1
|
Cidre-Aranaz F, Magrin C, Zimmermann M, Li J, Baffa A, Ciccaldo M, Hartmann W, Dirksen U, Sola M, Paganetti P, Grünewald TGP, Papin S. High Tau expression correlates with reduced invasion and prolonged survival in Ewing sarcoma. Cell Death Discov 2025; 11:216. [PMID: 40319030 PMCID: PMC12049433 DOI: 10.1038/s41420-025-02497-7] [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: 03/24/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
The microtubule-associated protein Tau (encoded by the MAPT gene) is linked to a family of neurodegenerative disorders defined as tauopathies, which are characterized by its brain accumulation in neurofibrillary tangles and neuropil threads. Newly described Tau functions comprise DNA protection, chromatin remodeling, p53 regulation and cell fate modulation, suggesting a role of Tau in oncogenesis. Bioinformatic-supported characterization of Tau in cancer reveals robust expression in bone cancer cells, in particular Ewing sarcoma (EwS) cell lines. EwS is an aggressive cancer caused by a fusion of members of the FET and ETS gene families, primarily EWSR1::FLI1. Here we found that MAPT is a EWSR1::ETS target gene and that higher Tau expression in EwS cells inhibited their migratory and invasive behavior, consistent with a more immobile and proliferative phenotype observed in EwS. Indeed, we report that high Tau expression is associated with improved overall survival of EwS patients. We also show that the sessile but proliferative phenotype of EWSR1::ETS-high cells may result from a modulatory role of Tau on focal adhesion to extracellular matrix proteins. Our data highlight the utility of determining Tau expression as a prognostic factor in EwS as well as the opportunity to target Tau expression as an innovative EwS therapy.
Collapse
Affiliation(s)
- Florencia Cidre-Aranaz
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Claudia Magrin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Malenka Zimmermann
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Jing Li
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Annalisa Baffa
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Matteo Ciccaldo
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Uta Dirksen
- Pediatrics III, University Hospital Essen, West German Cancer Center, German Cancer Consortium (DKTK) site Essen, National Center for Tumor Diseases (NCT) West, Essen, Germany
| | - Martina Sola
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Paolo Paganetti
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Thomas G P Grünewald
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Stéphanie Papin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| |
Collapse
|
2
|
Sugiyama M, Kosik KS, Panagiotou E. Geometry based prediction of tau protein sites and motifs associated with misfolding and aggregation. Sci Rep 2025; 15:10283. [PMID: 40133414 PMCID: PMC11937417 DOI: 10.1038/s41598-025-93304-x] [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: 09/18/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Recent studies of tau proteins point to specific sites or motifs along the protein related to its misfolding and aggregation propensity, which is associated with neurodegenerative diseases of structure-dependent pathology. In this manuscript we employ topology and geometry to analyze the local structure of tau proteins obtained from the Protein Data Bank. Our results show that mathematical topology/geometry of cryo-EM structures alone identify the PGGG motifs, and the PHF6(*) motifs as sites of interest and reveal a geometrical hierarchy of the PGGG motifs that differs for 3R+4R and 4R tauopathies. By employing the Local Topological Free Energy (LTE), we find that progressive supranuclear palsy (PSP) and globular glial tauopathy (GGT) have the highest LTE values around residues 302-305, which are inside the jR2R3 peptide and in the vicinity of the 301 site, experimentally associated with aggregation. By extending the LTE definition to estimate a global topological free energy, we find that the jR2R3 peptide of PSP and GGT, has in fact the lowest global topological free energy among other tauopathies. These results point to a possible correlation between the global topological free energy of parts of the protein and the LTE of specific sites.
Collapse
Affiliation(s)
- Masumi Sugiyama
- Department of Mathematics, University of Tennessee at Chattanooga, Chattanooga, TN, 37403, USA
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM2), Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Kenneth S Kosik
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Eleni Panagiotou
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, 85281, USA.
| |
Collapse
|
3
|
Chew CS, Lee JY, Ng KY, Koh RY, Chye SM. Resilience mechanisms underlying Alzheimer's disease. Metab Brain Dis 2025; 40:86. [PMID: 39760900 DOI: 10.1007/s11011-024-01507-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 12/14/2024] [Indexed: 01/07/2025]
Abstract
Alzheimer's disease (AD) consists of two main pathologies, which are the deposition of amyloid plaque as well as tau protein aggregation. Evidence suggests that not everyone who carries the AD-causing genes displays AD-related symptoms; they might never acquire AD as well. These individuals are referred to as non-demented individuals with AD neuropathology (NDAN). Despite the presence of extensive AD pathology in their brain, it was found that NDAN had better cognitive function than was expected, suggesting that they were more resilient (better at coping) to AD due to differences in their brains compared to other demented or cognitively impaired patients. Thus, identification of the mechanisms underlying resilience is crucial since it represents a promising therapeutic strategy for AD. In this review, we will explore the molecular mechanisms underpinning the role of genetic and molecular resilience factors in improving resilience to AD. These include protective genes and proteins such as APOE2, BDNF, RAB10, actin network proteins, scaffolding proteins, and the basal forebrain cholinergic system. A thorough understanding of these resilience mechanisms is crucial for not just comprehending the development of AD but may also open new treatment possibilities for AD by enhancing the neuroprotective pathway and targeting the pathogenic process.
Collapse
Affiliation(s)
- Chu Shi Chew
- School of Health Science, IMU University, 57000, Kuala Lumpur, Malaysia
| | - Jia Yee Lee
- School of Health Science, IMU University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, IMU University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, IMU University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
4
|
Chen X, Xu S, Chu B, Guo J, Zhang H, Sun S, Song L, Feng XQ. Applying Spatiotemporal Modeling of Cell Dynamics to Accelerate Drug Development. ACS NANO 2024; 18:29311-29336. [PMID: 39420743 DOI: 10.1021/acsnano.4c12599] [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: 10/19/2024]
Abstract
Cells act as physical computational programs that utilize input signals to orchestrate molecule-level protein-protein interactions (PPIs), generating and responding to forces, ultimately shaping all of the physiological and pathophysiological behaviors. Genome editing and molecule drugs targeting PPIs hold great promise for the treatments of diseases. Linking genes and molecular drugs with protein-performed cellular behaviors is a key yet challenging issue due to the wide range of spatial and temporal scales involved. Building predictive spatiotemporal modeling systems that can describe the dynamic behaviors of cells intervened by genome editing and molecular drugs at the intersection of biology, chemistry, physics, and computer science will greatly accelerate pharmaceutical advances. Here, we review the mechanical roles of cytoskeletal proteins in orchestrating cellular behaviors alongside significant advancements in biophysical modeling while also addressing the limitations in these models. Then, by integrating generative artificial intelligence (AI) with spatiotemporal multiscale biophysical modeling, we propose a computational pipeline for developing virtual cells, which can simulate and evaluate the therapeutic effects of drugs and genome editing technologies on various cell dynamic behaviors and could have broad biomedical applications. Such virtual cell modeling systems might revolutionize modern biomedical engineering by moving most of the painstaking wet-laboratory effort to computer simulations, substantially saving time and alleviating the financial burden for pharmaceutical industries.
Collapse
Affiliation(s)
- Xindong Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
- BioMap, Beijing 100144, China
| | - Shihao Xu
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bizhu Chu
- School of Pharmacy, Shenzhen University, Shenzhen 518055, China
- Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Guo
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Huikai Zhang
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Shuyi Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Le Song
- BioMap, Beijing 100144, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
5
|
Höhler M, Alcázar-Román AR, Schenk K, Aguirre-Huamani MP, Braun C, Zrieq R, Mölleken K, Hegemann JH, Fleig U. Direct targeting of host microtubule and actin cytoskeletons by a chlamydial pathogenic effector protein. J Cell Sci 2024; 137:jcs263450. [PMID: 39099397 PMCID: PMC11444262 DOI: 10.1242/jcs.263450] [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: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
To propagate within a eukaryotic cell, pathogenic bacteria hijack and remodulate host cell functions. The Gram-negative obligate intracellular Chlamydiaceae, which pose a serious threat to human and animal health, attach to host cells and inject effector proteins that reprogram host cell machineries. Members of the conserved chlamydial TarP family have been characterized as major early effectors that bind to and remodel the host actin cytoskeleton. We now describe a new function for the Chlamydia pneumoniae TarP member CPn0572, namely the ability to bind and alter the microtubule cytoskeleton. Thus, CPn0572 is unique in being the only prokaryotic protein that directly modulates both dynamic cytoskeletons of a eukaryotic cell. Ectopically expressed GFP-CPn0572 associates in a dose-independent manner with either cytoskeleton singly or simultaneously. In vitro, CPn0572 binds directly to microtubules. Expression of a microtubule-only CPn0572 variant resulted in the formation of an aberrantly thick, stabilized microtubule network. Intriguingly, during infection, secreted CPn0572 also colocalized with altered microtubules, suggesting that this protein also affects microtubule dynamics during infection. Our analysis points to a crosstalk between actin and microtubule cytoskeletons via chlamydial CPn0572.
Collapse
Affiliation(s)
- Mona Höhler
- Eukaryotic Microbiology, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | | | - Katharina Schenk
- Eukaryotic Microbiology, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | | | - Corinna Braun
- Institute of Functional Microbial Genomics, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Rafat Zrieq
- Department of Public Health, College of Public Health and Health Informatics, University of Ha'il, Ha'il City 2440, Saudi Arabia
- Applied Science Research Centre, Applied Science Private University, Amman 11931, Jordan
| | - Katja Mölleken
- Institute of Functional Microbial Genomics, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Johannes H Hegemann
- Institute of Functional Microbial Genomics, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Ursula Fleig
- Eukaryotic Microbiology, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| |
Collapse
|
6
|
deWeever A, Paudel SS, Zhou C, Francis CM, Tambe DT, Frank DW, Balczon R, Stevens T. cUMP elicits interendothelial gap formation during Pseudomonas aeruginosa infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L395-L405. [PMID: 39076085 PMCID: PMC11444506 DOI: 10.1152/ajplung.00164.2023] [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: 05/22/2023] [Revised: 05/08/2024] [Accepted: 06/30/2024] [Indexed: 07/31/2024] Open
Abstract
Pseudomonas aeruginosa utilizes a type 3 secretion system to intoxicate host cells with the nucleotidyl cyclase ExoY. After activation by its host cell cofactor, filamentous actin, ExoY produces purine and pyrimidine cyclic nucleotides, including cAMP, cGMP, and cUMP. ExoY-generated cyclic nucleotides promote interendothelial gap formation, impair motility, and arrest cell growth. The disruptive activities of cAMP and cGMP during the P. aeruginosa infection are established; however, little is known about the function of cUMP. Here, we tested the hypothesis that cUMP contributes to endothelial cell barrier disruption during P. aeruginosa infection. Using a membrane permeable cUMP analog, cUMP-AM, we revealed that during infection with catalytically inactive ExoY, cUMP promotes interendothelial gap formation in cultured pulmonary microvascular endothelial cells (PMVECs) and contributes to increased filtration coefficient in the isolated perfused lung. These findings indicate that cUMP contributes to endothelial permeability during P. aeruginosa lung infection.NEW & NOTEWORTHY During pneumonia, bacteria utilize a virulence arsenal to communicate with host cells. The Pseudomonas aeruginosa T3SS directly introduces virulence molecules into the host cell cytoplasm. These molecules are enzymes that trigger interkingdom communication. One of the exoenzymes is a nucleotidyl cyclase that produces noncanonical cyclic nucleotides like cUMP. Little is known about how cUMP acts in the cell. Here we found that cUMP instigates pulmonary edema during Pseudomonas aeruginosa infection of the lung.
Collapse
Grants
- R01 HL167997 NHLBI NIH HHS
- HL136689 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL066299 NHLBI NIH HHS
- AI104922 HHS | NIH | NIAID | Division of Microbiology and Infectious Diseases (DMID)
- R01 HL140182 NHLBI NIH HHS
- HL167997 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 AI104922 NIAID NIH HHS
- HL148069 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL148069 NHLBI NIH HHS
- HL140182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL66299 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Althea deWeever
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sunita S Paudel
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Chun Zhou
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - C Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dhananjay T Tambe
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
7
|
Iyer AK, Vermunt L, Mirfakhar FS, Minaya M, Acquarone M, Koppisetti RK, Renganathan A, You SF, Danhash EP, Verbeck A, Galasso G, Lee SM, Marsh J, Nana AL, Spina S, Seeley WW, Grinberg LT, Temple S, Teunissen CE, Sato C, Karch CM. Cell autonomous microglia defects in a stem cell model of frontotemporal dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.15.24307444. [PMID: 38798451 PMCID: PMC11118656 DOI: 10.1101/2024.05.15.24307444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Neuronal dysfunction has been extensively studied as a central feature of neurodegenerative tauopathies. However, across neurodegenerative diseases, there is strong evidence for active involvement of immune cells like microglia in driving disease pathophysiology. Here, we demonstrate that tau mRNA and protein are expressed in microglia in human brains and in human induced pluripotent stem cell (iPSC)-derived microglia like cells (iMGLs). Using iMGLs harboring the MAPT IVS10+16 mutation and isogenic controls, we demonstrate that a tau mutation is sufficient to alter microglial transcriptional states. We discovered that MAPT IVS10+16 microglia exhibit cytoskeletal abnormalities, stalled phagocytosis, disrupted TREM2/TYROBP networks, and altered metabolism. Additionally, we found that secretory factors from MAPT IVS10+16 iMGLs impact neuronal health, reducing synaptic density in neurons. Key features observed in vitro were recapitulated in human brain tissue and cerebrospinal fluid from MAPT mutations carriers. Together, our findings that MAPT IVS10+16 drives cell-intrinsic dysfunction in microglia that impacts neuronal health has major implications for development of therapeutic strategies.
Collapse
Affiliation(s)
- Abhirami K. Iyer
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University, Amsterdam UMC, The Netherlands
| | | | - Miguel Minaya
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Mariana Acquarone
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | | | - Arun Renganathan
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Shih-Feng You
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Emma P. Danhash
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Anthony Verbeck
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Grant Galasso
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Scott M. Lee
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Jacob Marsh
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Alissa L. Nana
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - William W. Seeley
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Lea T. Grinberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of Sao Paulo
| | | | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University, Amsterdam UMC, The Netherlands
| | - Chihiro Sato
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
- The Tracy Family Stable Isotope Labeling Quantitation Center, Washington University in St Louis, St Louis, MO, USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
8
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. Genome Res 2024; 34:590-605. [PMID: 38599684 PMCID: PMC11146598 DOI: 10.1101/gr.278576.123] [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] [Received: 09/29/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein TAU (current and approved symbol is MAPT) cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human TAU in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas-mediated gene editing to model frontotemporal dementia caused by the TAU P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for Tau P251L display age-dependent neurodegeneration, display metabolic defects, and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies, we performed single-cell RNA sequencing on approximately 130,000 cells from brains of Tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified. Gene expression was also altered in glial cells, suggestive of non-cell-autonomous regulation. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell type-specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy that faithfully recapitulates the genetic context and phenotypic features of the human disease, and use the results of comprehensive single-cell sequencing analysis to outline pathways of neurotoxicity and highlight the potential role of non-cell-autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Rachel A Battaglia
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Anthony Cicalo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Matthew J Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, Massachusetts 02139, USA
| | - Xianjun Dong
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
9
|
Sahayaraj AE, Abdul Vahid A, Dhara A, Babu AT, Vijayan V. Role of G326 in Determining the Aggregation Propensity of R3 Tau Repeat: Insights from Studies on R1R3 Tau Construct. J Phys Chem B 2024; 128:4325-4335. [PMID: 38676652 DOI: 10.1021/acs.jpcb.4c00123] [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: 04/29/2024]
Abstract
The Microtubule-binding repeat region (MTBR) of Tau has been studied extensively due to its pathological implications in neurodegenerative diseases like Alzheimer's disease. The pathological property of MTBR is mainly due to the R3 repeat's high propensity for self-aggregation, highlighting the critical molecular grammar of the repeat. Utilizing the R1R3 construct (WT) and its G326E mutant (EE), we determine the distinct characteristics of various peptide segments that modulate the aggregation propensity of the R3 repeat using NMR spectroscopy. Through time-dependent experiments, we have identified 317KVTSKCGS324 in R3 repeat as the aggregation initiating motif (AIM) due to its role at the initial stages of aggregation. The G326E mutation induces changes in conformation and dynamics at the AIM, thereby effectively abrogating the aggregation propensity of the R1R3 construct. We further corroborate our findings through MD simulations and propose that AIM is a robust site of interest for tauopathy drug design.
Collapse
Affiliation(s)
- Allwin Ebenezer Sahayaraj
- School of Chemistry, IISER-Thiruvananthapuram, Maruthamala PO, Thiruvananthapuram, Kerala 695551, India
| | - Arshad Abdul Vahid
- School of Chemistry, IISER-Thiruvananthapuram, Maruthamala PO, Thiruvananthapuram, Kerala 695551, India
| | - Asmita Dhara
- School of Chemistry, IISER-Thiruvananthapuram, Maruthamala PO, Thiruvananthapuram, Kerala 695551, India
| | - Ann Teres Babu
- School of Chemistry, IISER-Thiruvananthapuram, Maruthamala PO, Thiruvananthapuram, Kerala 695551, India
| | - Vinesh Vijayan
- School of Chemistry, IISER-Thiruvananthapuram, Maruthamala PO, Thiruvananthapuram, Kerala 695551, India
| |
Collapse
|
10
|
Elman JA, Schork NJ, Rangan AV, Alzheimer’s Disease Neuroimaging Initiative. Exploring the genetic heterogeneity of Alzheimer's disease: Evidence for genetic subtypes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.05.02.23289347. [PMID: 37205553 PMCID: PMC10187457 DOI: 10.1101/2023.05.02.23289347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background Alzheimer's disease (AD) exhibits considerable phenotypic heterogeneity, suggesting the potential existence of subtypes. AD is under substantial genetic influence, thus identifying systematic variation in genetic risk may provide insights into disease origins. Objective We investigated genetic heterogeneity in AD risk through a multi-step analysis. Methods We performed principal component analysis (PCA) on AD-associated variants in the UK Biobank (AD cases=2,739, controls=5,478) to assess structured genetic heterogeneity. Subsequently, a biclustering algorithm searched for distinct disease-specific genetic signatures among subsets of cases. Replication tests were conducted using the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset (AD cases=500, controls=470). We categorized a separate set of ADNI individuals with mild cognitive impairment (MCI; n=399) into genetic subtypes and examined cognitive, amyloid, and tau trajectories. Results PCA revealed three distinct clusters ("constellations") driven primarily by different correlation patterns in a region of strong LD surrounding the MAPT locus. Constellations contained a mixture of cases and controls, reflecting disease-relevant but not disease-specific structure. We found two disease-specific biclusters among AD cases. Pathway analysis linked bicluster-associated variants to neuron morphogenesis and outgrowth. Disease-relevant and disease-specific structure replicated in ADNI, and bicluster 2 exhibited increased CSF p-tau and cognitive decline over time. Conclusions This study unveils a hierarchical structure of AD genetic risk. Disease-relevant constellations may represent haplotype structure that does not increase risk directly but may alter the relative importance of other genetic risk factors. Biclusters may represent distinct AD genetic subtypes. This structure is replicable and relates to differential pathological accumulation and cognitive decline over time.
Collapse
Affiliation(s)
- Jeremy A. Elman
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Nicholas J. Schork
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- The Translational Genomics Research Institute, Quantitative Medicine and Systems Biology, Phoenix, AZ, USA
| | - Aaditya V. Rangan
- Department of Mathematics, New York University, New York, New York, USA
| | | |
Collapse
|
11
|
Tong CS, Su M, Sun H, Chua XL, Xiong D, Guo S, Raj R, Ong NWP, Lee AG, Miao Y, Wu M. Collective dynamics of actin and microtubule and its crosstalk mediated by FHDC1. Front Cell Dev Biol 2024; 11:1261117. [PMID: 38567385 PMCID: PMC10985548 DOI: 10.3389/fcell.2023.1261117] [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: 07/18/2023] [Accepted: 12/19/2023] [Indexed: 04/04/2024] Open
Abstract
The coordination between actin and microtubule network is crucial, yet this remains a challenging problem to dissect and our understanding of the underlying mechanisms remains limited. In this study, we used travelling waves in the cell cortex to characterize the collective dynamics of cytoskeletal networks. Our findings show that Cdc42 and F-BAR-dependent actin waves in mast cells are mainly driven by formin-mediated actin polymerization, with the microtubule-binding formin FH2 domain-containing protein 1 (FHDC1) as an early regulator. Knocking down FHDC1 inhibits actin wave formation, and this inhibition require FHDC1's interaction with both microtubule and actin. The phase of microtubule depolymerization coincides with the nucleation of actin waves and microtubule stabilization inhibit actin waves, leading us to propose that microtubule shrinking and the concurrent release of FHDC1 locally regulate actin nucleation. Lastly, we show that FHDC1 is crucial for multiple cellular processes such as cell division and migration. Our data provided molecular insights into the nucleation mechanisms of actin waves and uncover an antagonistic interplay between microtubule and actin polymerization in their collective dynamics.
Collapse
Affiliation(s)
- Chee San Tong
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
| | - Maohan Su
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - He Sun
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xiang Le Chua
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
| | - Ding Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Su Guo
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
| | - Ravin Raj
- Special Programme in Science, National University of Singapore, Singapore, Singapore
| | - Nicole Wen Pei Ong
- Special Programme in Science, National University of Singapore, Singapore, Singapore
| | - Ann Gie Lee
- Special Programme in Science, National University of Singapore, Singapore, Singapore
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Min Wu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biological Sciences, Centre for Bioimaging Sciences, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578624. [PMID: 38352559 PMCID: PMC10862891 DOI: 10.1101/2024.02.02.578624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein tau cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human tau in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease, but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas mediated gene editing to model frontotemporal dementia caused by the tau P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for tau P251L display age-dependent neurodegeneration, metabolic defects and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies we performed single-cell RNA sequencing on approximately 130,000 cells from brains of tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified and non-cell autonomously in glial cells. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell-type specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy, which faithfully recapitulates the genetic context and phenotypic features of the human disease and use the results of comprehensive single cell sequencing analysis to outline pathways of neurotoxicity and highlight the role of non-cell autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Rachel A. Battaglia
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Anthony Cicalo
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Genomics and Bioinformatics Hub, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Matthew J. Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA 02139
| | - Xianjun Dong
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Genomics and Bioinformatics Hub, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Mel B. Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| |
Collapse
|
13
|
Huang H, Majumder T, Khot B, Suriyaarachchi H, Yang T, Shao Q, Tirukovalluru S, Liu G. The role of microtubule-associated protein tau in netrin-1 attractive signaling. J Cell Sci 2024; 137:jcs261244. [PMID: 38197773 PMCID: PMC10906489 DOI: 10.1242/jcs.261244] [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: 04/12/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024] Open
Abstract
Direct binding of netrin receptors with dynamic microtubules (MTs) in the neuronal growth cone plays an important role in netrin-mediated axon guidance. However, how netrin-1 (NTN1) regulates MT dynamics in axon turning remains a major unanswered question. Here, we show that the coupling of netrin-1 receptor DCC with tau (MAPT)-regulated MTs is involved in netrin-1-promoted axon attraction. Tau directly interacts with DCC and partially overlaps with DCC in the growth cone of primary neurons. Netrin-1 induces this interaction and the colocalization of DCC and tau in the growth cone. The netrin-1-induced interaction of tau with DCC relies on MT dynamics and TUBB3, a highly dynamic β-tubulin isotype in developing neurons. Netrin-1 increased cosedimentation of DCC with tau and TUBB3 in MTs, and knockdown of either tau or TUBB3 mutually blocked this effect. Downregulation of endogenous tau levels by tau shRNAs inhibited netrin-1-induced axon outgrowth, branching and commissural axon attraction in vitro, and led to defects in spinal commissural axon projection in vivo. These findings suggest that tau is a key MT-associated protein coupling DCC with MT dynamics in netrin-1-promoted axon attraction.
Collapse
Affiliation(s)
- Huai Huang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Harindi Suriyaarachchi
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Shraddha Tirukovalluru
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| |
Collapse
|
14
|
Elman JA, Schork NJ, Rangan AV. Exploring the Genetic Heterogeneity of Alzheimer's Disease: Evidence for Genetic Subtypes. J Alzheimers Dis 2024; 100:1209-1226. [PMID: 38995775 PMCID: PMC11636402 DOI: 10.3233/jad-231252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Background Alzheimer's disease (AD) exhibits considerable phenotypic heterogeneity, suggesting the potential existence of subtypes. AD is under substantial genetic influence, thus identifying systematic variation in genetic risk may provide insights into disease origins. Objective We investigated genetic heterogeneity in AD risk through a multi-step analysis. Methods We performed principal component analysis (PCA) on AD-associated variants in the UK Biobank (AD cases = 2,739, controls = 5,478) to assess structured genetic heterogeneity. Subsequently, a biclustering algorithm searched for distinct disease-specific genetic signatures among subsets of cases. Replication tests were conducted using the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset (AD cases = 500, controls = 470). We categorized a separate set of ADNI individuals with mild cognitive impairment (MCI; n = 399) into genetic subtypes and examined cognitive, amyloid, and tau trajectories. Results PCA revealed three distinct clusters ("constellations") driven primarily by different correlation patterns in a region of strong LD surrounding the MAPT locus. Constellations contained a mixture of cases and controls, reflecting disease-relevant but not disease-specific structure. We found two disease-specific biclusters among AD cases. Pathway analysis linked bicluster-associated variants to neuron morphogenesis and outgrowth. Disease-relevant and disease-specific structure replicated in ADNI, and bicluster 2 exhibited increased cerebrospinal fluid p-tau and cognitive decline over time. Conclusions This study unveils a hierarchical structure of AD genetic risk. Disease-relevant constellations may represent haplotype structure that does not increase risk directly but may alter the relative importance of other genetic risk factors. Biclusters may represent distinct AD genetic subtypes. This structure is replicable and relates to differential pathological accumulation and cognitive decline over time.
Collapse
Affiliation(s)
- Jeremy A. Elman
- Department of Psychiatry, University of California San
Diego, La Jolla, CA, USA
- Center for Behavior Genetics of Aging, University of
California San Diego, La Jolla, CA, USA
| | - Nicholas J. Schork
- Department of Psychiatry, University of California San
Diego, La Jolla, CA, USA
- The Translational Genomics Research Institute, Quantitative
Medicine and Systems Biology, Phoenix, AZ, USA
| | - Aaditya V. Rangan
- Department of Mathematics, New York University, New York,
New York, USA
| |
Collapse
|
15
|
Gélin M, Schaeffer A, Gaillard J, Guérin C, Vianay B, Orhant-Prioux M, Braun M, Leterrier C, Blanchoin L, Théry M. Microtubules under mechanical pressure can breach dense actin networks. J Cell Sci 2023; 136:jcs261667. [PMID: 37870087 DOI: 10.1242/jcs.261667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The crosstalk between the actin network and microtubules is essential for cell polarity. It orchestrates microtubule organization within the cell, driven by the asymmetry of actin architecture along the cell periphery. The physical intertwining of these networks regulates spatial organization and force distribution in the microtubule network. Although their biochemical interactions are becoming clearer, the mechanical aspects remain less understood. To explore this mechanical interplay, we developed an in vitro reconstitution assay to investigate how dynamic microtubules interact with various actin filament structures. Our findings revealed that microtubules can align and move along linear actin filament bundles through polymerization force. However, they are unable to pass through when encountering dense branched actin meshworks, similar to those present in the lamellipodium along the periphery of the cell. Interestingly, immobilizing microtubules through crosslinking with actin or other means allow the buildup of pressure, enabling them to breach these dense actin barriers. This mechanism offers insights into microtubule progression towards the cell periphery, with them overcoming obstacles within the denser parts of the actin network and ultimately contributing to cell polarity establishment.
Collapse
Affiliation(s)
- Matthieu Gélin
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Alexandre Schaeffer
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Jérémie Gaillard
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Christophe Guérin
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Benoit Vianay
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Magali Orhant-Prioux
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 25250 Vestec, Prague West, Czech Republic
| | - Christophe Leterrier
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto, 13385, Marseille, France
| | - Laurent Blanchoin
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Manuel Théry
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| |
Collapse
|
16
|
Chakraborty R, Darido C, Chien A, Tay A, Vickery K, Hu H, Liu F, Ranganathan S. Preclinical 3D-model supports an invisibility cloak for adenoid cystic carcinoma. Sci Rep 2023; 13:17033. [PMID: 37813936 PMCID: PMC10562364 DOI: 10.1038/s41598-023-44329-7] [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: 02/15/2023] [Accepted: 10/06/2023] [Indexed: 10/11/2023] Open
Abstract
The tumour-cell based initiation of immune evasion project evaluated the role of Gipie in adenoid cystic carcinoma (ACC) and mucoepidermoid carcinoma (A-253), from ninety-six 3D-ACC and A-253-immune co-culture models using natural killer cells (NK), and Jurkat cells (JK). Abnormal ACC morphology was observed in 3D-ACC immune co-culture models. Gipie-silencing conferred a "lymphoblast-like" morphology to ACC cells, a six-fold increase in apoptotic cells (compared to unaltered ACC cells, P ≤ 0.0001), a two-fold decrease in T regulatory cells (FoxP3+/IL-2Rα+/CD25+) (P ≤ 0.0001), and a three-fold increase in activated NK cells (NKp30+/IFN-γ+) (P ≤ 0.0001) with significantly higher release of granzyme (P ≤ 0.001) and perforin (P ≤ 0.0001).
Collapse
Affiliation(s)
- Rajdeep Chakraborty
- Applied Biosciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia.
| | - Charbel Darido
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Arthur Chien
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Aidan Tay
- Applied Biosciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Karen Vickery
- Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Honghua Hu
- Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Fei Liu
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Shoba Ranganathan
- Applied Biosciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
17
|
Frost B. Alzheimer's disease and related tauopathies: disorders of disrupted neuronal identity. Trends Neurosci 2023; 46:797-813. [PMID: 37591720 PMCID: PMC10528597 DOI: 10.1016/j.tins.2023.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/20/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023]
Abstract
Postmitotic neurons require persistently active controls to maintain terminal differentiation. Unlike dividing cells, aberrant cell cycle activation in mature neurons causes apoptosis rather than transformation. In Alzheimer's disease (AD) and related tauopathies, evidence suggests that pathogenic forms of tau drive neurodegeneration via neuronal cell cycle re-entry. Multiple interconnected mechanisms linking tau to cell cycle activation have been identified, including, but not limited to, tau-induced overstabilization of the actin cytoskeleton, consequent changes to nuclear architecture, and disruption of heterochromatin-mediated gene silencing. Cancer- and development-associated pathways are upregulated in human and cellular models of tauopathy, and many tau-induced cellular phenotypes are also present in various cancers and progenitor/stem cells. In this review, I delve into mechanistic parallels between tauopathies, cancer, and development, and highlight the role of tau in cancer and in the developing brain. Based on these studies, I put forth a model by which pathogenic forms of tau disrupt the program that maintains terminal neuronal differentiation, driving cell cycle re-entry and consequent neuronal death. This framework presents tauopathies as conditions involving the profound toxic disruption of neuronal identity.
Collapse
Affiliation(s)
- Bess Frost
- Sam & Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
18
|
DeGiosio RA, Needham PG, Andrews OA, Tristan H, Grubisha MJ, Brodsky JL, Camacho C, Sweet RA. Differential regulation of MAP2 by phosphorylation events in proline-rich versus C-terminal domains. FASEB J 2023; 37:e23194. [PMID: 37702880 PMCID: PMC10539048 DOI: 10.1096/fj.202300486r] [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: 03/13/2023] [Revised: 07/31/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023]
Abstract
MAP2 is a critical cytoskeletal regulator in neurons. The phosphorylation of MAP2 (MAP2-P) is well known to regulate core functions of MAP2, including microtubule (MT)/actin binding and facilitation of tubulin polymerization. However, site-specific studies of MAP2-P function in regions outside of the MT-binding domain (MTBD) are lacking. We previously identified a set of MAP2 phosphopeptides which are differentially expressed and predominantly increased in the cortex of individuals with schizophrenia relative to nonpsychiatric comparison subjects. The phosphopeptides originated not from the MTBD, but from the flanking proline-rich and C-terminal domains of MAP2. We sought to understand the contribution of MAP2-P at these sites on MAP2 function. To this end, we isolated a series of phosphomimetic MAP2C constructs and subjected them to cell-free tubulin polymerization, MT-binding, actin-binding, and actin polymerization assays. A subset of MAP2-P events significantly impaired these functions, with the two domains displaying different patterns of MAP2 regulation: proline-rich domain mutants T293E and T300E impaired MT assembly and actin-binding affinity but did not affect MT-binding, while C-terminal domain mutants S426E and S439D impaired all three functions. S443D also impaired MT assembly with minimal effects on MT- or actin-binding. Using heterologous cells, we also found that S426E but not T293E had a lower capability for process formation than the wild-type protein. These findings demonstrate the functional utility of MAP2-P in the proline-rich and C-terminal domains and point to distinct, domain-dependent regulations of MAP2 function, which can go on to affect cellular morphology.
Collapse
Affiliation(s)
- R A DeGiosio
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - P G Needham
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - O A Andrews
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - H Tristan
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - M J Grubisha
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - J L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - C Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - R A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Duarte ML, Wang M, Gomes I, Liu C, Sharma A, Fakira AK, Gupta A, Mack SM, Zhang B, Devi LA. Multiomics Analyses Identify Proline Endopeptidase-Like Protein As a Key Regulator of Protein Trafficking, a Pathway Underlying Alzheimer's Disease Pathogenesis. Mol Pharmacol 2023; 104:1-16. [PMID: 37147110 PMCID: PMC10289242 DOI: 10.1124/molpharm.122.000641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/22/2023] [Accepted: 04/12/2023] [Indexed: 05/07/2023] Open
Abstract
Current treatments for Alzheimer's disease (AD) help reduce symptoms for a limited time but do not treat the underlying pathology. To identify potential therapeutic targets for AD, an integrative network analysis was previously carried out using 364 human postmortem control, mild cognitive impairment, and AD brains. This analysis identified proline endopeptidase-like protein (PREPL), an understudied protein, as a downregulated protein in late-onset AD patients. In this study we investigate the role of PREPL. Analyses of data from human postmortem samples and PREPL knockdown (KD) cells suggest that PREPL expression modulates pathways associated with protein trafficking, synaptic activities, and lipid metabolism. Furthermore, PREPL KD impairs cell proliferation and modulates the structure of vesicles, levels of neuropeptide-processing enzymes, and secretion of neuropeptides. In addition, decrease in PREPL levels leads to changes in the levels of a number of synaptic proteins as well as changes in the levels of secreted amyloid beta (Aβ) 42 peptide and Tau phosphorylation. Finally, we report that local decrease in PREPL levels in mouse hippocampus attenuates long-term potentiation, suggesting a role in synaptic plasticity. Together, our results indicate that PREPL affects neuronal function by modulating protein trafficking and synaptic function, an important mechanism of AD pathogenesis. SIGNIFICANCE STATEMENT: Integrative network analysis reveals proline endopeptidase-like protein (PREPL) to be downregulated in human sporadic late-onset Alzheimer's disease brains. Down regulation of PREPL leads to increases in amyloid beta secretion, Tau phosphorylation, and decreases in protein trafficking and long-term potentiation.
Collapse
Affiliation(s)
- Mariana Lemos Duarte
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Minghui Wang
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ivone Gomes
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Chenge Liu
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ali Sharma
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amanda K Fakira
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Achla Gupta
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Seshat M Mack
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bin Zhang
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lakshmi A Devi
- Department of Pharmacological Sciences (M.L.D., I.G., C.L., A.S., A.K.F., A.G., S.M.M., L.A.D.), Department of Genetics and Genomics (M.W., B.Z.), and Department of Neurology (M.L.D.), Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
20
|
Pinzi L, Bisi N, Sorbi C, Franchini S, Tonali N, Rastelli G. Insights into the Structural Conformations of the Tau Protein in Different Aggregation Status. Molecules 2023; 28:4544. [PMID: 37299020 PMCID: PMC10254443 DOI: 10.3390/molecules28114544] [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: 03/29/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Tau is a protein characterized by large structural portions displaying extended conformational changes. Unfortunately, the accumulation of this protein into toxic aggregates in neuronal cells leads to a number of severe pathologies, collectively named tauopathies. In the last decade, significant research advancements were achieved, including a better understanding of Tau structures and their implication in different tauopathies. Interestingly, Tau is characterized by a high structural variability depending on the type of disease, the crystallization conditions, and the formation of pathologic aggregates obtained from in vitro versus ex vivo samples. In this review, we reported an up-to-date and comprehensive overview of Tau structures reported in the Protein Data Bank, with a special focus on discussing the connections between structural features, different tauopathies, different crystallization conditions, and the use of in vitro or ex vivo samples. The information reported in this article highlights very interesting links between all these aspects, which we believe may be of particular relevance for a more informed structure-based design of compounds able to modulate Tau aggregation.
Collapse
Affiliation(s)
- Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (C.S.); (S.F.)
| | - Nicolò Bisi
- Centre National de la Recherche Scientifique (CNRS), Université de Paris-Saclay, BioCIS, Bat. Henri Moissan, 17 Av. des Sciences, 91400 Orsay, France; (N.B.); (N.T.)
| | - Claudia Sorbi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (C.S.); (S.F.)
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (C.S.); (S.F.)
| | - Nicolò Tonali
- Centre National de la Recherche Scientifique (CNRS), Université de Paris-Saclay, BioCIS, Bat. Henri Moissan, 17 Av. des Sciences, 91400 Orsay, France; (N.B.); (N.T.)
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (C.S.); (S.F.)
| |
Collapse
|
21
|
Inaba H, Oikawa K, Ishikawa K, Kodama Y, Matsuura K, Numata K. Binding of Tau-derived peptide-fused GFP to plant microtubules in Arabidopsis thaliana. PLoS One 2023; 18:e0286421. [PMID: 37267323 DOI: 10.1371/journal.pone.0286421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/16/2023] [Indexed: 06/04/2023] Open
Abstract
Studies on how exogenous molecules modulate properties of plant microtubules, such as their stability, structure, and dynamics, are important for understanding and modulating microtubule functions in plants. We have developed a Tau-derived peptide (TP) that binds to microtubules and modulates their properties by binding of TP-conjugated molecules in vitro. However, there was no investigation of TPs on microtubules in planta. Here, we generated transgenic Arabidopsis thaliana plants stably expressing TP-fused superfolder GFP (sfGFP-TP) and explored the binding properties and effects of sfGFP-TP on plant microtubules. Our results indicate that the expressed sfGFP-TP binds to the plant microtubules without inhibiting plant growth. A transgenic line strongly expressing sfGFP-TP produced thick fibrous structures that were stable under conditions where microtubules normally depolymerize. This study generates a new tool for analyzing and modulating plant microtubules.
Collapse
Affiliation(s)
- Hiroshi Inaba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan
| | - Kazusato Oikawa
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kazuya Ishikawa
- Center for Bioscience Research and Education, Utsunomiya University, Tochigi, Japan
| | - Yutaka Kodama
- Center for Bioscience Research and Education, Utsunomiya University, Tochigi, Japan
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan
| | - Keiji Numata
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| |
Collapse
|
22
|
Zhang H, Lu F, Liu P, Qiu Z, Li J, Wang X, Xu H, Zhao Y, Li X, Wang H, Lu D, Qi R. A direct interaction between RhoGDIα/Tau alleviates hyperphosphorylation of Tau in Alzheimer's disease and vascular dementia. J Neuroimmune Pharmacol 2023; 18:58-71. [PMID: 35080740 DOI: 10.1007/s11481-021-10049-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023]
Abstract
RhoGDIα is an inhibitor of RhoGDP dissociation that involves in Aβ metabolism and NFTs production in Alzheimer's disease (AD) by regulating of RhoGTP enzyme activity. Our previous research revealed that RhoGDIα, as the target of Polygala saponin (Sen), might alleviate apoptosis of the nerve cells caused by hypoxia/reoxygenation (H/R). To further clarify the role of RhoGDIα in the generation of NFTs, we explored the relationship between RhoGDIα and Tau. We found out that RhoGDIα and Tau can bind with each other and interact by using coimmunoprecipitation (Co-IP) and GST pulldown methods in vitro. This RhoGDIα-Tau partnership was further verified by using immunofluorescence colocalization and fluorescence resonance energy transfer (FRET) approaches in PC12 cells. Using the RNA interference (RNAi) technique, we found that the RhoGDIα may be involved in an upstream signaling pathway for Tau. Subsequently, in Aβ25-35- and H/R-induced PC12 cells, forced expression of RhoGDIα via cDNA plasmid transfection was found to reduce the hyperphosphorylation of Tau, augment the expression of bcl-2 protein, and inhibit the expression of Bax protein (reducing the Bax/bcl-2 ratio) and the activity of caspase-3. In mouse AD and VaD models, forced expression of RhoGDIα via injection of a viral vector (pAAV-EGFP-RhoGDIα) into the lateral ventricle of the brain alleviated the pathological symptoms of AD and VaD. Finally, GST pulldown confirmed that the binding sites on RhoGDIα for Tau were located in the range of the ΔC33 fragment (aa 1-33). These results indicate that RhoGDIα is involved in the phosphorylation of Tau and apoptosis in AD and VaD. Overexpression of RhoGDIα can inhibit the generation of NFTs and delay the progress of these two types of dementia.
Collapse
Affiliation(s)
- Heping Zhang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Fan Lu
- Department of Emergency, First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Panhong Liu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- Department of Pathology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Zhaohui Qiu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-Sen University, ShenZhen, 518033, China
| | - Jianling Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- Department of Anesthesiology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xiaotong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Hui Xu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Yandong Zhao
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Xuemin Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Anhui, 230031, China
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Daxiang Lu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Renbin Qi
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
23
|
Nadel CM, Thwin AC, Callahan M, Lee K, Connelly E, Craik CS, Southworth DR, Gestwicki JE. The E3 Ubiquitin Ligase, CHIP/STUB1, Inhibits Aggregation of Phosphorylated Proteoforms of Microtubule-associated Protein Tau (MAPT). J Mol Biol 2023; 435:168026. [PMID: 37330289 PMCID: PMC10491737 DOI: 10.1016/j.jmb.2023.168026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 06/19/2023]
Abstract
Hyper-phosphorylated tau accumulates as insoluble fibrils in Alzheimer's disease (AD) and related dementias. The strong correlation between phosphorylated tau and disease has led to an interest in understanding how cellular factors discriminate it from normal tau. Here, we screen a panel of chaperones containing tetratricopeptide repeat (TPR) domains to identify those that might selectively interact with phosphorylated tau. We find that the E3 ubiquitin ligase, CHIP/STUB1, binds 10-fold more strongly to phosphorylated tau than unmodified tau. The presence of even sub-stoichiometric concentrations of CHIP strongly suppresses aggregation and seeding of phosphorylated tau. We also find that CHIP promotes rapid ubiquitination of phosphorylated tau, but not unmodified tau, in vitro. Binding to phosphorylated tau requires CHIP's TPR domain, but the binding mode is partially distinct from the canonical one. In cells, CHIP restricts seeding by phosphorylated tau, suggesting that it could be an important barrier in cell-to-cell spreading. Together, these findings show that CHIP recognizes a phosphorylation-dependent degron on tau, establishing a pathway for regulating the solubility and turnover of this pathological proteoform.
Collapse
Affiliation(s)
- Cory M Nadel
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Aye C Thwin
- Biochemistry & Biophysics and the University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Matthew Callahan
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Kanghyun Lee
- Biochemistry & Biophysics and the University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Emily Connelly
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA
| | - Charles S Craik
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA
| | - Daniel R Southworth
- Biochemistry & Biophysics and the University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA.
| | - Jason E Gestwicki
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA.
| |
Collapse
|
24
|
Beckmann A, Ramirez P, Gamez M, Gonzalez E, De Mange J, Bieniek KF, Ray WJ, Frost B. Moesin is an effector of tau-induced actin overstabilization, cell cycle activation, and neurotoxicity in Alzheimer's disease. iScience 2023; 26:106152. [PMID: 36879821 PMCID: PMC9984563 DOI: 10.1016/j.isci.2023.106152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/01/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
In Alzheimer's disease, neurons acquire phenotypes that are also present in various cancers, including aberrant activation of the cell cycle. Unlike cancer, cell cycle activation in post-mitotic neurons is sufficient to induce cell death. Multiple lines of evidence suggest that abortive cell cycle activation is a consequence of pathogenic forms of tau, a protein that drives neurodegeneration in Alzheimer's disease and related "tauopathies." Here we combine network analyses of human Alzheimer's disease and mouse models of Alzheimer's disease and primary tauopathy with studies in Drosophila to discover that pathogenic forms of tau drive cell cycle activation by disrupting a cellular program involved in cancer and the epithelial-mesenchymal transition (EMT). Moesin, an EMT driver, is elevated in cells harboring disease-associated phosphotau, over-stabilized actin, and ectopic cell cycle activation. We further find that genetic manipulation of Moesin mediates tau-induced neurodegeneration. Taken together, our study identifies novel parallels between tauopathy and cancer.
Collapse
Affiliation(s)
- Adrian Beckmann
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Paulino Ramirez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Maria Gamez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Elias Gonzalez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jasmine De Mange
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Kevin F. Bieniek
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - William J. Ray
- The Neurodegeneration Consortium, Therapeutics Discovery Division, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bess Frost
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
25
|
Biomolecular condensation involving the cytoskeleton. Brain Res Bull 2023; 194:105-117. [PMID: 36690162 DOI: 10.1016/j.brainresbull.2023.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Biomolecular condensation of proteins contributes to the organization of the cytoplasm and nucleoplasm. A number of condensation processes appear to be directly involved in regulating the structure, function and dynamics of the cytoskeleton. Liquid-liquid phase separation of cytoskeleton proteins, together with polymerization modulators, promotes cytoskeletal fiber nucleation and branching. Furthermore, the attachment of protein condensates to the cytoskeleton can contribute to cytoskeleton stability and organization, regulate transport, create patterns of functional reaction containers, and connect the cytoskeleton with membranes. Surface-bound condensates can exert and buffer mechanical forces that give stability and flexibility to the cytoskeleton, thus, may play a large role in cell biology. In this review, we introduce the concept and role of cellular biomolecular condensation, explain its special function on cytoskeletal fiber surfaces, and point out potential definition and experimental caveats. We review the current literature on protein condensation processes related to the actin, tubulin, and intermediate filament cytoskeleton, and discuss some of them in the context of neurobiology. In summary, we provide an overview about biomolecular condensation in relation to cytoskeleton structure and function, which offers a base for the exploration and interpretation of cytoskeletal condensates in neurobiology.
Collapse
|
26
|
Tabeshmehr P, Eftekharpour E. Tau; One Protein, So Many Diseases. BIOLOGY 2023; 12:244. [PMID: 36829521 PMCID: PMC9953016 DOI: 10.3390/biology12020244] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023]
Abstract
Tau, a member of the microtubule-associated proteins, is a known component of the neuronal cytoskeleton; however, in the brain tissue, it is involved in other vital functions beyond maintaining the cellular architecture. The pathologic tau forms aggregates inside the neurons and ultimately forms the neurofibrillary tangles. Intracellular and extracellular accumulation of different tau isoforms, including dimers, oligomers, paired helical filaments and tangles, lead to a highly heterogenous group of diseases named "Tauopathies". About twenty-six different types of tauopathy diseases have been identified that have different clinical phenotypes or pathophysiological characteristics. Although all these diseases are identified by tau aggregation, they are distinguishable based on the specific tau isoforms, the affected cell types and the brain regions. The neuropathological and phenotypical heterogeneity of these diseases impose significant challenges for discovering new diagnostic and therapeutic strategies. Here, we review the recent literature on tau protein and the pathophysiological mechanisms of tauopathies. This article mainly focuses on physiologic and pathologic tau and aims to summarize the upstream and downstream events and discuss the current diagnostic approaches and therapeutic strategies.
Collapse
Affiliation(s)
| | - Eftekhar Eftekharpour
- Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
27
|
Sohn C, Ma J, Ray WJ, Frost B. Pathogenic tau decreases nuclear tension in cultured neurons. FRONTIERS IN AGING 2023; 4:1058968. [PMID: 36756194 PMCID: PMC9901542 DOI: 10.3389/fragi.2023.1058968] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
Neurodegenerative tauopathies, including Alzheimer's disease, are pathologically defined by the presence of aggregated forms of tau protein in brains of affected individuals. Previous studies report that the negative effects of pathogenic tau on the actin cytoskeleton and microtubules cause a toxic destabilization of the lamin nucleoskeleton and formation of nuclear invaginations and blebs. Based on the known function of the nucleus as a mechanosensor, as well as the high incidence of nuclear pleomorphism in human Alzheimer's disease and related tauopathies, we investigated the effects of pathogenic tau on nuclear tension. We first find that tau-dependent nuclear envelope invagination and relocalization of LInker of Nucleoskeleton and Cytoskeleton (LINC) complex components are conserved in a newly-developed neuroblastoma cell line that features doxycycline-inducible expression of a tau mutant associated with autosomal dominant frontotemporal dementia. We next determine that a Förster resonance energy transfer (FRET)-based sensor of nuclear tension responds to cytoskeletal stabilization and destabilization when expressed in neuroblastoma cells. Using this nuclear tension sensor, we find that induced expression of pathogenic tau is sufficient to decrease nuclear tension. This work provides the initial proof-of-concept evidence that pathogenic forms of tau alter nuclear tension, paving the way for the future study of altered nuclear mechanosensing in the context of tau-mediated neurodegenerative disorders.
Collapse
Affiliation(s)
- Claira Sohn
- Department of Cell Systems and Anatomy, Barshop Institute for Longevity and Aging Studies, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jiacheng Ma
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - William J. Ray
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bess Frost
- Department of Cell Systems and Anatomy, Barshop Institute for Longevity and Aging Studies, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
28
|
Septins mediate a microtubule-actin crosstalk that enables actin growth on microtubules. Proc Natl Acad Sci U S A 2022; 119:e2202803119. [PMID: 36475946 PMCID: PMC9897426 DOI: 10.1073/pnas.2202803119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cellular morphogenesis and processes such as cell division and migration require the coordination of the microtubule and actin cytoskeletons. Microtubule-actin crosstalk is poorly understood and largely regarded as the capture and regulation of microtubules by actin. Septins are filamentous guanosine-5'-triphosphate (GTP) binding proteins, which comprise the fourth component of the cytoskeleton along microtubules, actin, and intermediate filaments. Here, we report that septins mediate microtubule-actin crosstalk by coupling actin polymerization to microtubule lattices. Superresolution and platinum replica electron microscopy (PREM) show that septins localize to overlapping microtubules and actin filaments in the growth cones of neurons and non-neuronal cells. We demonstrate that recombinant septin complexes directly crosslink microtubules and actin filaments into hybrid bundles. In vitro reconstitution assays reveal that microtubule-bound septins capture and align stable actin filaments with microtubules. Strikingly, septins enable the capture and polymerization of growing actin filaments on microtubule lattices. In neuronal growth cones, septins are required for the maintenance of the peripheral actin network that fans out from microtubules. These findings show that septins directly mediate microtubule interactions with actin filaments, and reveal a mechanism of microtubule-templated actin growth with broader significance for the self-organization of the cytoskeleton and cellular morphogenesis.
Collapse
|
29
|
Chen K, Li W, Xu K. Super-multiplexing excitation spectral microscopy with multiple fluorescence bands. BIOMEDICAL OPTICS EXPRESS 2022; 13:6048-6060. [PMID: 36733753 PMCID: PMC9872899 DOI: 10.1364/boe.473241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 06/18/2023]
Abstract
Fluorescence microscopy, with high molecular specificity and selectivity, is a valuable tool for studying complex biological systems and processes. However, the ability to distinguish a large number of distinct subcellular structures in a single sample is impeded by the broad spectra of molecular fluorescence. We have recently shown that excitation spectral microscopy provides a powerful means to unmix up to six fluorophores in a single fluorescence band. Here, by working with multiple fluorescence bands, we extend this approach to the simultaneous imaging of up to ten targets, with the potential for further expansions. By covering the excitation/emission bandwidth across the full visible range, an ultra-broad 24-wavelength excitation scheme is established through frame-synchronized scanning of the excitation wavelength from a white lamp via an acousto-optic tunable filter (AOTF), so that full-frame excitation-spectral images are obtained every 24 camera frames, offering superior spectral information and multiplexing capability. With numerical simulations, we validate the concurrent imaging of 10 fluorophores spanning the visible range to achieve exceptionally low (∼0.5%) crosstalks. For cell imaging experiments, we demonstrate unambiguous identification of up to eight different intracellular structures labeled by common fluorophores of substantial spectral overlap with minimal color crosstalks. We thus showcase an easy-to-implement, cost-effective microscopy system for visualizing complex cellular components with more colors and lower crosstalks.
Collapse
Affiliation(s)
- Kun Chen
- Department of Chemistry & California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Wan Li
- Department of Chemistry & California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ke Xu
- Department of Chemistry & California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
30
|
Cazzaro S, Fang C, Khan H, Witas R, Kee TR, Woo JAA, Kang DE. Slingshot homolog-1 mediates the secretion of small extracellular vesicles containing misfolded proteins by regulating autophagy cargo receptors and actin dynamics. Front Aging Neurosci 2022; 14:933979. [PMID: 36092812 PMCID: PMC9452914 DOI: 10.3389/fnagi.2022.933979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing evidence indicates that the accumulation misfolded proteins in Alzheimer's disease (AD) arises from clearance defects in the autophagy-lysosome pathway. Misfolded proteins such as Aβ and tau are secreted in small extracellular vesicles (i.e., exosomes) and are propagated from cell to cell in part through secreted small extracellular vesicles (sEVs). Recent studies suggest that autophagic activity and exosome secretion are coregulated events, and multiple autophagy-related proteins are found in sEVs, including the cargo receptors Sqstm1/p62 and optineurin. However, whether and how autophagy cargo receptors per se regulate the secretion of sEVs is unknown. Moreover, despite the prominent role of actin dynamics in secretory vesicle release, its role in EV secretion is unknown. In this study, we leveraged the dual axes of Slingshot Homolog-1 (SSH1), which inhibits Sqstm1/p62-mediated autophagy and activates cofilin-mediated actin dynamics, to study the regulation of sEV secretion. Here we show that cargo receptors Sqstm1/p62 and optineurin inhibit sEV secretion, an activity that requires their ability to bind ubiquitinated cargo. Conversely, SSH1 increases sEV secretion by dephosphorylating Sqstm1/p62 at pSer403, the phospho-residue that allows Sqstm1/p62 to bind ubiquitinated cargo. In addition, increasing actin dynamics through the SSH1-cofilin activation pathway also increases sEV secretion, which is mimicked by latrunculin B treatment. Finally, Aβ42 oligomers and mutant tau increase sEV secretion and are physically associated with secreted sEVs. These findings suggest that increasing cargo receptor engagement with autophagic cargo and reducing actin dynamics (i.e., SSH1 inhibition) represents an attractive strategy to promote misfolded protein degradation while reducing sEV-mediated cell to cell spread of pathology.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Cenxiao Fang
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Hirah Khan
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Richard Witas
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Teresa R. Kee
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Jung-A. A. Woo
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - David E. Kang
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, United States
| |
Collapse
|
31
|
Goel P, Chakrabarti S, Goel K, Bhutani K, Chopra T, Bali S. Neuronal cell death mechanisms in Alzheimer's disease: An insight. Front Mol Neurosci 2022; 15:937133. [PMID: 36090249 PMCID: PMC9454331 DOI: 10.3389/fnmol.2022.937133] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regulated cell death (RCD) is an ordered and tightly orchestrated set of changes/signaling events in both gene expression and protein activity and is responsible for normal development as well as maintenance of tissue homeostasis. Aberrant activation of this pathway results in cell death by various mechanisms including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. Such pathological changes in neurons alone or in combination have been observed in the pathogenesis of various neurodegenerative diseases including Alzheimer's disease (AD). Pathological hallmarks of AD focus primarily on the accumulation of two main protein markers: amyloid β peptides and abnormally phosphorylated tau proteins. These protein aggregates result in the formation of A-β plaques and neuro-fibrillary tangles (NFTs) and induce neuroinflammation and neurodegeneration over years to decades leading to a multitude of cognitive and behavioral deficits. Autopsy findings of AD reveal massive neuronal death manifested in the form of cortical volume shrinkage, reduction in sizes of gyri to up to 50% and an increase in the sizes of sulci. Multiple forms of cell death have been recorded in neurons from different studies conducted so far. However, understanding the mechanism/s of neuronal cell death in AD patients remains a mystery as the trigger that results in aberrant activation of RCD is unknown and because of the limited availability of dying neurons. This review attempts to elucidate the process of Regulated cell death, how it gets unregulated in response to different intra and extracellular stressors, various forms of unregulated cell death, their interplay and their role in pathogenesis of Alzheimer's Disease in both human and experimental models of AD. Further we plan to explore the correlation of both amyloid-beta and Tau with neuronal loss as seen in AD.
Collapse
Affiliation(s)
- Parul Goel
- Department of Biochemistry, Shri Atal Bihari Vajpayee Government Medical College Chhainsa, Faridabad, India
| | - Sasanka Chakrabarti
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Kapil Goel
- Department of Community Medicine and School of Public Health, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Karanpreet Bhutani
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Tanya Chopra
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Sharadendu Bali
- Department of Surgery, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| |
Collapse
|
32
|
Fourest-Lieuvin A, Vinit A, Blot B, Perrot A, Denarier E, Saudou F, Arnal I. Controlled Tau Cleavage in Cells Reveals Abnormal Localizations of Tau Fragments. Neuroscience 2022; 518:162-177. [PMID: 35995336 DOI: 10.1016/j.neuroscience.2022.08.016] [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: 12/16/2021] [Revised: 06/08/2022] [Accepted: 08/16/2022] [Indexed: 11/15/2022]
Abstract
In several forms of dementia, such as Alzheimer's disease, the cytoskeleton-associated protein tau undergoes proteolysis, giving rise to fragments that have a toxic impact on neuronal homeostasis. How these fragments interact with cellular structures, in particular with the cytoskeleton, is currently incompletely understood. Here, we developed a method, derived from a Tobacco Etch Virus (TEV) protease system, to induce controlled cleavage of tau at specific sites. Five tau proteins containing specific TEV recognition sites corresponding to pathological proteolytic sites were engineered, and tagged with GFP at one end and mCherry at the other. Following controlled cleavage to produce GFP-N-terminal and C-terminal-mCherry fragments, we followed the fate of tau fragments in cells. Our results showed that whole engineered tau proteins associate with the cytoskeleton similarly to the non-modified tau, whereas tau fragments adopted different localizations with respect to the actin and microtubule cytoskeletons. These distinct localizations were confirmed by expressing each separate fragment in cells. Some cleavages - in particular cleavages at amino-acid positions 124 or 256 - displayed a certain level of cellular toxicity, with an unusual relocalization of the N-terminal fragments to the nucleus. Based on the data presented here, inducible cleavage of tau by the TEV protease appears to be a valuable tool to reproduce tau fragmentation in cells and study the resulting consequences on cell physiology.
Collapse
Affiliation(s)
- Anne Fourest-Lieuvin
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| | - Angélique Vinit
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Béatrice Blot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Anthime Perrot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eric Denarier
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédéric Saudou
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Isabelle Arnal
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
33
|
Alipour M, Motavaf M, Abdolmaleki P, Zali A, Ashrafi F, Safari S, Hajipour-Verdom B. Structural Analysis and Conformational Dynamics of Short Helical Hyperphosphorylated Segments of Tau Protein (Sequence 254–290) in Alzheimer’s Disease: A Molecular Dynamics Simulation Study. Front Mol Biosci 2022; 9:884705. [PMID: 36003083 PMCID: PMC9393928 DOI: 10.3389/fmolb.2022.884705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder whose early diagnosis leads to a chance for successful treatment and decreases the side effects. Hyperphosphorylation of tau proteins is a pathological hallmark of AD that causes it to lose its attachment ability to the microtubules. Alteration of tau structure due to its hyperphosphorylation is an exciting challenge regarding AD treatments. Here, we aimed to examine the structural alterations of short helical segments of tau protein with one to three phosphorylated sites by molecular dynamics simulation. Results indicated that the interaction of two similar segments with three phosphorylated sites (P-Ser262, 285, and 289) formed a compact and more stable structure than the one phosphorylated site complex (P-Ser262). Moreover, due to the high dynamics of the P-Ser262 complex, several structures were made with different conformational dynamics, but there was only one stable cluster of the P-Ser262, 285, and 289 complex during simulation. It seems that the P-Ser262, 285, and 289 complex plays an important role in the formation of paired helical filaments (PHFs) by forming a stable dimer. Generally, it is important to identify how structural features of segments in tau protein change when the phosphorylated sites increase from one to three sites and their effects on the formation of PHFs for drug design and diagnostic biomarkers.
Collapse
Affiliation(s)
- Mozhgan Alipour
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahsa Motavaf
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Ashrafi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Safari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Saeid Safari, ; Behnam Hajipour-Verdom,
| | - Behnam Hajipour-Verdom
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- *Correspondence: Saeid Safari, ; Behnam Hajipour-Verdom,
| |
Collapse
|
34
|
Silver nanoparticles induced hippocampal neuronal damage involved in mitophagy, mitochondrial biogenesis and synaptic degeneration. Food Chem Toxicol 2022; 166:113227. [PMID: 35697184 DOI: 10.1016/j.fct.2022.113227] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
Silver nanoparticles (AgNPs) could accumulate in the central nervous system (CNS) and induce neurotoxicity for their widespread use in industry and medicine. Mitochondria are vulnerable to toxicity of AgNPs, however, their role in the neurotoxicity remains unclear. This study aimed to evaluate AgNPs-induced synaptic degeneration in mouse hippocampal neurons (at a dose of 12-120 mg/kg BW via intravenous injection), and to further investigate mechanism of mitophagy, mitochondrial biogenesis process in the neurotoxicity. The results indicated that AgNPs accumulated in mouse hippocampal neurons and induced neurological deficits of learning and memory, which involved in synaptic degeneration accompanied with mitochondrial damage. Mechanistically, AgNPs exposure increased protein expression of PTEN-induced kinase 1 (PINK1), Parkin and inhibited peroxisome proliferator-activated receptor coactivator 1 alpha (PGC-1α) protein expression, caused disturbed mitophagy and mitochondrial biogenesis. AgNPs also induced synaptic damage by increasing the protein expression of synaptophysin and decreasing PSD95, MAP2 protein expression. AgNPs exposure even promoted protein expression of amyloid precursor protein (APP) using in amyloid-β (Aβ) cleavage. Furthermore, AgNPs induced hippocampal neuronal synaptic degeneration, mitophagy and mitochondrial biogenesis is dependent on particle-specific AgNPs rather than released silver ions. Our research could provide insights into the regulatory mechanisms of AgNPs-induced neurotoxicity. This study will shed the light of neurotoxicological evaluation of nanoparticles and possible early warning of biomedical applications.
Collapse
|
35
|
Xia P, Chen J, Bai X, Li M, Wang L, Lu Z. Key gene network related to primary ciliary dyskinesia in hippocampus of patients with Alzheimer’s disease revealed by weighted gene co-expression network analysis. BMC Neurol 2022; 22:198. [PMID: 35637434 PMCID: PMC9150314 DOI: 10.1186/s12883-022-02724-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD) is closely related to aging, showing an increasing incidence rate for years. As one of the main brain regions involved in AD, hippocampus has been extensively studied due to its association with many human diseases. However, little is known about its association with primary ciliary dyskinesia (PCD).
Material and Methods
The microarray data of hippocampus on AD were retrieved from the Gene Expression Omnibus (GEO) database to construct the co-expression network by weighted gene co-expression network analysis (WGCNA). The gene network modules associated with AD screened with the common genes were further annotated based on Gene Ontology (GO) database and enriched based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. The protein-protein interaction (PPI) network was constructed based on STRING database to identify the hub genes in the network.
Results
Genes involved in PCD were identified in the hippocampus of AD patients. Functional analysis revealed that these genes were mainly enriched in ciliary tissue, ciliary assembly, axoneme assembly, ciliary movement, microtubule based process, microtubule based movement, organelle assembly, axoneme dynamin complex, cell projection tissue, and microtubule cytoskeleton tissue. A total of 20 central genes, e.g., DYNLRB2, ZMYND10, DRC1, DNAH5, WDR16, TTC25, and ARMC4 were identified as hub genes related to PCD in hippocampus of AD patients.
Conclusion
Our study demonstrated that AD and PCD have common metabolic pathways. These common pathways provide novel evidence for further investigation of the pathophysiological mechanism and the hub genes suggest new therapeutic targets for the diagnosis and treatment of AD and PCD.
Subjects
Bioinformatics, Cell Biology, Molecular Biology, Neurology.
Collapse
|
36
|
Tsujikawa K, Hamanaka K, Riku Y, Hattori Y, Hara N, Iguchi Y, Ishigaki S, Hashizume A, Miyatake S, Mitsuhashi S, Miyazaki Y, Kataoka M, Jiayi L, Yasui K, Kuru S, Koike H, Kobayashi K, Sahara N, Ozaki N, Yoshida M, Kakita A, Saito Y, Iwasaki Y, Miyashita A, Iwatsubo T, Japanese Alzheimer’s Disease Neuroimaging Initiative (J-ADNI), Ikeuchi T, Japanese Longitudinal Biomarker Study in PSP and CBD (JALPAC) Consortium, Miyata T, Sobue G, Matsumoto N, Sahashi K, Katsuno M. Actin-binding protein filamin-A drives tau aggregation and contributes to progressive supranuclear palsy pathology. SCIENCE ADVANCES 2022; 8:eabm5029. [PMID: 35613261 PMCID: PMC9132466 DOI: 10.1126/sciadv.abm5029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
While amyloid-β lies upstream of tau pathology in Alzheimer's disease, key drivers for other tauopathies, including progressive supranuclear palsy (PSP), are largely unknown. Various tau mutations are known to facilitate tau aggregation, but how the nonmutated tau, which most cases with PSP share, increases its propensity to aggregate in neurons and glial cells has remained elusive. Here, we identified genetic variations and protein abundance of filamin-A in the PSP brains without tau mutations. We provided in vivo biochemical evidence that increased filamin-A levels enhance the phosphorylation and insolubility of tau through interacting actin filaments. In addition, reduction of filamin-A corrected aberrant tau levels in the culture cells from PSP cases. Moreover, transgenic mice carrying human filamin-A recapitulated tau pathology in the neurons. Our data highlight that filamin-A promotes tau aggregation, providing a potential mechanism by which filamin-A contributes to PSP pathology.
Collapse
Affiliation(s)
- Koyo Tsujikawa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neurology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
- Department of Neurology , National Hospital Organization Suzuka National Hospital, Suzuka, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuichi Riku
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yohei Iguchi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Hashizume
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Japan
| | - Satomi Mitsuhashi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Genomic Function and Diversity, Medical Research Institute Tokyo Medical and Dental University, Tokyo, Japan
| | - Yu Miyazaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mayumi Kataoka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Li Jiayi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keizo Yasui
- Department of Neurology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Satoshi Kuru
- Department of Neurology , National Hospital Organization Suzuka National Hospital, Suzuka, Japan
| | - Haruki Koike
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuko Saito
- Department of Neurology and Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Yasushi Iwasaki
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gen Sobue
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Corresponding author.
| |
Collapse
|
37
|
Caamaño-Moreno M, Gargini R. Tauopathies: the role of tau in cellular crosstalk and synaptic dysfunctions. Neuroscience 2022; 518:38-53. [PMID: 35272005 DOI: 10.1016/j.neuroscience.2022.02.034] [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: 12/30/2021] [Revised: 02/20/2022] [Accepted: 02/28/2022] [Indexed: 10/18/2022]
Abstract
Tauopathies are a group of neurodegenerative diseases among which are many of the most prevalent and with higher incidence worldwide, such as Alzheimer's disease (AD). According to the World Health Organization, this set of diseases will continue to increase their incidence, affecting millions of people by 2050. All of them are characterized by aberrant aggregation of tau protein in neurons and glia that are distributed in different brain regions according to their susceptibility. Numerous studies reveal that synaptic regulation not only has a neuronal component, but glia plays a fundamental role in it beyond its neuroinflammatory role. Despite this, it has not been emphasized how the glial inclusions of tau in this cell type directly affect this and many other essential functions, whose alterations have been related to the development of tauopathies. In this way, this review shows how tau inclusions in glia influence the synaptic dysfunctions that result in the cognitive symptoms characteristic of tauopathies. Thus, the mechanisms affected by inclusions in neurons, astrocytes, and oligodendrocytes are unraveled.
Collapse
Affiliation(s)
- Marta Caamaño-Moreno
- Instituto de investigaciones Biomédicas I+12, Hospital 12 de Octubre, Madrid, Spain
| | - Ricardo Gargini
- Instituto de investigaciones Biomédicas I+12, Hospital 12 de Octubre, Madrid, Spain; Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, 28220 Madrid, Spain.
| |
Collapse
|
38
|
Di Meo D, Ravindran P, Sadhanasatish T, Dhumale P, Püschel AW. The balance of mitochondrial fission and fusion in cortical axons depends on the kinases SadA and SadB. Cell Rep 2021; 37:110141. [PMID: 34936879 DOI: 10.1016/j.celrep.2021.110141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/17/2021] [Accepted: 11/29/2021] [Indexed: 01/21/2023] Open
Abstract
Neurons are highly polarized cells that display characteristic differences in the organization of their organelles in axons and dendrites. The kinases SadA and SadB (SadA/B) promote the formation of distinct axonal and dendritic extensions during the development of cortical and hippocampal neurons. Here, we show that SadA/B are required for the specific dynamics of axonal mitochondria. Ankyrin B (AnkB) stimulates the activity of SadA/B that function as regulators of mitochondrial dynamics through the phosphorylation of tau. Suppression of SadA/B or AnkB in cortical neurons induces the elongation of mitochondria by disrupting the balance of fission and fusion. SadA/B-deficient neurons show an accumulation of hyper-fused mitochondria and activation of the integrated stress response (ISR). The normal dynamics of axonal mitochondria could be restored by mild actin destabilization. Thus, the elongation after loss of SadA/B results from an excessive stabilization of actin filaments and reduction of Drp1 recruitment to mitochondria.
Collapse
Affiliation(s)
- Danila Di Meo
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität, Schloßplatz 5, 48149 Münster, Germany; Cells-in-Motion Interfaculty Center, University of Münster, 48149 Münster, Germany
| | - Priyadarshini Ravindran
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität, Schloßplatz 5, 48149 Münster, Germany
| | - Tanmay Sadhanasatish
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität, Schloßplatz 5, 48149 Münster, Germany; Cells-in-Motion Interfaculty Center, University of Münster, 48149 Münster, Germany
| | - Pratibha Dhumale
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität, Schloßplatz 5, 48149 Münster, Germany; Cells-in-Motion Interfaculty Center, University of Münster, 48149 Münster, Germany
| | - Andreas W Püschel
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität, Schloßplatz 5, 48149 Münster, Germany; Cells-in-Motion Interfaculty Center, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
39
|
Zhovmer AS, Manning A, Smith C, Hayes JB, Burnette DT, Wang J, Cartagena-Rivera AX, Dokholyan NV, Singh RK, Tabdanov ED. Mechanical Counterbalance of Kinesin and Dynein Motors in a Microtubular Network Regulates Cell Mechanics, 3D Architecture, and Mechanosensing. ACS NANO 2021; 15:17528-17548. [PMID: 34677937 PMCID: PMC9291236 DOI: 10.1021/acsnano.1c04435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Microtubules (MTs) and MT motor proteins form active 3D networks made of unstretchable cables with rod-like bending mechanics that provide cells with a dynamically changing structural scaffold. In this study, we report an antagonistic mechanical balance within the dynein-kinesin microtubular motor system. Dynein activity drives the microtubular network inward compaction, while isolated activity of kinesins bundles and expands MTs into giant circular bands that deform the cell cortex into discoids. Furthermore, we show that dyneins recruit MTs to sites of cell adhesion, increasing the topographic contact guidance of cells, while kinesins antagonize it via retraction of MTs from sites of cell adhesion. Actin-to-microtubule translocation of septin-9 enhances kinesin-MT interactions, outbalances the activity of kinesins over that of dyneins, and induces the discoid architecture of cells. These orthogonal mechanisms of MT network reorganization highlight the existence of an intricate mechanical balance between motor activities of kinesins and dyneins that controls cell 3D architecture, mechanics, and cell-microenvironment interactions.
Collapse
Affiliation(s)
- Alexander S. Zhovmer
- Center
for Biologics Evaluation and Research, U.S.
Food and Drug Administration, Silver Spring, Maryland 20903, United States
| | - Alexis Manning
- Center
for Biologics Evaluation and Research, U.S.
Food and Drug Administration, Silver Spring, Maryland 20903, United States
| | - Chynna Smith
- Section
on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - James B. Hayes
- Department
of Cell and Developmental Biology, Vanderbilt Medical Center, University of Vanderbilt, Nashville, Tennessee 37232, United States
| | - Dylan T. Burnette
- Department
of Cell and Developmental Biology, Vanderbilt Medical Center, University of Vanderbilt, Nashville, Tennessee 37232, United States
| | - Jian Wang
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
| | - Alexander X. Cartagena-Rivera
- Section
on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nikolay V. Dokholyan
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
- Department
of Biochemistry & Molecular Biology, Penn State College of Medicine, Pennsylvania State University, Hershey, Pennsylvania 17033, United States
| | - Rakesh K. Singh
- Department
of Obstetrics and Gynecology, University
of Rochester Medical Center, Rochester, New York 14620, United States
| | - Erdem D. Tabdanov
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
| |
Collapse
|
40
|
Pathak BK, Dey S, Mozumder S, Sengupta J. The role of membranes in function and dysfunction of intrinsically disordered amyloidogenic proteins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:397-434. [PMID: 35034725 DOI: 10.1016/bs.apcsb.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Membrane-protein interactions play a major role in human physiology as well as in diseases pathology. Interaction of a protein with the membrane was previously thought to be dependent on well-defined three-dimensional structure of the protein. In recent decades, however, it has become evident that a large fraction of the proteome, particularly in eukaryotes, stays disordered in solution and these proteins are termed as intrinsically disordered proteins (IDPs). Also, a vast majority of human proteomes have been reported to contain substantially long disordered regions, called intrinsically disordered regions (IDRs), in addition to the structurally ordered regions. IDPs exist in an ensemble of conformations and the conformational flexibility enables IDPs to achieve functional diversity. IDPs (and IDRs) are found to be important players in cell signaling, where biological membranes act as anchors for signaling cascades. Therefore, IDPs modulate the membrane architectures, at the same time membrane composition also affects the binding of IDPs. Because of intrinsic disorders, misfolding of IDPs often leads to formation of oligomers, protofibrils and mature fibrils through progressive self-association. Accumulation of amyloid-like aggregates of some of the IDPs is a known causative agent for numerous diseases. In this chapter we highlight recent advances in understanding membrane interactions of some of the intrinsically disordered proteins involved in the pathogenesis of human diseases.
Collapse
Affiliation(s)
- Bani Kumar Pathak
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sandip Dey
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sukanya Mozumder
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jayati Sengupta
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
41
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
42
|
Myrka A, Buck L. Cytoskeletal Arrest: An Anoxia Tolerance Mechanism. Metabolites 2021; 11:metabo11080561. [PMID: 34436502 PMCID: PMC8401981 DOI: 10.3390/metabo11080561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/16/2022] Open
Abstract
Polymerization of actin filaments and microtubules constitutes a ubiquitous demand for cellular adenosine-5′-triphosphate (ATP) and guanosine-5′-triphosphate (GTP). In anoxia-tolerant animals, ATP consumption is minimized during overwintering conditions, but little is known about the role of cell structure in anoxia tolerance. Studies of overwintering mammals have revealed that microtubule stability in neurites is reduced at low temperature, resulting in withdrawal of neurites and reduced abundance of excitatory synapses. Literature for turtles is consistent with a similar downregulation of peripheral cytoskeletal activity in brain and liver during anoxic overwintering. Downregulation of actin dynamics, as well as modification to microtubule organization, may play vital roles in facilitating anoxia tolerance. Mitochondrial calcium release occurs during anoxia in turtle neurons, and subsequent activation of calcium-binding proteins likely regulates cytoskeletal stability. Production of reactive oxygen species (ROS) formation can lead to catastrophic cytoskeletal damage during overwintering and ROS production can be regulated by the dynamics of mitochondrial interconnectivity. Therefore, suppression of ROS formation is likely an important aspect of cytoskeletal arrest. Furthermore, gasotransmitters can regulate ROS levels, as well as cytoskeletal contractility and rearrangement. In this review we will explore the energetic costs of cytoskeletal activity, the cellular mechanisms regulating it, and the potential for cytoskeletal arrest being an important mechanism permitting long-term anoxia survival in anoxia-tolerant species, such as the western painted turtle and goldfish.
Collapse
Affiliation(s)
- Alexander Myrka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
| | - Leslie Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Correspondence: ; Tel.: +1-416-978-3506
| |
Collapse
|
43
|
Abstract
Actin filaments and microtubules are cytoskeletal polymers that participate in many vital cell functions including division, morphogenesis, phagocytosis, and motility. Despite the persistent dogma that actin filament and microtubule networks are distinct in localization, structure, and function, a growing body of evidence shows that these elements are choreographed through intricate mechanisms sensitive to either polymer. Many proteins and cellular signals that mediate actin–microtubule interactions have already been identified. However, the impact of these regulators is typically assessed with actin filament or microtubule polymers alone, independent of the other system. Further, unconventional modes and regulators coordinating actin–microtubule interactions are still being discovered. Here we examine several methods of actin–microtubule crosstalk with an emphasis on the molecular links between both polymer systems and their higher-order interactions.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Jessica L Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210.,Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
44
|
Membrane interaction and disulphide-bridge formation in the unconventional secretion of Tau. Biosci Rep 2021; 41:229358. [PMID: 34308969 PMCID: PMC8350431 DOI: 10.1042/bsr20210148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/04/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Misfolded, pathological tau protein propagates from cell to cell causing neuronal degeneration in Alzheimer's disease and other tauopathies. The molecular mechanisms of this process have remained elusive. Unconventional secretion of tau takes place via several different routes, including direct penetration through the plasma membrane. Here, we show that tau secretion requires membrane interaction via disulphide bridge formation. Mutating residues that reduce tau interaction with membranes or formation of disulphide bridges decrease both tau secretion from cells, and penetration through artificial lipid membranes. Our results demonstrate that tau is indeed able to penetrate protein-free membranes in a process independent of active cellular processes and that both membrane interaction and disulphide bridge formation are needed for this process. QUARK-based de novo modelling of the second and third microtubule-binding repeat domains (MTBDs), in which the two cysteine residues of 4R isoforms of tau are located, supports the concept that this region of tau could form transient amphipathic helices for membrane interaction.
Collapse
|
45
|
Mages B, Fuhs T, Aleithe S, Blietz A, Hobusch C, Härtig W, Schob S, Krueger M, Michalski D. The Cytoskeletal Elements MAP2 and NF-L Show Substantial Alterations in Different Stroke Models While Elevated Serum Levels Highlight Especially MAP2 as a Sensitive Biomarker in Stroke Patients. Mol Neurobiol 2021; 58:4051-4069. [PMID: 33931805 PMCID: PMC8280005 DOI: 10.1007/s12035-021-02372-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
In the setting of ischemic stroke, the neurofilament subunit NF-L and the microtubule-associated protein MAP2 have proven to be exceptionally ischemia-sensitive elements of the neuronal cytoskeleton. Since alterations of the cytoskeleton have been linked to the transition from reversible to irreversible tissue damage, the present study investigates underlying time- and region-specific alterations of NF-L and MAP2 in different animal models of focal cerebral ischemia. Although NF-L is increasingly established as a clinical stroke biomarker, MAP2 serum measurements after stroke are still lacking. Therefore, the present study further compares serum levels of MAP2 with NF-L in stroke patients. In the applied animal models, MAP2-related immunofluorescence intensities were decreased in ischemic areas, whereas the abundance of NF-L degradation products accounted for an increase of NF-L-related immunofluorescence intensity. Accordingly, Western blot analyses of ischemic areas revealed decreased protein levels of both MAP2 and NF-L. The cytoskeletal alterations are further reflected at an ultrastructural level as indicated by a significant reduction of detectable neurofilaments in cortical axons of ischemia-affected areas. Moreover, atomic force microscopy measurements confirmed altered mechanical properties as indicated by a decreased elastic strength in ischemia-affected tissue. In addition to the results from the animal models, stroke patients exhibited significantly elevated serum levels of MAP2, which increased with infarct size, whereas serum levels of NF-L did not differ significantly. Thus, MAP2 appears to be a more sensitive stroke biomarker than NF-L, especially for early neuronal damage. This perspective is strengthened by the results from the animal models, showing MAP2-related alterations at earlier time points compared to NF-L. The profound ischemia-induced alterations further qualify both cytoskeletal elements as promising targets for neuroprotective therapies.
Collapse
Affiliation(s)
- Bianca Mages
- Institute of Anatomy, Leipzig University, Leipzig, Germany.
| | - Thomas Fuhs
- Section of Soft Matter Physics, Faculty of Physics and Geosciences, Leipzig University, Leipzig, Germany
| | - Susanne Aleithe
- Department of Neurology, Leipzig University, Leipzig, Germany
| | | | | | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Stefan Schob
- Department of Neuroradiology, Leipzig University, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
46
|
Rai SK, Savastano A, Singh P, Mukhopadhyay S, Zweckstetter M. Liquid-liquid phase separation of tau: From molecular biophysics to physiology and disease. Protein Sci 2021; 30:1294-1314. [PMID: 33930220 PMCID: PMC8197432 DOI: 10.1002/pro.4093] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022]
Abstract
Biomolecular condensation via liquid-liquid phase separation (LLPS) of intrinsically disordered proteins/regions (IDPs/IDRs), with and without nucleic acids, has drawn widespread interest due to the rapidly unfolding role of phase-separated condensates in a diverse range of cellular functions and human diseases. Biomolecular condensates form via transient and multivalent intermolecular forces that sequester proteins and nucleic acids into liquid-like membrane-less compartments. However, aberrant phase transitions into gel-like or solid-like aggregates might play an important role in neurodegenerative and other diseases. Tau, a microtubule-associated neuronal IDP, is involved in microtubule stabilization, regulates axonal outgrowth and transport in neurons. A growing body of evidence indicates that tau can accomplish some of its cellular activities via LLPS. However, liquid-to-solid transition resulting in the abnormal aggregation of tau is associated with neurodegenerative diseases. The physical chemistry of tau is crucial for governing its propensity for biomolecular condensation which is governed by various intermolecular and intramolecular interactions leading to simple one-component and complex multi-component condensates. In this review, we aim at capturing the current scientific state in unveiling the intriguing molecular mechanism of phase separation of tau. We particularly focus on the amalgamation of existing and emerging biophysical tools that offer unique spatiotemporal resolutions on a wide range of length- and time-scales. We also discuss the link between quantitative biophysical measurements and novel biological insights into biomolecular condensation of tau. We believe that this account will provide a broad and multidisciplinary view of phase separation of tau and its association with physiology and disease.
Collapse
Affiliation(s)
- Sandeep K. Rai
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, and Department of Chemical SciencesIndian Institute of Science Education and Research (IISER)MohaliIndia
| | - Adriana Savastano
- Research group Translational Structural BiologyGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Priyanka Singh
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, and Department of Chemical SciencesIndian Institute of Science Education and Research (IISER)MohaliIndia
| | - Samrat Mukhopadhyay
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, and Department of Chemical SciencesIndian Institute of Science Education and Research (IISER)MohaliIndia
| | - Markus Zweckstetter
- Research group Translational Structural BiologyGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department for NMR‐based Structural BiologyMax Planck Institute for Biophysical ChemistryGöttingenGermany
| |
Collapse
|
47
|
Mahato RR, Shandilya E, Dasgupta B, Maiti S. Dictating Catalytic Preference and Activity of a Nanoparticle by Modulating Its Multivalent Engagement. ACS Catal 2021. [DOI: 10.1021/acscatal.1c01991] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Rishi Ram Mahato
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Manauli 140306, India
| | - Ekta Shandilya
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Manauli 140306, India
| | - Basundhara Dasgupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Manauli 140306, India
| | - Subhabrata Maiti
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Manauli 140306, India
| |
Collapse
|
48
|
Ricolo D, Castro-Ribera J, Araújo SJ. Cytoskeletal players in single-cell branching morphogenesis. Dev Biol 2021; 477:22-34. [PMID: 34004181 DOI: 10.1016/j.ydbio.2021.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022]
Abstract
Branching networks are a very common feature of multicellular animals and underlie the formation and function of numerous organs including the nervous system, the respiratory system, the vasculature and many internal glands. These networks range from subcellular structures such as dendritic trees to large multicellular tissues such as the lungs. The production of branched structures by single cells, so called subcellular branching, which has been better described in neurons and in cells of the respiratory and vascular systems, involves complex cytoskeletal remodelling events. In Drosophila, tracheal system terminal cells (TCs) and nervous system dendritic arborisation (da) neurons are good model systems for these subcellular branching processes. During development, the generation of subcellular branches by single-cells is characterized by extensive remodelling of the microtubule (MT) network and actin cytoskeleton, followed by vesicular transport and membrane dynamics. In this review, we describe the current knowledge on cytoskeletal regulation of subcellular branching, based on the terminal cells of the Drosophila tracheal system, but drawing parallels with dendritic branching and vertebrate vascular subcellular branching.
Collapse
Affiliation(s)
- Delia Ricolo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Judith Castro-Ribera
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Sofia J Araújo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
49
|
Dioli C, Patrício P, Pinto LG, Marie C, Morais M, Vyas S, Bessa JM, Pinto L, Sotiropoulos I. Adult neurogenic process in the subventricular zone-olfactory bulb system is regulated by Tau protein under prolonged stress. Cell Prolif 2021; 54:e13027. [PMID: 33988263 PMCID: PMC8249793 DOI: 10.1111/cpr.13027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/08/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives The area of the subventricular zone (SVZ) in the adult brain exhibits the highest number of proliferative cells, which, together with the olfactory bulb (OB), maintains constant brain plasticity through the generation, migration and integration of newly born neurons. Despite Tau and its malfunction is increasingly related to deficits of adult hippocampal neurogenesis and brain plasticity under pathological conditions [e.g. in Alzheimer's disease (AD)], it remains unknown whether Tau plays a role in the neurogenic process of the SVZ and OB system under conditions of chronic stress, a well‐known sculptor of brain and risk factor for AD. Materials and methods Different types of newly born cells in SVZ and OB were analysed in animals that lack Tau gene (Tau‐KO) and their wild‐type littermates (WT) under control or chronic stress conditions. Results We demonstrate that chronic stress reduced the number of proliferating cells and neuroblasts in the SVZ leading to decreased number of newborn neurons in the OB of adult WT, but not Tau‐KO, mice. Interestingly, while stress‐evoked changes were not detected in OB granular cell layer, Tau‐KO exhibited increased number of mature neurons in this layer indicating altered neuronal migration due to Tau loss. Conclusions Our findings suggest the critical involvement of Tau in the neurogenesis suppression of SVZ and OB neurogenic niche under stressful conditions highlighting the role of Tau protein as an essential regulator of stress‐driven plasticity deficits.
Collapse
Affiliation(s)
- Chrysoula Dioli
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Institute of Biology Paris Seine, Team Gene Regulation and Adaptive Behaviors, Department of Neurosciences Paris Seine, Sorbonne Université, CNRS UMR 8246, INSERM U1130, Paris, France
| | - Patrícia Patrício
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lucilia-Goreti Pinto
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Clemence Marie
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mónica Morais
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sheela Vyas
- Institute of Biology Paris Seine, Team Gene Regulation and Adaptive Behaviors, Department of Neurosciences Paris Seine, Sorbonne Université, CNRS UMR 8246, INSERM U1130, Paris, France
| | - João M Bessa
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Luisa Pinto
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ioannis Sotiropoulos
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
50
|
Anderson SJ, Garamella J, Adalbert S, McGorty RJ, Robertson-Anderson RM. Subtle changes in crosslinking drive diverse anomalous transport characteristics in actin-microtubule networks. SOFT MATTER 2021; 17:4375-4385. [PMID: 33908593 PMCID: PMC8189643 DOI: 10.1039/d1sm00093d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Anomalous diffusion in crowded and complex environments is widely studied due to its importance in intracellular transport, fluid rheology and materials engineering. Specifically, diffusion through the cytoskeleton, a network comprised of semiflexible actin filaments and rigid microtubules that interact both sterically and via crosslinking, plays a principal role in viral infection, vesicle transport and targeted drug delivery. Here, we elucidate the impact of crosslinking on particle diffusion in composites of actin and microtubules with actin-actin, microtubule-microtubule and actin-microtubule crosslinking. We analyze a suite of transport metrics by coupling single-particle tracking and differential dynamic microscopy. Using these complementary techniques, we find that particles display non-Gaussian and non-ergodic subdiffusion that is markedly enhanced by cytoskeletal crosslinking, which we attribute to suppressed microtubule mobility. However, the extent to which transport deviates from normal Brownian diffusion depends strongly on the crosslinking motif - with actin-microtubule crosslinking inducing the most pronounced anomalous characteristics. Our results reveal that subtle changes to actin-microtubule interactions can have complex impacts on particle diffusion in cytoskeleton composites, and suggest that a combination of reduced filament mobility and more variance in actin mobilities leads to more strongly anomalous particle transport.
Collapse
Affiliation(s)
- S J Anderson
- Department of Physics & Biophysics, University of San Diego, San Diego, CA 92110, USA.
| | - J Garamella
- Department of Physics & Biophysics, University of San Diego, San Diego, CA 92110, USA.
| | - S Adalbert
- Department of Physics & Biophysics, University of San Diego, San Diego, CA 92110, USA.
| | - R J McGorty
- Department of Physics & Biophysics, University of San Diego, San Diego, CA 92110, USA.
| | | |
Collapse
|