mechanism of communication that holds great significance. In particular, the interplay facilitated by TNTs among various cell types within the brain, including neurons, astrocytes, oligodendrocytes, glial cells, and microglia, can be essential for the normal development and optimal functioning of this complex organ. The involvement of TNTs in neurodegenerative disorders, such as Alzheimer's disease, Huntington's disease, and Parkinson's disease, has attracted significant attention. These disorders are characterized by the progressive degeneration of neurons and the subsequent decline in brain function. Studies have predicted that TNTs likely play critical roles in the propagation and spread of pathological factors, contributing to the advancement of these diseases. Thus, there is a growing interest in understanding the precise functions and mechanisms of TNTs within the nervous system. This review article, based on our recent work on Rhes-mediated TNTs, aims to explore the functions of TNTs within the brain and investigate their implications for neurodegenerative diseases. Using the knowledge gained from studying TNTs could offer novel opportunities for designing targeted treatments that can stop the progression of neurodegenerative disorders.
Collapse
Affiliation(s)
- Sunayana Dagar
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Srinivasa Subramaniam
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
- The Scripps Research Institute, La Jolla, CA 92037, USA
- Norman Fixel Institute for Neurological Diseases, 130 Scripps Way, C323, Jupiter, FL 33458, USA
| |
Collapse
|
34
|
Rose K, Jepson T, Shukla S, Maya-Romero A, Kampmann M, Xu K, Hurley JH. Tau fibrils induce nanoscale membrane damage and nucleate cytosolic tau at lysosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555157. [PMID: 37693477 PMCID: PMC10491128 DOI: 10.1101/2023.08.28.555157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The prion-like spread of protein aggregates is a leading hypothesis for the propagation of neurofibrillary lesions in the brain, including the spread of tau inclusions associated with Alzheimer's disease. The mechanisms of cellular uptake of tau seeds and subsequent nucleated polymerization of cytosolic tau are major questions in the field, and the potential for coupling between the entry and nucleation mechanisms has been little explored. We found that in primary astrocytes, endocytosis of tau seeds leads to their accumulation in lysosomes. This in turn leads to lysosomal swelling, deacidification and recruitment of ESCRT proteins, but not Galectin-3, to the lysosomal membrane. These observations are consistent with nanoscale damage of the lysosomal membrane. Using live cell and STORM, imaging, nucleation of cytosolic tau occurs primarily at the lysosome membrane under these conditions. These data suggest that tau seeds escape from lysosomes via nanoscale damage rather than wholesale rupture, and that nucleation of cytosolic tau commences as soon as tau fibril ends emerge from the lysosomal membrane.
Collapse
Affiliation(s)
- Kevin Rose
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Tyler Jepson
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, University of California, Berkeley, CA 94720
| | - Sankalp Shukla
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Alex Maya-Romero
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California 94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158
| | - Ke Xu
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, University of California, Berkeley, CA 94720
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - James H. Hurley
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, University of California, Berkeley, CA 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720
| |
Collapse
|
35
|
Capobianco DL, Simone L, Svelto M, Pisani F. Intercellular crosstalk mediated by tunneling nanotubes between central nervous system cells. What we need to advance. Front Physiol 2023; 14:1214210. [PMID: 37670766 PMCID: PMC10475722 DOI: 10.3389/fphys.2023.1214210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/11/2023] [Indexed: 09/07/2023] Open
Abstract
Long-range intercellular communication between Central Nervous System (CNS) cells is an essential process for preserving CNS homeostasis. Paracrine signaling, extracellular vesicles, neurotransmitters and synapses are well-known mechanisms involved. A new form of intercellular crosstalk mechanism based on Tunneling Nanotubes (TNTs), suggests a new way to understand how neural cells interact with each other in controlling CNS functions. TNTs are long intercellular bridges that allow the intercellular transfer of cargoes and signals from one cell to another contributing to the control of tissue functionality. CNS cells communicate with each other via TNTs, through which ions, organelles and other signals are exchanged. Unfortunately, almost all these results were obtained through 2D in-vitro models, and fundamental mechanisms underlying TNTs-formation still remain elusive. Consequently, many questions remain open, and TNTs role in CNS remains largely unknown. In this review, we briefly discuss the state of the art regarding TNTs identification and function. We highlight the gaps in the knowledge of TNTs and discuss what is needed to accelerate TNTs-research in CNS-physiology. To this end, it is necessary to: 1) Develop an ad-hoc TNTs-imaging and software-assisted processing tool to improve TNTs-identification and quantification, 2) Identify specific molecular pathways involved into TNTs-formation, 3) Use in-vitro 3D-CNS and animal models to investigate TNTs-role in a more physiological context pushing the limit of live-microscopy techniques. Although there are still many steps to be taken, we believe that the study of TNTs is a new and fascinating frontier that could significantly contribute to deciphering CNS physiology.
Collapse
Affiliation(s)
- D. L. Capobianco
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Bari, Italy
| | - L. Simone
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Casa Sollievo della Sofferenza, Cancer Stem Cells Unit, San Giovanni Rotondo, Italy
| | - M. Svelto
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Bari, Italy
| | - F. Pisani
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Bari, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
36
|
Flavin WP, Hosseini H, Ruberti JW, Kavehpour HP, Giza CC, Prins ML. Traumatic brain injury and the pathways to cerebral tau accumulation. Front Neurol 2023; 14:1239653. [PMID: 37638180 PMCID: PMC10450935 DOI: 10.3389/fneur.2023.1239653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Tau is a protein that has received national mainstream recognition for its potential negative impact to the brain. This review succinctly provides information on the structure of tau and its normal physiological functions, including in hibernation and changes throughout the estrus cycle. There are many pathways involved in phosphorylating tau including diabetes, stroke, Alzheimer's disease (AD), brain injury, aging, and drug use. The common mechanisms for these processes are put into context with changes observed in mild and repetitive mild traumatic brain injury (TBI). The phosphorylation of tau is a part of the progression to pathology, but the ability for tau to aggregate and propagate is also addressed. Summarizing both the functional and dysfunctional roles of tau can help advance our understanding of this complex protein, improve our care for individuals with a history of TBI, and lead to development of therapeutic interventions to prevent or reverse tau-mediated neurodegeneration.
Collapse
Affiliation(s)
- William P. Flavin
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Helia Hosseini
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
| | - Jeffrey W. Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - H. Pirouz Kavehpour
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Mechanical and Aerospace Engineering, UCLA, Los Angeles, CA, United States
| | - Christopher C. Giza
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Mayumi L. Prins
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
37
|
Needs HI, Wilkinson KA, Henley JM, Collinson I. Aggregation-prone Tau impairs mitochondrial import, which affects organelle morphology and neuronal complexity. J Cell Sci 2023; 136:jcs260993. [PMID: 37303235 PMCID: PMC10357015 DOI: 10.1242/jcs.260993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
Mitochondrial protein import is essential for organellar biogenesis, and thereby for the sufficient supply of cytosolic ATP - which is particularly important for cells with high energy demands like neurons. This study explores the prospect of import machinery perturbation as a cause of neurodegeneration instigated by the accumulation of aggregating proteins linked to disease. We found that the aggregation-prone Tau variant (TauP301L) reduces the levels of components of the import machinery of the outer (TOM20, encoded by TOMM20) and inner membrane (TIM23, encoded by TIMM23) while associating with TOM40 (TOMM40). Intriguingly, this interaction affects mitochondrial morphology, but not protein import or respiratory function; raising the prospect of an intrinsic rescue mechanism. Indeed, TauP301L induced the formation of tunnelling nanotubes (TNTs), potentially for the recruitment of healthy mitochondria from neighbouring cells and/or the disposal of mitochondria incapacitated by aggregated Tau. Consistent with this, inhibition of TNT formation (and rescue) reveals Tau-induced import impairment. In primary neuronal cultures, TauP301L induced morphological changes characteristic of neurodegeneration. Interestingly, these effects were mirrored in cells where the import sites were blocked artificially. Our results reveal a link between aggregation-prone Tau and defective mitochondrial import relevant to disease.
Collapse
Affiliation(s)
- Hope I. Needs
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | - Jeremy M. Henley
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Ian Collinson
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
38
|
Arnold FJ, Nguyen AD, Bedlack RS, Bennett CL, La Spada AR. Intercellular transmission of pathogenic proteins in ALS: Exploring the pathogenic wave. Neurobiol Dis 2023:106218. [PMID: 37394036 DOI: 10.1016/j.nbd.2023.106218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023] Open
Abstract
In patients with amyotrophic lateral sclerosis (ALS), disease symptoms and pathology typically spread in a predictable spatiotemporal pattern beginning at a focal site of onset and progressing along defined neuroanatomical tracts. Like other neurodegenerative diseases, ALS is characterized by the presence of protein aggregates in postmortem patient tissue. Cytoplasmic, ubiquitin-positive aggregates of TDP-43 are observed in approximately 97% of sporadic and familial ALS patients, while SOD1 inclusions are likely specific to cases of SOD1-ALS. Additionally, the most common subtype of familial ALS, caused by a hexanucleotide repeat expansion in the first intron of the C9orf72 gene (C9-ALS), is further characterized by the presence of aggregated dipeptide repeat proteins (DPRs). As we will describe, cell-to-cell propagation of these pathological proteins tightly correlates with the contiguous spread of disease. While TDP-43 and SOD1 are capable of seeding protein misfolding and aggregation in a prion-like manner, C9orf72 DPRs appear to induce (and transmit) a 'disease state' more generally. Multiple mechanisms of intercellular transport have been described for all of these proteins, including anterograde and retrograde axonal transport, extracellular vesicle secretion, and macropinocytosis. In addition to neuron-to-neuron transmission, transmission of pathological proteins occurs between neurons and glia. Given that the spread of ALS disease pathology corresponds with the spread of symptoms in patients, the various mechanisms by which ALS-associated protein aggregates propagate through the central nervous system should be closely examined.
Collapse
Affiliation(s)
- F J Arnold
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA; Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - A D Nguyen
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - R S Bedlack
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - C L Bennett
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA; Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - A R La Spada
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA; Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA; Departments of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, Irvine, CA, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; UCI Center for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
39
|
Avallone M, Pardo J, Mergiya TF, Rájová J, Räsänen A, Davidsson M, Åkerblom M, Quintino L, Kumar D, Bramham CR, Björklund T. Visualizing Arc protein dynamics and localization in the mammalian brain using AAV-mediated in situ gene labeling. Front Mol Neurosci 2023; 16:1140785. [PMID: 37415832 PMCID: PMC10321715 DOI: 10.3389/fnmol.2023.1140785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/11/2023] [Indexed: 07/08/2023] Open
Abstract
The activity-regulated cytoskeleton-associated (Arc) protein is essential for synaptic plasticity and memory formation. The Arc gene, which contains remnants of a structural GAG retrotransposon sequence, produces a protein that self-assembles into capsid-like structures harboring Arc mRNA. Arc capsids, released from neurons, have been proposed as a novel intercellular mechanism for mRNA transmission. Nevertheless, evidence for intercellular transport of Arc in the mammalian brain is still lacking. To enable the tracking of Arc molecules from individual neurons in vivo, we devised an adeno-associated virus (AAV) mediated approach to tag the N-terminal of the mouse Arc protein with a fluorescent reporter using CRISPR/Cas9 homologous independent targeted integration (HITI). We show that a sequence coding for mCherry can successfully be knocked in at the 5' end of the Arc open reading frame. While nine spCas9 gene editing sites surround the Arc start codon, the accuracy of the editing was highly sequence-dependent, with only a single target resulting in an in-frame reporter integration. When inducing long-term potentiation (LTP) in the hippocampus, we observed an increase of Arc protein highly correlated with an increase in fluorescent intensity and the number of mCherry-positive cells. By proximity ligation assay (PLA), we demonstrated that the mCherry-Arc fusion protein retains the Arc function by interacting with the transmembrane protein stargazin in postsynaptic spines. Finally, we recorded mCherry-Arc interaction with presynaptic protein Bassoon in mCherry-negative surrounding neurons at close proximity to mCherry-positive spines of edited neurons. This is the first study to provide support for inter-neuronal in vivo transfer of Arc in the mammalian brain.
Collapse
Affiliation(s)
- Martino Avallone
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Joaquín Pardo
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner” (INIBIOLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Tadiwos F. Mergiya
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Jana Rájová
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Atte Räsänen
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Marcus Davidsson
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Malin Åkerblom
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Luis Quintino
- CNS Gene Therapy, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | | | - Clive R. Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
40
|
Padmanabhan S, Manjithaya R. Leaderless secretory proteins of the neurodegenerative diseases via TNTs: a structure-function perspective. Front Mol Neurosci 2023; 16:983108. [PMID: 37396786 PMCID: PMC10308029 DOI: 10.3389/fnmol.2023.983108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
Neurodegenerative disease-causing proteins such as alpha-synuclein, tau, and huntingtin are known to traverse across cells via exosomes, extracellular vesicles and tunneling nanotubes (TNTs). There seems to be good synergy between exosomes and TNTs in intercellular communication. Interestingly, many of the known major neurodegenerative proteins/proteolytic products are leaderless and are also reported to be secreted out of the cell via unconventional protein secretion. Such classes contain intrinsically disordered proteins and regions (IDRs) within them. The dynamic behavior of these proteins is due to their heterogenic conformations that is exhibited owing to various factors that occur inside the cells. The amino acid sequence along with the chemical modifications has implications on the functional roles of IDRs inside the cells. Proteins that form aggregates resulting in neurodegeneration become resistant to degradation by the processes of autophagy and proteasome system thus leading to Tunneling nanotubes, TNT formation. The proteins that traverse across TNTs may or may not be dependent on the autophagy machinery. It is not yet clear whether the conformation of the protein plays a crucial role in its transport from one cell to another without getting degraded. Although there is some experimental data, there are many grey areas which need to be revisited. This review provides a different perspective on the structural and functional aspects of these leaderless proteins that get secreted outside the cell. In this review, attention has been focused on the characteristic features that lead to aggregation of leaderless secretory proteins (from structural-functional aspect) with special emphasis on TNTs.
Collapse
|
41
|
Whitfield JF, Rennie K, Chakravarthy B. Alzheimer's Disease and Its Possible Evolutionary Origin: Hypothesis. Cells 2023; 12:1618. [PMID: 37371088 PMCID: PMC10297544 DOI: 10.3390/cells12121618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aβ1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-β oligomers (AβOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AβOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AβOs from ground zero is supported by Aβ's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AβOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.
Collapse
Affiliation(s)
- James F. Whitfield
- Human Health Therapeutics, National Research Council, Ottawa, ON K1A 0R6, Canada
| | | | | |
Collapse
|
42
|
Valdebenito S, Ono A, Rong L, Eugenin EA. The role of tunneling nanotubes during early stages of HIV infection and reactivation: implications in HIV cure. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:169-186. [PMID: 37476291 PMCID: PMC10355284 DOI: 10.1515/nipt-2022-0015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/30/2022] [Indexed: 07/22/2023]
Abstract
Tunneling nanotubes (TNTs), also called cytonemes or tumor microtubes, correspond to cellular processes that enable long-range communication. TNTs are plasma membrane extensions that form tubular processes that connect the cytoplasm of two or more cells. TNTs are mostly expressed during the early stages of development and poorly expressed in adulthood. However, in disease conditions such as stroke, cancer, and viral infections such as HIV, TNTs proliferate, but their role is poorly understood. TNTs function has been associated with signaling coordination, organelle sharing, and the transfer of infectious agents such as HIV. Here, we describe the critical role and function of TNTs during HIV infection and reactivation, as well as the use of TNTs for cure strategies.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Akira Ono
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Libin Rong
- Department of Mathematics, University of Florida, Gainesville, FL, USA
| | - Eliseo A. Eugenin
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| |
Collapse
|
43
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
44
|
Mazzotta GM, Ceccato N, Conte C. Synucleinopathies Take Their Toll: Are TLRs a Way to Go? Cells 2023; 12:cells12091231. [PMID: 37174631 PMCID: PMC10177040 DOI: 10.3390/cells12091231] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/17/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
The misfolding and subsequent abnormal accumulation and aggregation of α-Synuclein (αSyn) as insoluble fibrils in Lewy bodies and Lewy neurites is the pathological hallmark of Parkinson's disease (PD) and several neurodegenerative disorders. A combination of environmental and genetic factors is linked to αSyn misfolding, among which neuroinflammation is recognized to play an important role. Indeed, a number of studies indicate that a Toll-like receptor (TLR)-mediated neuroinflammation might lead to a dopaminergic neural loss, suggesting that TLRs could participate in the pathogenesis of PD as promoters of immune/neuroinflammatory responses. Here we will summarize our current understanding on the mechanisms of αSyn aggregation and misfolding, focusing on the contribution of TLRs to the progression of α-synucleinopathies and speculating on their link with the non-motor disturbances associated with aging and neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Nadia Ceccato
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, 06100 Perugia, Italy
| |
Collapse
|
45
|
Girolamo F, Lim YP, Virgintino D, Stonestreet BS, Chen XF. Inter-Alpha Inhibitor Proteins Modify the Microvasculature after Exposure to Hypoxia-Ischemia and Hypoxia in Neonatal Rats. Int J Mol Sci 2023; 24:6743. [PMID: 37047713 PMCID: PMC10094872 DOI: 10.3390/ijms24076743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Microvasculature develops during early brain development. Hypoxia-ischemia (HI) and hypoxia (H) predispose to brain injury in neonates. Inter-alpha inhibitor proteins (IAIPs) attenuate injury to the neonatal brain after exposure to HI. However, the effects of IAIPs on the brain microvasculature after exposure to HI have not been examined in neonates. Postnatal day-7 rats were exposed to sham treatment or right carotid artery ligation and 8% oxygen for 90 min. HI comprises hypoxia (H) and ischemia to the right hemisphere (HI-right) and hypoxia to the whole body, including the left hemisphere (H-left). Human IAIPs (hIAIPs, 30 mg/kg) or placebo were injected immediately, 24 and 48 h after HI/H. The brains were analyzed 72 h after HI/H to determine the effects of hIAIPs on the microvasculature by laminin immunohistochemistry and calculation of (1) the percentage area stained by laminin, (2) cumulative microvessel length, and (3) density of tunneling nanotubes (TNTs), which are sensitive indicators of the earliest phases of neo-vascularization/collateralization. hIAIPs mainly affected the percent of the laminin-stained area after HI/H, cumulative vessel length after H but not HI, and TNT density in females but not males. hIAIPs modify the effects of HI/H on the microvasculature after brain injury in neonatal rats and exhibit sex-related differential effects. Our findings suggest that treatment with hIAIPs after exposure to H and HI in neonatal rats affects the laminin content of the vessel basal lamina and angiogenic responses in a sex-related fashion.
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Daniela Virgintino
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi F. Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
46
|
Mu S, Turner NL, Silversmith WM, Jordan CS, Kemnitz N, Sorek M, David C, Jones DL, Bland D, Moore M, Sterling AR, Seung HS. Special nuclear layer contacts between starburst amacrine cells in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1129463. [PMID: 38983098 PMCID: PMC11182129 DOI: 10.3389/fopht.2023.1129463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/07/2023] [Indexed: 07/11/2024]
Abstract
Starburst amacrine cells are a prominent neuron type in the mammalian retina that has been well-studied for its role in direction-selective information processing. One specific property of these cells is that their dendrites tightly stratify at specific depths within the inner plexiform layer (IPL), which, together with their unique expression of choline acetyltransferase (ChAT), has made them the most common depth marker for studying other retinal neurons in the IPL. This stratifying property makes it unexpected that they could routinely have dendrites reaching into the nuclear layer or that they could have somatic contact specializations, which is exactly what we have found in this study. Specifically, an electron microscopic image volume of sufficient size from a mouse retina provided us with the opportunity to anatomically observe both microscopic details and collective patterns, and our detailed cell reconstructions revealed interesting cell-cell contacts between starburst amacrine neurons. The contact characteristics differ between the respective On and Off starburst amacrine subpopulations, but both occur within the soma layers, as opposed to their regular contact laminae within the inner plexiform layer.
Collapse
Affiliation(s)
- Shang Mu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Nicholas L. Turner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
- Computer Science Department, Princeton University, Princeton, NJ, United States
| | | | - Chris S. Jordan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Nico Kemnitz
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Marissa Sorek
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Celia David
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Devon L. Jones
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Doug Bland
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Merlin Moore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - Amy Robinson Sterling
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| | - H. Sebastian Seung
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
- Computer Science Department, Princeton University, Princeton, NJ, United States
| |
Collapse
|
47
|
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
|
48
|
Hebisch M, Klostermeier S, Wolf K, Boccaccini AR, Wolf SE, Tanzi RE, Kim DY. The Impact of the Cellular Environment and Aging on Modeling Alzheimer's Disease in 3D Cell Culture Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205037. [PMID: 36642841 PMCID: PMC10015857 DOI: 10.1002/advs.202205037] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/29/2022] [Indexed: 06/13/2023]
Abstract
Creating a cellular model of Alzheimer's disease (AD) that accurately recapitulates disease pathology has been a longstanding challenge. Recent studies showed that human AD neural cells, integrated into three-dimensional (3D) hydrogel matrix, display key features of AD neuropathology. Like in the human brain, the extracellular matrix (ECM) plays a critical role in determining the rate of neuropathogenesis in hydrogel-based 3D cellular models. Aging, the greatest risk factor for AD, significantly alters brain ECM properties. Therefore, it is important to understand how age-associated changes in ECM affect accumulation of pathogenic molecules, neuroinflammation, and neurodegeneration in AD patients and in vitro models. In this review, mechanistic hypotheses is presented to address the impact of the ECM properties and their changes with aging on AD and AD-related dementias. Altered ECM characteristics in aged brains, including matrix stiffness, pore size, and composition, will contribute to disease pathogenesis by modulating the accumulation, propagation, and spreading of pathogenic molecules of AD. Emerging hydrogel-based disease models with differing ECM properties provide an exciting opportunity to study the impact of brain ECM aging on AD pathogenesis, providing novel mechanistic insights. Understanding the role of ECM aging in AD pathogenesis should also improve modeling AD in 3D hydrogel systems.
Collapse
Affiliation(s)
- Matthias Hebisch
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Stefanie Klostermeier
- Institute of Medical PhysicsFriedrich‐Alexander Universität Erlangen‐Nürnberg91052ErlangenGermany
- Max‐Planck‐Zentrum für Physik und Medizin91054ErlangenGermany
| | - Katharina Wolf
- Department of Medicine 1Friedrich‐Alexander‐Universität Erlangen‐Nürnberg91054ErlangenGermany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg91058ErlangenGermany
| | - Stephan E. Wolf
- Institute of Glass and CeramicsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg91058ErlangenGermany
| | - Rudolph E. Tanzi
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Doo Yeon Kim
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| |
Collapse
|
49
|
Okumura H, Kawasaki T, Nakamura K. Probing protein misfolding and dissociation with an infrared free-electron laser. Methods Enzymol 2022; 679:65-96. [PMID: 36682873 DOI: 10.1016/bs.mie.2022.08.047] [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: 12/24/2022]
Abstract
Misfolding is observed in the mutant proteins that are causative for neurodegenerative disorders such as polyglutamine diseases. These proteins are prone to aggregate in the cytoplasm and nucleus of cells. To reproduce cells with the aggregated proteins, gene expression system is usually applied, in which the expression construct having the mutated DNA sequence of the interest is transfected into cells. The transfected DNA is finally converted into the mutant protein, which is gradually aggregated in the cells. In addition, a simple method to prepare the cells having aggregates inside has been recently applied. Peptides were first aggregated by incubating them in water. The aggregates are spontaneously taken up by cells because aggregated proteins generally transfer between cells. Peptides with different degrees of aggregation can be made by changing the incubation times and temperatures, which enables to examine contribution of aggregation to the toxicity to the recipient cells. Moreover, such cells can be used for therapeutic researches of diseases in which aggregates are involved. In this chapter, we show methods to induce aggregation of peptides. The functional analyses of the cells with aggregates are also described. Then, experimental dissociation of the aggregates produced using this method by mid infrared free electron laser irradiation and its theoretical support by molecular dynamics simulation are introduced as the therapeutic research for neurodegenerative disorders.
Collapse
Affiliation(s)
- Hisashi Okumura
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan; Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Aichi, Japan; Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Takayasu Kawasaki
- Accelerator Laboratory, High Energy Accelerator Research Organization, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Nakamura
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Gunma, Japan.
| |
Collapse
|
50
|
Common and Specific Marks of Different Tau Strains Following Intra-Hippocampal Injection of AD, PiD, and GGT Inoculum in hTau Transgenic Mice. Int J Mol Sci 2022; 23:ijms232415940. [PMID: 36555581 PMCID: PMC9787745 DOI: 10.3390/ijms232415940] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Heterozygous hTau mice were used for the study of tau seeding. These mice express the six human tau isoforms, with a high predominance of 3Rtau over 4Rtau. The following groups were assessed: (i) non-inoculated mice aged 9 months (n = 4); (ii) Alzheimer's Disease (AD)-inoculated mice (n = 4); (iii) Globular Glial Tauopathy (GGT)-inoculated mice (n = 4); (iv) Pick's disease (PiD)-inoculated mice (n = 4); (v) control-inoculated mice (n = 4); and (vi) inoculated with vehicle alone (n = 2). AD-inoculated mice showed AT8-immunoreactive neuronal pre-tangles, granular aggregates, and dots in the CA1 region of the hippocampus, dentate gyrus (DG), and hilus, and threads and dots in the ipsilateral corpus callosum. GGT-inoculated mice showed unique or multiple AT8-immunoreactive globular deposits in neurons, occasionally extended to the proximal dendrites. PiD-inoculated mice showed a few loose pre-tangles in the CA1 region, DG, and cerebral cortex near the injection site. Coiled bodies were formed in the corpus callosum in AD-inoculated mice, but GGT-inoculated mice lacked globular glial inclusions. Tau deposits in inoculated mice co-localized active kinases p38-P and SAPK/JNK-P, thus suggesting active phosphorylation of the host tau. Tau deposits were absent in hTau mice inoculated with control homogenates and vehicle alone. Deposits in AD-inoculated hTau mice contained 3Rtau and 4Rtau; those in GGT-inoculated mice were mainly stained with anti-4Rtau antibodies, but a small number of deposits contained 3Rtau. Deposits in PiD-inoculated mice were stained with anti-3Rtau antibodies, but rare neuronal, thread-like, and dot-like deposits showed 4Rtau immunoreactivity. These findings show that tau strains produce different patterns of active neuronal seeding, which also depend on the host tau. Unexpected 3Rtau and 4Rtau deposits after inoculation of homogenates from 4R and 3R tauopathies, respectively, suggests the regulation of exon 10 splicing of the host tau during the process of seeding, thus modulating the plasticity of the cytoskeleton.
Collapse
|