1
|
Grigor'eva EV, Malakhova AA, Yarkova ES, Minina JM, Vyatkin YV, Nadtochy JA, Khabarova EA, Rzaev JA, Medvedev SP, Zakian SM. Generation and characterization of two induced pluripotent stem cell lines (ICGi052-A and ICGi052-B) from a patient with frontotemporal dementia with parkinsonism-17 associated with the pathological variant c.2013T>G in the MAPT gene. Vavilovskii Zhurnal Genet Selektsii 2024; 28:679-687. [PMID: 39722675 PMCID: PMC11668817 DOI: 10.18699/vjgb-24-76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 12/28/2024] Open
Abstract
Frontotemporal dementia with parkinsonism-17 is a neurodegenerative disease characterised by pathological aggregation of the tau protein with the formation of neurofibrillary tangles and subsequent neuronal death. The inherited form of frontotemporal dementia can be caused by mutations in several genes, including the MAPT gene on chromosome 17, which encodes the tau protein. As there are currently no medically approved treatments for frontotemporal dementia, there is an urgent need for research using in vitro cell models to understand the molecular genetic mechanisms that lead to the development of the disease, to identify targets for therapeutic intervention and to test potential drugs to prevent neuronal death. Analysis of exome sequencing data from a 46-year-old patient with a clinical diagnosis of Parkinson's disease revealed the presence of the pathological variant c.2013T>G (rs63750756) in the MAPT gene, which is associated with frontotemporal dementia with parkinsonism-17. By reprogramming the patient's peripheral blood mononuclear cells, we obtained induced pluripotent stem cells (iPSCs). Two iPSC lines were characterised in detail. Reprogramming was performed by transfection with non-integrating episomal vectors expressing the OCT4, SOX2, KLF4, LIN28, L-MYC and mp53DD proteins. The iPSC lines ICGi052-A and ICGi052-B proliferate stably, form colonies with a morphology characteristic of human pluripotent cells, have a normal diploid karyotype (46,XX), express endogenous alkaline phosphatase and pluripotency markers (OCT4, NANOG, SSEA-4 and TRA-1-60) and are able to differentiate into derivatives of three germ layers: ento-, ecto- and mesoderm. The iPSC lines obtained and characterised in detail in this work represent a unique tool for studying the molecular genetic mechanisms of the pathogenesis of frontotemporal dementia with parkinsonism-17, as well as for testing potential drugs in vitro.
Collapse
Affiliation(s)
- E V Grigor'eva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Malakhova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E S Yarkova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - J M Minina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - J A Nadtochy
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - E A Khabarova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Federal Neurosurgical Center of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - J A Rzaev
- Federal Neurosurgical Center of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - S P Medvedev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S M Zakian
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
2
|
Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol 2024; 25:845-864. [PMID: 39014245 DOI: 10.1038/s41580-024-00753-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/18/2024]
Abstract
Tau protein is involved in various cellular processes, including having a canonical role in binding and stabilization of microtubules in neurons. Tauopathies are neurodegenerative diseases marked by the abnormal accumulation of tau protein aggregates in neurons, as seen, for example, in conditions such as frontotemporal dementia and Alzheimer disease. Mutations in tau coding regions or that disrupt tau mRNA splicing, tau post-translational modifications and cellular stress factors (such as oxidative stress and inflammation) increase the tendency of tau to aggregate and interfere with its clearance. Pathological tau is strongly implicated in the progression of neurodegenerative diseases, and the propagation of tau aggregates is associated with disease severity. Recent technological advancements, including cryo-electron microscopy and disease models derived from human induced pluripotent stem cells, have increased our understanding of tau-related pathology in neurodegenerative conditions. Substantial progress has been made in deciphering tau aggregate structures and the molecular mechanisms that underlie protein aggregation and toxicity. In this Review, we discuss recent insights into the diverse cellular functions of tau and the pathology of tau inclusions and explore the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sarah A Naguib
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
3
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
4
|
Kindler D, Maschio C, Ni R, Zerbi V, Razansky D, Klohs J. Arterial spin labeling demonstrates preserved regional cerebral blood flow in the P301L mouse model of tauopathy. J Cereb Blood Flow Metab 2022; 42:686-693. [PMID: 34822744 PMCID: PMC8943618 DOI: 10.1177/0271678x211062274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is growing evidence for the vascular contribution to cognitive impairment and dementia in Alzheimer's disease (AD) and other neurodegenerative diseases. While perfusion deficits have been observed in patients with Alzheimer's disease and tauopaties, little is known about the role of tau in vascular dysfunction. In the present study, regional cerebral blood (rCBF) was characterized in P301L mice with arterial spin labeling. No differences in rCBF in P301L mice compared to their age-matched non-transgenic littermates at mid (10-12 months of age) and advanced (19-21 months of age) disease stages. This was concomitant with preservation of cortical brain structure as assessed with structural T2-weighted magnetic resonance imaging. These results show that hypoperfusion and neurodegeneration are not a phenotype of P301L mice. More studies are thus needed to understand the relationship of tau, neurodegeneration and vascular dysfunction and its modulators in AD and primary tauopathies.
Collapse
Affiliation(s)
- Diana Kindler
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland
| | - Cinzia Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| | - Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| | - Valerio Zerbi
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland.,Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, 27219ETH Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| |
Collapse
|
5
|
Kühn R, Mahajan A, Canoll P, Hargus G. Human Induced Pluripotent Stem Cell Models of Frontotemporal Dementia With Tau Pathology. Front Cell Dev Biol 2021; 9:766773. [PMID: 34858989 PMCID: PMC8631302 DOI: 10.3389/fcell.2021.766773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/27/2021] [Indexed: 12/04/2022] Open
Abstract
Neurodegenerative dementias are the most common group of neurodegenerative diseases affecting more than 40 million people worldwide. One of these diseases is frontotemporal dementia (FTD), an early onset dementia and one of the leading causes of dementia in people under the age of 60. FTD is a heterogeneous group of neurodegenerative disorders with pathological accumulation of particular proteins in neurons and glial cells including the microtubule-associated protein tau, which is deposited in its hyperphosphorylated form in about half of all patients with FTD. As for other patients with dementia, there is currently no cure for patients with FTD and thus several lines of research focus on the characterization of underlying pathogenic mechanisms with the goal to identify therapeutic targets. In this review, we provide an overview of reported disease phenotypes in induced pluripotent stem cell (iPSC)-derived neurons and glial cells from patients with tau-associated FTD with the aim to highlight recent progress in this fast-moving field of iPSC disease modeling. We put a particular focus on genetic forms of the disease that are linked to mutations in the gene encoding tau and summarize mutation-associated changes in FTD patient cells related to tau splicing and tau phosphorylation, microtubule function and cell metabolism as well as calcium homeostasis and cellular stress. In addition, we discuss challenges and limitations but also opportunities using differentiated patient-derived iPSCs for disease modeling and biomedical research on neurodegenerative diseases including FTD.
Collapse
Affiliation(s)
- Rebekka Kühn
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Aayushi Mahajan
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| |
Collapse
|
6
|
Wang P, Ye Y. Astrocytes in Neurodegenerative Diseases: A Perspective from Tauopathy and α-Synucleinopathy. Life (Basel) 2021; 11:life11090938. [PMID: 34575087 PMCID: PMC8471224 DOI: 10.3390/life11090938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases are aging-associated chronic pathological conditions affecting primarily neurons in humans. Inclusion bodies containing misfolded proteins have emerged as a common pathologic feature for these diseases. In many cases, misfolded proteins produced by a neuron can be transmitted to another neuron or a non-neuronal cell, leading to the propagation of disease-associated pathology. While undergoing intercellular transmission, misfolded proteins released from donor cells can often change the physiological state of recipient cells. Accumulating evidence suggests that astrocytes are highly sensitive to neuron-originated proteotoxic insults, which convert them into an active inflammatory state. Conversely, activated astrocytes can release a plethora of factors to impact neuronal functions. This review summarizes our current understanding of the complex molecular interplays between astrocyte and neuron, emphasizing on Tau and α-synuclein (α-syn), the disease-driving proteins for Alzheimer's and Parkinson's diseases, respectively.
Collapse
Affiliation(s)
| | - Yihong Ye
- Correspondence: ; Tel.: +1-301-594-0845; Fax: +1-301-496-0201
| |
Collapse
|
7
|
Chung DEC, Roemer S, Petrucelli L, Dickson DW. Cellular and pathological heterogeneity of primary tauopathies. Mol Neurodegener 2021; 16:57. [PMID: 34425874 PMCID: PMC8381569 DOI: 10.1186/s13024-021-00476-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
Microtubule-associated protein tau is abnormally aggregated in neuronal and glial cells in a range of neurodegenerative diseases that are collectively referred to as tauopathies. Multiple studies have suggested that pathological tau species may act as a seed that promotes aggregation of endogenous tau in naïve cells and contributes to propagation of tau pathology. While they share pathological tau aggregation as a common feature, tauopathies are distinct from one another with respect to predominant tau isoforms that accumulate and the selective vulnerability of brain regions and cell types that have tau inclusions. For instance, primary tauopathies present with glial tau pathology, while it is mostly neuronal in Alzheimer's disease (AD). Also, morphologies of tau inclusions can greatly vary even within the same cell type, suggesting distinct mechanisms or distinct tau conformers in each tauopathy. Neuropathological heterogeneity across tauopathies challenges our understanding of pathophysiology behind tau seeding and aggregation, as well as our efforts to develop effective therapeutic strategies for AD and other tauopathies. In this review, we describe diverse neuropathological features of tau inclusions in neurodegenerative tauopathies and discuss what has been learned from experimental studies with mouse models, advanced transcriptomics, and cryo-electron microscopy (cryo-EM) on the biology underlying cell type-specific tau pathology.
Collapse
Affiliation(s)
- Dah-eun Chloe Chung
- Department of Neuroscience, Mayo Clinic, 32224 Jacksonville, FL USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, 77030 Houston, TX USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 77030 Houston, TX USA
| | - Shanu Roemer
- Department of Neuroscience, Mayo Clinic, 32224 Jacksonville, FL USA
| | | | | |
Collapse
|
8
|
Maté de Gérando A, d'Orange M, Augustin E, Joséphine C, Aurégan G, Gaudin-Guérif M, Guillermier M, Hérard AS, Stimmer L, Petit F, Gipchtein P, Jan C, Escartin C, Selingue E, Carvalho K, Blum D, Brouillet E, Hantraye P, Gaillard MC, Bonvento G, Bemelmans AP, Cambon K. Neuronal tau species transfer to astrocytes and induce their loss according to tau aggregation state. Brain 2021; 144:1167-1182. [PMID: 33842937 DOI: 10.1093/brain/awab011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/24/2020] [Accepted: 11/01/2020] [Indexed: 11/12/2022] Open
Abstract
Deposits of different abnormal forms of tau in neurons and astrocytes represent key anatomo-pathological features of tauopathies. Although tau protein is highly enriched in neurons and poorly expressed by astrocytes, the origin of astrocytic tau is still elusive. Here, we used innovative gene transfer tools to model tauopathies in adult mouse brains and to investigate the origin of astrocytic tau. We showed in our adeno-associated virus (AAV)-based models and in Thy-Tau22 transgenic mice that astrocytic tau pathology can emerge secondarily to neuronal pathology. By designing an in vivo reporter system, we further demonstrated bidirectional exchanges of tau species between neurons and astrocytes. We then determined the consequences of tau accumulation in astrocytes on their survival in models displaying various status of tau aggregation. Using stereological counting of astrocytes, we report that, as for neurons, soluble tau species are highly toxic to some subpopulations of astrocytes in the hippocampus, whereas the accumulation of tau aggregates does not affect their survival. Thus, astrocytes are not mere bystanders of neuronal pathology. Our results strongly suggest that tau pathology in astrocytes may significantly contribute to clinical symptoms.
Collapse
Affiliation(s)
- Anastasie Maté de Gérando
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Marie d'Orange
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Emma Augustin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Charlène Joséphine
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Gwénaelle Aurégan
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Mylène Gaudin-Guérif
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Lev Stimmer
- MIRCen, INSERM-CEA, Platform for experimental pathology, U1169 and US27, F-92265 Fontenay-aux-Roses, France
| | - Fanny Petit
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Gipchtein
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Caroline Jan
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Erwan Selingue
- Université Paris-Saclay, CEA, Neurospin, 91191, Gif-sur-Yvette, France
| | - Kévin Carvalho
- Université Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Université Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Emmanuel Brouillet
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Alexis-Pierre Bemelmans
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Karine Cambon
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| |
Collapse
|
9
|
Karikari TK, Keeling S, Hill E, Lantero Rodrı́guez J, Nagel DA, Becker B, Höglund K, Zetterberg H, Blennow K, Hill EJ, Moffat KG. Extensive Plasmid Library to Prepare Tau Protein Variants and Study Their Functional Biochemistry. ACS Chem Neurosci 2020; 11:3117-3129. [PMID: 32833429 DOI: 10.1021/acschemneuro.0c00469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tau neurofibrillary tangles are key pathological features of Alzheimer's disease and other tauopathies. Recombinant protein technology is vital for studying the structure and function of tau in physiology and aggregation in pathophysiology. However, open-source and well-characterized plasmids for efficiently expressing and purifying different tau variants are lacking. We generated 44 sequence-verified plasmids including those encoding full length (FL) tau-441, its four-repeat microtubule-binding (K18) fragment, and their respective selected familial pathological variants (N279K, V337M, P301L, C291R, and S356T). Moreover, plasmids for expressing single (C291A), double (C291A/C322A), and triple (C291A/C322A/I260C) cysteine-modified variants were generated to study alterations in cysteine content and locations. Furthermore, protocols for producing representative tau forms were developed. We produced and characterized the aggregation behavior of the triple cysteine-modified tau-K18, often used in real-time cell internalization and aggregation studies because it can be fluorescently labeled on a cysteine outside the microtubule-binding core. Similar to the wild type (WT), triple cysteine-modified tau-K18 aggregated by progressive β-sheet enrichment, albeit at a slower rate. On prolonged incubation, cysteine-modified K18 formed paired helical filaments similar to those in Alzheimer's disease, sharing morphological phenotypes with WT tau-K18 filaments. Nonetheless, cysteine-modified tau-K18 filaments were significantly shorter (p = 0.002) and mostly wider than WT filaments, explainable by their different principal filament elongation pathways: vertical (end-to-end) and lateral growth for WT and cysteine-modified, respectively. Cysteine rearrangement may therefore induce filament polymorphism. Together, the plasmid library, the protein production methods, and the new insights into cysteine-dependent aggregation should facilitate further studies and the design of antiaggregation agents.
Collapse
Affiliation(s)
- Thomas K. Karikari
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
- Midlands Integrative Biosciences Training Partnership, University of Warwick, Coventry CV4 7AL, U.K
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg SE 43180, Sweden
| | - Sophie Keeling
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
| | - Emily Hill
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
| | - Juan Lantero Rodrı́guez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg SE 43180, Sweden
| | - David A. Nagel
- School of Life and Health Sciences, Aston University, Birmingham B4 7ET, U.K
| | - Bruno Becker
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg SE 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
| | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg SE 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg SE 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1E 6BT, U.K
- UK Dementia Research Institute at UCL, London WC1E 6BT, U.K
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg SE 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
| | - Eric J. Hill
- School of Life and Health Sciences, Aston University, Birmingham B4 7ET, U.K
| | - Kevin G. Moffat
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
| |
Collapse
|
10
|
TCW J. Human iPSC application in Alzheimer’s disease and Tau-related neurodegenerative diseases. Neurosci Lett 2019; 699:31-40. [DOI: 10.1016/j.neulet.2019.01.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/23/2018] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
|
11
|
Ritter ML, Avila J, García-Escudero V, Hernández F, Pérez M. Frontotemporal Dementia-Associated N279K Tau Mutation Localizes at the Nuclear Compartment. Front Cell Neurosci 2018; 12:202. [PMID: 30050413 PMCID: PMC6052045 DOI: 10.3389/fncel.2018.00202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/21/2018] [Indexed: 11/25/2022] Open
Abstract
Tau is a microtubule-associated protein that plays an important role in Alzheimer’s disease and related tauopathies. Approximately one-half of all cases of Frontotemporal dementia with parkinsonism-17 (FTDP-17) are caused by mutations in the MAPT gene. The N279K mutation is one of the three mutations more prevalent in FTDP-17 cases. Several studies have demonstrated that N279K Tau mutation alters alternative splicing inducing the presence of exon 10. Tau is mainly found in the cytosol of neuronal cells although it has also been localized within the nucleus. Here we demonstrate by biochemical and immunohistochemistry studies in COS-7 cells, that the proportion of mutant N279K Tau increases compared with wild-type at the cell nucleus although cell viability is not affected. These data will provide us with a better outline of the nuclear role of tau protein offering new clues related with this tauopathie.
Collapse
Affiliation(s)
- Maxi L Ritter
- Departamento de Anatomía Histología y Neurociencia, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Jesús Avila
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autonoma de Madrid (UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Carlos III Institute of Health, Madrid, Spain
| | - Vega García-Escudero
- Departamento de Anatomía Histología y Neurociencia, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autonoma de Madrid (UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Carlos III Institute of Health, Madrid, Spain
| | - Mar Pérez
- Departamento de Anatomía Histología y Neurociencia, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
12
|
Valachova B, Brezovakova V, Bugos O, Jadhav S, Smolek T, Novak P, Zilka N. A comparative study on pathological features of transgenic rat lines expressing either three or four repeat misfolded tau. J Comp Neurol 2018; 526:1777-1789. [DOI: 10.1002/cne.24447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Bernadeta Valachova
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
- Axon Neuroscience R&D Services SE; Bratislava Slovak Republic
| | - Veronika Brezovakova
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
| | - Ondrej Bugos
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
| | - Santosh Jadhav
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
- Axon Neuroscience R&D Services SE; Bratislava Slovak Republic
| | - Tomas Smolek
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
- Axon Neuroscience R&D Services SE; Bratislava Slovak Republic
| | - Petr Novak
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
| | - Norbert Zilka
- Centre of Excellence for Alzheimer's Disease and Related Disorders; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava Slovak Republic
- Axon Neuroscience R&D Services SE; Bratislava Slovak Republic
| |
Collapse
|
13
|
Nonclinical data supporting orphan medicinal product designations: lessons from rare neurological conditions. Drug Discov Today 2018; 23:26-48. [DOI: 10.1016/j.drudis.2017.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/01/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022]
|
14
|
Alzheimer’s Disease and Frontotemporal Lobar Degeneration: Mouse Models. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
15
|
Tau Isoforms Imbalance Impairs the Axonal Transport of the Amyloid Precursor Protein in Human Neurons. J Neurosci 2017; 37:58-69. [PMID: 28053030 DOI: 10.1523/jneurosci.2305-16.2016] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/21/2016] [Accepted: 11/04/2016] [Indexed: 01/11/2023] Open
Abstract
Tau, as a microtubule (MT)-associated protein, participates in key neuronal functions such as the regulation of MT dynamics, axonal transport, and neurite outgrowth. Alternative splicing of exon 10 in the tau primary transcript gives rise to protein isoforms with three (3R) or four (4R) MT binding repeats. Although tau isoforms are balanced in the normal adult human brain, imbalances in 3R:4R ratio have been tightly associated with the pathogenesis of several neurodegenerative disorders, yet the underlying molecular mechanisms remain elusive. Several studies exploiting tau overexpression and/or mutations suggested that perturbations in tau metabolism impair axonal transport. Nevertheless, no physiological model has yet demonstrated the consequences of altering the endogenous relative content of tau isoforms over axonal transport regulation. Here, we addressed this issue using a trans-splicing strategy that allows modulating tau exon 10 inclusion/exclusion in differentiated human-derived neurons. Upon changes in 3R:4R tau relative content, neurons showed no morphological changes, but live imaging studies revealed that the dynamics of the amyloid precursor protein (APP) were significantly impaired. Single trajectory analyses of the moving vesicles showed that predominance of 3R tau favored the anterograde movement of APP vesicles, increasing anterograde run lengths and reducing retrograde runs and segmental velocities. Conversely, the imbalance toward the 4R isoform promoted a retrograde bias by a significant reduction of anterograde velocities. These findings suggest that changes in 3R:4R tau ratio has an impact on the regulation of axonal transport and specifically in APP dynamics, which might link tau isoform imbalances with APP abnormal metabolism in neurodegenerative processes. SIGNIFICANCE STATEMENT The tau protein has a relevant role in the transport of cargos throughout neurons. Dysfunction in tau metabolism underlies several neurological disorders leading to dementia. In the adult human brain, two tau isoforms are found in equal amounts, whereas changes in such equilibrium have been associated with neurodegenerative diseases. We investigated the role of tau in human neurons in culture and found that perturbations in the endogenous balance of tau isoforms were sufficient to impair the transport of the Alzheimer's disease-related amyloid precursor protein (APP), although neuronal morphology was normal. Our results provide evidence of a direct relationship between tau isoform imbalance and defects in axonal transport, which induce an abnormal APP metabolism with important implications in neurodegeneration.
Collapse
|
16
|
Leyns CEG, Holtzman DM. Glial contributions to neurodegeneration in tauopathies. Mol Neurodegener 2017; 12:50. [PMID: 28662669 PMCID: PMC5492997 DOI: 10.1186/s13024-017-0192-x] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023] Open
Abstract
Tauopathies are a broad set of neurodegenerative dementias characterized by aggregation of the tau protein into filamentous inclusions that can be found in neurons and glial cells. Activated microglia, astrocytes and elevated levels of proinflammatory molecules are also pathological hallmarks that are found in brain regions affected by tau pathology. There has been abundant research in recent years to understand the role of gliosis and neuroinflammation in neurodegenerative diseases, particularly in Alzheimer's disease (AD) which is the most common form of dementia. AD is a tauopathy characterized by both extracellular amyloid-β plaques in addition to intracellular neurofibrillary tangles and neuropil threads containing aggregated tau protein. Accumulating evidence suggests that neuroinflammation offers a possible mechanistic link between these pathologies. Additionally, there appears to be a role for neuroinflammation in aggravating tau pathology and neurodegeneration in tauopathies featuring tau deposits as the predominant pathological signature. In this review, we survey the literature regarding inflammatory mechanisms that may impact neurodegeneration in AD and related tauopathies. We consider a physical role for microglia in the spread of tau pathology as well as the non-cell autonomous effects of secreted proinflammatory cytokines, specifically interleukin 1 beta, interleukin 6, tumor necrosis factor alpha and complement proteins. These molecules appear to have direct effects on tau pathophysiology and overall neuronal health. They also indirectly impact neuronal homeostasis by altering glial function. We conclude by proposing a complex role for gliosis and neuroinflammation in accelerating the progression of AD and other tauopathies.
Collapse
Affiliation(s)
- Cheryl E. G. Leyns
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - David M. Holtzman
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| |
Collapse
|
17
|
Laskowitz DT, Wang H, Chen T, Lubkin DT, Cantillana V, Tu TM, Kernagis D, Zhou G, Macy G, Kolls BJ, Dawson HN. Neuroprotective pentapeptide CN-105 is associated with reduced sterile inflammation and improved functional outcomes in a traumatic brain injury murine model. Sci Rep 2017; 7:46461. [PMID: 28429734 PMCID: PMC5399447 DOI: 10.1038/srep46461] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
At present, there are no proven pharmacological treatments demonstrated to improve long term functional outcomes following traumatic brain injury(TBI). In the setting of non-penetrating TBI, sterile brain inflammatory responses are associated with the development of cerebral edema, intracranial hypertension, and secondary neuronal injury. There is increasing evidence that endogenous apolipoprotein E(apoE) modifies the neuroinflammatory response through its role in downregulating glial activation, however, the intact apoE holoprotein does not cross the blood-brain barrier due to its size. To address this limitation, we developed a small 5 amino acid apoE mimetic peptide(CN-105) that mimics the polar face of the apoE helical domain involved in receptor interactions. The goal of this study was to investigate the therapeutic potential of CN-105 in a murine model of closed head injury. Treatment with CN-105 was associated with a durable improvement in functional outcomes as assessed by Rotarod and Morris Water Maze and a reduction in positive Fluoro-Jade B stained injured neurons and microglial activation. Administration of CN-105 was also associated with reduction in mRNA expression of a subset of inflammatory and immune-related genes.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA.,Aegis-CN LLC., Durham, NC, USA
| | - Haichen Wang
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tony Chen
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - David T Lubkin
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Viviana Cantillana
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tian Ming Tu
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Dawn Kernagis
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Guanen Zhou
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gary Macy
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bradley J Kolls
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hana N Dawson
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
18
|
Wobst HJ, Denk F, Oliver PL, Livieratos A, Taylor TN, Knudsen MH, Bengoa-Vergniory N, Bannerman D, Wade-Martins R. Increased 4R tau expression and behavioural changes in a novel MAPT-N296H genomic mouse model of tauopathy. Sci Rep 2017; 7:43198. [PMID: 28233851 PMCID: PMC5324134 DOI: 10.1038/srep43198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/20/2017] [Indexed: 11/29/2022] Open
Abstract
The microtubule-associated protein tau is implicated in various neurodegenerative diseases including Alzheimer's disease, progressive supranuclear palsy and corticobasal degeneration, which are characterized by intracellular accumulation of hyperphosphorylated tau. Mutations in the tau gene MAPT cause frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17). In the human central nervous system, six tau isoforms are expressed, and imbalances in tau isoform ratios are associated with pathology. To date, few animal models of tauopathy allow for the potential influence of these protein isoforms, relying instead on cDNA-based transgene expression. Using the P1-derived artificial chromosome (PAC) technology, we created mouse lines expressing all six tau isoforms from the human MAPT locus, harbouring either the wild-type sequence or the disease-associated N296H mutation on an endogenous Mapt-/- background. Animals expressing N296H mutant tau recapitulated early key features of tauopathic disease, including a tau isoform imbalance and tau hyperphosphorylation in the absence of somatodendritic tau inclusions. Furthermore, N296H animals displayed behavioural anomalies such as hyperactivity, increased time in the open arms of the elevated plus maze and increased immobility during the tail suspension test. The mouse models described provide an excellent model to study the function of wild-type or mutant tau in a highly physiological setting.
Collapse
Affiliation(s)
- Heike J. Wobst
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Franziska Denk
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter L. Oliver
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Achilleas Livieratos
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Tonya N. Taylor
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Maria H. Knudsen
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Nora Bengoa-Vergniory
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - David Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Bondulich MK, Guo T, Meehan C, Manion J, Rodriguez Martin T, Mitchell JC, Hortobagyi T, Yankova N, Stygelbout V, Brion JP, Noble W, Hanger DP. Tauopathy induced by low level expression of a human brain-derived tau fragment in mice is rescued by phenylbutyrate. Brain 2016; 139:2290-306. [PMID: 27297240 PMCID: PMC4958900 DOI: 10.1093/brain/aww137] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/01/2016] [Indexed: 12/11/2022] Open
Abstract
Human neurodegenerative tauopathies exhibit pathological tau aggregates in the brain along with diverse clinical features including cognitive and motor dysfunction. Post-translational modifications including phosphorylation, ubiquitination and truncation, are characteristic features of tau present in the brain in human tauopathy. We have previously reported an N-terminally truncated form of tau in human brain that is associated with the development of tauopathy and is highly phosphorylated. We have generated a new mouse model of tauopathy in which this human brain-derived, 35 kDa tau fragment (Tau35) is expressed in the absence of any mutation and under the control of the human tau promoter. Most existing mouse models of tauopathy overexpress mutant tau at levels that do not occur in human neurodegenerative disease, whereas Tau35 transgene expression is equivalent to less than 10% of that of endogenous mouse tau. Tau35 mice recapitulate key features of human tauopathies, including aggregated and abnormally phosphorylated tau, progressive cognitive and motor deficits, autophagic/lysosomal dysfunction, loss of synaptic protein, and reduced life-span. Importantly, we found that sodium 4-phenylbutyrate (Buphenyl®), a drug used to treat urea cycle disorders and currently in clinical trials for a range of neurodegenerative diseases, reverses the observed abnormalities in tau and autophagy, behavioural deficits, and loss of synapsin 1 in Tau35 mice. Our results show for the first time that, unlike other tau transgenic mouse models, minimal expression of a human disease-associated tau fragment in Tau35 mice causes a profound and progressive tauopathy and cognitive changes, which are rescued by pharmacological intervention using a clinically approved drug. These novel Tau35 mice therefore represent a highly disease-relevant animal model in which to investigate molecular mechanisms and to develop novel treatments for human tauopathies.
Collapse
Affiliation(s)
- Marie K Bondulich
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Tong Guo
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Christopher Meehan
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - John Manion
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Teresa Rodriguez Martin
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Jacqueline C Mitchell
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Tibor Hortobagyi
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Natalia Yankova
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Virginie Stygelbout
- 2 Laboratory of Histology, Neuroanatomy and Neuropathology (CP 620), ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine 808, route de Lennik, 1070 Brussels, Belgium
| | - Jean-Pierre Brion
- 2 Laboratory of Histology, Neuroanatomy and Neuropathology (CP 620), ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine 808, route de Lennik, 1070 Brussels, Belgium
| | - Wendy Noble
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Diane P Hanger
- 1 King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
20
|
Schoch KM, DeVos SL, Miller RL, Chun SJ, Norrbom M, Wozniak DF, Dawson HN, Bennett CF, Rigo F, Miller TM. Increased 4R-Tau Induces Pathological Changes in a Human-Tau Mouse Model. Neuron 2016; 90:941-7. [PMID: 27210553 DOI: 10.1016/j.neuron.2016.04.042] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/19/2015] [Accepted: 04/26/2016] [Indexed: 01/09/2023]
Abstract
Pathological evidence for selective four-repeat (4R) tau deposition in certain dementias and exon 10-positioned MAPT mutations together suggest a 4R-specific role in causing disease. However, direct assessments of 4R toxicity have not yet been accomplished in vivo. Increasing 4R-tau expression without change to total tau in human tau-expressing mice induced more severe seizures and nesting behavior abnormality, increased tau phosphorylation, and produced a shift toward oligomeric tau. Exon 10 skipping could also be accomplished in vivo, providing support for a 4R-tau targeted approach to target 4R-tau toxicity and, in cases of primary MAPT mutation, eliminate the disease-causing mutation.
Collapse
Affiliation(s)
- Kathleen M Schoch
- Department of Neurology, Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO 63110
| | - Sarah L DeVos
- Department of Neurology, Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO 63110
| | - Rebecca L Miller
- Department of Neurology, Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO 63110
| | | | | | - David F Wozniak
- Taylor Family Institute for Innovative Psychiatric Research, Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110
| | - Hana N Dawson
- Department of Neurology, Duke University Medical Center, Durham, NC 27710
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010
| | - Timothy M Miller
- Department of Neurology, Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO 63110.
| |
Collapse
|
21
|
Kadri F, Pacifici M, Wilk A, Parker-Struckhoff A, Del Valle L, Hauser KF, Knapp PE, Parsons C, Jeansonne D, Lassak A, Peruzzi F. HIV-1-Tat Protein Inhibits SC35-mediated Tau Exon 10 Inclusion through Up-regulation of DYRK1A Kinase. J Biol Chem 2015; 290:30931-46. [PMID: 26534959 PMCID: PMC4692221 DOI: 10.1074/jbc.m115.675751] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/02/2015] [Indexed: 01/08/2023] Open
Abstract
The HIV-1 transactivator protein Tat is implicated in the neuronal damage that contributes to neurocognitive impairment affecting people living with HIV/AIDS. Aberrant splicing of TAU exon 10 results in tauopathies characterized by alterations in the proportion of TAU isoforms containing three (3R) or four (4R) microtubule-binding repeats. The splicing factor SC35/SRSF2 binds to nuclear RNA and facilitates the incorporation of exon 10 in the TAU molecule. Here, we utilized clinical samples, an animal model, and neuronal cell cultures and found that Tat promotes TAU 3R up-regulation through increased levels of phosphorylated SC35, which is retained in nuclear speckles. This mechanism involved Tat-mediated increased expression of DYRK1A and was prevented by DYRK1A silencing. In addition, we found that Tat associates with TAU RNA, further demonstrating that Tat interferes with host RNA metabolism in the absence of viral infection. Altogether, our data unravel a novel mechanism of Tat-mediated neuronal toxicity through dysregulation of the SC35-dependent alternative splicing of TAU exon 10. Furthermore, the increased immunostaining of DYRK1A in HIV+ brains without pathology points at dysregulation of DYRK1A as an early event in the neuronal complications of HIV infection.
Collapse
Affiliation(s)
- Ferdous Kadri
- From the Department of Medicine, Stanley S. Scott Cancer Center, and Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112 and
| | - Marco Pacifici
- From the Department of Medicine, Stanley S. Scott Cancer Center, and
| | - Anna Wilk
- From the Department of Medicine, Stanley S. Scott Cancer Center, and
| | | | - Luis Del Valle
- From the Department of Medicine, Stanley S. Scott Cancer Center, and
| | | | - Pamela E Knapp
- Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia 23284
| | | | - Duane Jeansonne
- From the Department of Medicine, Stanley S. Scott Cancer Center, and
| | - Adam Lassak
- From the Department of Medicine, Stanley S. Scott Cancer Center, and
| | - Francesca Peruzzi
- From the Department of Medicine, Stanley S. Scott Cancer Center, and
| |
Collapse
|
22
|
Vernay A, Sellal F, René F. Evaluating Behavior in Mouse Models of the Behavioral Variant of Frontotemporal Dementia: Which Test for Which Symptom? NEURODEGENER DIS 2015; 16:127-39. [PMID: 26517704 DOI: 10.1159/000439253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/07/2015] [Indexed: 11/19/2022] Open
Abstract
The behavioral variant of frontotemporal dementia (bvFTD) is a neurodegenerative disease affecting people in their early sixties, characterized by dramatic changes in individual and social behavior. Despite the heterogeneity in the presentation of the clinical symptoms of bvFTD, some characteristic changes can be highlighted. Social disinhibition, changes in food preferences as well as loss of empathy and apathy are commonly described. This is accompanied by a characteristic and dramatic atrophy of the prefrontal cortex with the accumulation of protein aggregates in the neurons in this area. Several causative mutations in different genes have been discovered, allowing the development of transgenic animal models, especially mouse models. In mice, attention has been focused on the histopathological aspects of the pathology, but now studies are taking interest in assessing the behavioral phenotype of FTD models. Finding the right test corresponding to human symptoms is quite challenging, especially since the frontal cortex is much less developed in mice than in humans. Although challenging, the ability to detect relevant prefrontal cortex impairments in mice is crucial for therapeutic approaches. In this review, we aim to present the approaches that have been used to model the behavioral symptoms of FTD and to explore other relevant approaches to assess behavior involving the prefrontal cortex, as well as the deficits associated with FTD.
Collapse
Affiliation(s)
- Aurélia Vernay
- INSERM, U1118, Laboratoire des Mx00E9;canismes Centraux et Px00E9;riphx00E9;riques de la Neurodx00E9;gx00E9;nx00E9;rescence, Strasbourg, France
| | | | | |
Collapse
|
23
|
McCarthy A, Lonergan R, Olszewska DA, O'Dowd S, Cummins G, Magennis B, Fallon EM, Pender N, Huey ED, Cosentino S, O'Rourke K, Kelly BD, O'Connell M, Delon I, Farrell M, Spillantini MG, Rowland LP, Fahn S, Craig P, Hutton M, Lynch T. Closing the tau loop: the missing tau mutation. Brain 2015; 138:3100-9. [PMID: 26297556 DOI: 10.1093/brain/awv234] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/27/2015] [Indexed: 12/30/2022] Open
Abstract
Frontotemporal lobar degeneration comprises a group of disorders characterized by behavioural, executive, language impairment and sometimes features of parkinsonism and motor neuron disease. In 1994 we described an Irish-American family with frontotemporal dementia linked to chromosome 17 associated with extensive tau pathology. We named this disinhibition-dementia-parkinsonism-amyotrophy complex. We subsequently identified mutations in the MAPT gene. Eleven MAPT gene splice site stem loop mutations were identified over time except for 5' splice site of exon 10. We recently identified another Irish family with autosomal dominant early amnesia and behavioural change or parkinsonism associated with the 'missing' +15 mutation at the intronic boundary of exon 10. We performed a clinical, neuropsychological and neuroimaging study on the proband and four siblings, including two affected siblings. We sequenced MAPT and performed segregation analysis. We looked for a biological effect of the tau variant by performing real-time polymerase chain reaction analysis of RNA extracted from human embryonic kidney cells transfected with exon trapping constructs. We found a c.915+15A>C exon 10/intron 10 stem loop mutation in all affected subjects but not in the unaffected. The c.915+15A>C variant caused a shift in tau splicing pattern to a predominantly exon 10+ pattern presumably resulting in predominant 4 repeat tau and little 3 repeat tau. This strongly suggests that the c.915+15A>C variant is a mutation and that it causes frontotemporal dementia linked to chromosome 17 in this pedigree by shifting tau transcription and translation to +4 repeat tau. Tau (MAPT) screening should be considered in families where amnesia or atypical parkinsonism coexists with behavioural disturbance early in the disease process. We describe the final missing stem loop tau mutation predicted 15 years ago. Mutations have now been identified at all predicted sites within the 'stem' when the stem-loop model was first proposed and no mutations have been found within the 'loop' region as expected. Therefore we 'close the tau loop' having 'opened the loop' 21 years ago.
Collapse
Affiliation(s)
- Allan McCarthy
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Roisin Lonergan
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Diana A Olszewska
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Sean O'Dowd
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Gemma Cummins
- 2 Department of Clinical Neuroscience, Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Brian Magennis
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Emer M Fallon
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Niall Pender
- 3 Department of Psychology, Beaumont Hospital, Beaumont Rd, Dublin 9, Ireland, Department of Psychology, Royal College of Surgeons in Ireland
| | - Edward D Huey
- 4 Departments of Psychiatry and Neurology, College of Physicians and Surgeons, University Medical Centre, 630 West 168th Street, New York, NY 10032, USA
| | - Stephanie Cosentino
- 5 Cognitive Neuroscience Section, Department of Neurology, Columbia University Medical Centre, 630 West 168th Street, New York, NY 10032, USA
| | - Killian O'Rourke
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Brendan D Kelly
- 6 Department of Psychiatry, Mater Misericordiae University Hospital, 63 Eccles Street, Dublin 7, Ireland
| | - Martin O'Connell
- 7 Department of Radiology, Mater Misericordiae University Hospital, North Circular Road, Dublin 7, Ireland
| | - Isabelle Delon
- 8 Medical Genetics Service, Cambridge University Hospital NHS Foundation Trust, Addenbrooke's Treatment Centre, Hills Road, Cambridge, CB2 0QQ, UK
| | - Michael Farrell
- 9 Department of Neuropathology, Beaumont Hospital, Beaumont Road, Dublin, Ireland
| | - Maria Grazia Spillantini
- 10 Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Lewis P Rowland
- 11 The Neurological Institute, Columbia University, 710 West 168th Street, New York, NY 10032-3784, USA
| | - Stanley Fahn
- 11 The Neurological Institute, Columbia University, 710 West 168th Street, New York, NY 10032-3784, USA
| | - Peter Craig
- 12 Eli Lilly, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Michael Hutton
- 12 Eli Lilly, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Tim Lynch
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| |
Collapse
|
24
|
Dujardin S, Colin M, Buée L. Invited review: Animal models of tauopathies and their implications for research/translation into the clinic. Neuropathol Appl Neurobiol 2015; 41:59-80. [DOI: 10.1111/nan.12200] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/23/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Simon Dujardin
- Inserm, UMR1172 Jean-Pierre Aubert Research Centre; Lille France
- Faculté de Médecine; Université de Lille; France
- Memory Clinic; CHRU; Lille France
| | - Morvane Colin
- Inserm, UMR1172 Jean-Pierre Aubert Research Centre; Lille France
- Faculté de Médecine; Université de Lille; France
- Memory Clinic; CHRU; Lille France
| | - Luc Buée
- Inserm, UMR1172 Jean-Pierre Aubert Research Centre; Lille France
- Faculté de Médecine; Université de Lille; France
- Memory Clinic; CHRU; Lille France
| |
Collapse
|
25
|
Intravenous immunoglobulin G improves neurobehavioral and histological outcomes after traumatic brain injury in mice. J Neuroimmunol 2014; 276:112-8. [DOI: 10.1016/j.jneuroim.2014.08.626] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/25/2014] [Accepted: 08/27/2014] [Indexed: 11/21/2022]
|
26
|
Siva K, Covello G, Denti MA. Exon-skipping antisense oligonucleotides to correct missplicing in neurogenetic diseases. Nucleic Acid Ther 2014; 24:69-86. [PMID: 24506781 DOI: 10.1089/nat.2013.0461] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Alternative splicing is an important regulator of the transcriptome. However, mutations may cause alteration of splicing patterns, which in turn leads to disease. During the past 10 years, exon skipping has been looked upon as a powerful tool for correction of missplicing in disease and progress has been made towards clinical trials. In this review, we discuss the use of antisense oligonucleotides to correct splicing defects through exon skipping, with a special focus on diseases affecting the nervous system, and the latest stage achieved in its progress.
Collapse
Affiliation(s)
- Kavitha Siva
- 1 Center for Integrative Biology (CIBIO), University of Trento , Trento, Italy
| | | | | |
Collapse
|
27
|
Roberts JM, Ennajdaoui H, Edmondson C, Wirth B, Sanford J, Chen B. Splicing factor TRA2B is required for neural progenitor survival. J Comp Neurol 2014; 522:372-92. [PMID: 23818142 PMCID: PMC3855887 DOI: 10.1002/cne.23405] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/20/2013] [Accepted: 06/21/2013] [Indexed: 01/08/2023]
Abstract
Alternative splicing of pre-mRNAs can rapidly regulate the expression of large groups of proteins. The RNA binding protein TRA2B (SFRS10) plays well-established roles in developmentally regulated alternative splicing during Drosophila sexual differentiation. TRA2B is also essential for mammalian embryogenesis and is implicated in numerous human diseases. Precise regulation of alternative splicing is critical to the development and function of the central nervous system; however, the requirements for specific splicing factors in neurogenesis are poorly understood. This study focuses on the role of TRA2B in mammalian brain development. We show that, during murine cortical neurogenesis, TRA2B is expressed in both neural progenitors and cortical projection neurons. Using cortex-specific Tra2b mutant mice, we show that TRA2B depletion results in apoptosis of the neural progenitor cells as well as disorganization of the cortical plate. Thus, TRA2B is essential for proper development of the cerebral cortex.
Collapse
Affiliation(s)
- Jacqueline M Roberts
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Hanane Ennajdaoui
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Carina Edmondson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Brunhilde Wirth
- Institute of Human Genetics, Institute for Genetics and Center for Molecular Medicine Cologne, University of Cologne, Cologne 50931, Germany
| | - Jeremy Sanford
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Bin Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| |
Collapse
|
28
|
Alzheimer’s Disease and Frontotemporal Lobar Degeneration: Mouse Models. NEURODEGENER DIS 2014. [DOI: 10.1007/978-1-4471-6380-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
29
|
Balczon R, Prasain N, Ochoa C, Prater J, Zhu B, Alexeyev M, Sayner S, Frank DW, Stevens T. Pseudomonas aeruginosa exotoxin Y-mediated tau hyperphosphorylation impairs microtubule assembly in pulmonary microvascular endothelial cells. PLoS One 2013; 8:e74343. [PMID: 24023939 PMCID: PMC3762819 DOI: 10.1371/journal.pone.0074343] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 08/01/2013] [Indexed: 12/21/2022] Open
Abstract
Pseudomonas aeruginosa uses a type III secretion system to introduce the adenylyl and guanylyl cyclase exotoxin Y (ExoY) into the cytoplasm of endothelial cells. ExoY induces Tau hyperphosphorylation and insolubility, microtubule breakdown, barrier disruption and edema, although the mechanism(s) responsible for microtubule breakdown remain poorly understood. Here we investigated both microtubule behavior and centrosome activity to test the hypothesis that ExoY disrupts microtubule dynamics. Fluorescence microscopy determined that infected pulmonary microvascular endothelial cells contained fewer microtubules than control cells, and further studies demonstrated that the microtubule-associated protein Tau was hyperphosphorylated following infection and dissociated from microtubules. Disassembly/reassembly studies determined that microtubule assembly was disrupted in infected cells, with no detectable effects on either microtubule disassembly or microtubule nucleation by centrosomes. This effect of ExoY on microtubules was abolished when the cAMP-dependent kinase phosphorylation site (Ser-214) on Tau was mutated to a non-phosphorylatable form. These studies identify Tau in microvascular endothelial cells as the target of ExoY in control of microtubule architecture following pulmonary infection by Pseudomonas aeruginosa and demonstrate that phosphorylation of tau following infection decreases microtubule assembly.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- * E-mail:
| | - Nutan Prasain
- Department of Pediatrics, University of Indiana School of Medicine, Indianapolis, Indiana, United States of America
| | - Cristhiaan Ochoa
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
| | - Jason Prater
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Medicine, University of South Alabama, Mobile, Alabama, United States of America
| | - Bing Zhu
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
| | - Mikhail Alexeyev
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Sarah Sayner
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama, United States of America
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
| | - Dara W. Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Troy Stevens
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, United States of America
- Department of Medicine, University of South Alabama, Mobile, Alabama, United States of America
| |
Collapse
|
30
|
Umeda T, Yamashita T, Kimura T, Ohnishi K, Takuma H, Ozeki T, Takashima A, Tomiyama T, Mori H. Neurodegenerative Disorder FTDP-17–Related Tau Intron 10 +16C→T Mutation Increases Tau Exon 10 Splicing and Causes Tauopathy in Transgenic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:211-25. [DOI: 10.1016/j.ajpath.2013.03.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 02/27/2013] [Accepted: 03/21/2013] [Indexed: 01/12/2023]
|
31
|
Wolfe MS. The role of tau in neurodegenerative diseases and its potential as a therapeutic target. SCIENTIFICA 2012; 2012:796024. [PMID: 24278740 PMCID: PMC3820460 DOI: 10.6064/2012/796024] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/05/2012] [Indexed: 06/01/2023]
Abstract
The abnormal deposition of proteins in and around neurons is a common pathological feature of many neurodegenerative diseases. Among these pathological proteins, the microtubule-associated protein tau forms intraneuronal filaments in a spectrum of neurological disorders. The discovery that dominant mutations in the MAPT gene encoding tau are associated with familial frontotemporal dementia strongly supports abnormal tau protein as directly involved in disease pathogenesis. This and other evidence suggest that tau is a worthwhile target for the prevention or treatment of tau-associated neurodegenerative diseases, collectively called tauopathies. However, it is critical to understand the normal biological roles of tau, the specific molecular events that induce tau to become neurotoxic, the biochemical nature of pathogenic tau, the means by which pathogenic tau exerts neurotoxicity, and how tau pathology propagates. Based on known differences between normal and abnormal tau, a number of approaches have been taken toward the discovery of potential therapeutics. Key questions still remain open, such as the nature of the connection between the amyloid- β protein of Alzheimer's disease and tau pathology. Answers to these questions should help better understand the nature of tauopathies and may also reveal new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Michael S. Wolfe
- Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, H.I.M. 754, Boston, MA 02115, USA
| |
Collapse
|
32
|
Cárdenas AM, Ardiles AO, Barraza N, Baéz-Matus X, Caviedes P. Role of tau protein in neuronal damage in Alzheimer's disease and Down syndrome. Arch Med Res 2012; 43:645-54. [PMID: 23142525 DOI: 10.1016/j.arcmed.2012.10.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 10/22/2012] [Indexed: 01/09/2023]
Abstract
Neurodegenerative disorders constitute a growing concern worldwide. Their incidence has increased steadily, in particular among the elderly, a high-risk population that is becoming an important segment of society. Neurodegenerative mechanisms underlie many ailments such as Parkinson's disease, Huntington's disease, Alzheimer's disease (AD) and Down syndrome (DS, trisomy 21). Interestingly, there is increasing evidence suggesting that many such diseases share pathogenic mechanisms at the cellular and subcellular levels. These include altered protein misfolding, impaired autophagy, mitochondrial dysfunction, membrane damage, and altered axonal transport. Regarding AD and DS, the first common link comes from observations that DS patients undergo AD-like pathology early in adulthood. Also, the gene encoding for the amyloid precursor protein is present in human autosome 21 and in murine chromosome 16, an animal model of DS. Important functions related to preservation of normal neuronal architecture are impaired in both conditions. In particular, the stable assembly of microtubules, which is critical for the cytoskeleton, is impaired in AD and DS. In this process, tau protein plays a pivotal role in controlling microtubule stability. Abnormal tau expression and hyperphosphorylation are common features in both conditions, yet the mechanisms leading to these phenomena remain obscure. In the present report we review possible common mechanisms that may alter tau expression and function, in particular in relation to the effect of certain overexpressed DS-related genes, using cellular models of human DS. The latter contributes to the identification of possible therapeutic targets that could aid in the treatment of both AD and DS.
Collapse
Affiliation(s)
- Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| | | | | | | | | |
Collapse
|
33
|
Laskowitz DT, Lei B, Dawson HN, Wang H, Bellows ST, Christensen DJ, Vitek MP, James ML. The apoE-mimetic peptide, COG1410, improves functional recovery in a murine model of intracerebral hemorrhage. Neurocrit Care 2012; 16:316-26. [PMID: 21989844 DOI: 10.1007/s12028-011-9641-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Apolipoprotein E has previously been demonstrated to modulate acute brain injury responses, and administration of COG1410, an apoE-mimetic peptide derived from the receptor-binding region of apoE, improves outcome in preclinical models of acute neurological injury. In the current study, we sought to establish the optimal dose and timing of peptide administration associated with improved functional outcome in a murine model of intracerebral hemorrhage (ICH). METHODS Ten to twelve-week-old C57/BL6 male mice were injured by collagenase-induced ICH and randomly selected to receive either vehicle or one of four doses of COG1410 (0.5, 1, 2, or 4 mg/kg) via tail vein injection at 30 min after injury and then daily for 5 days. The injured mice were euthanized at various time points to assess inflammatory mediators, cerebral edema, and hematoma volume. Over the first 5 days following injury, vestibulomotor function was tested via Rotorod (RR) latency. After an optimal dose was demonstrated, a final cohort of animals was injured with ICH and randomly assigned to receive the first dose of COG1410 or vehicle at increasingly longer treatment initiation times after injury. The mice were then assessed for functional deficit via RR testing over the first 5 days following injury. RESULTS The mice receiving 2 mg/kg of COG1410 after injury demonstrated reduced functional deficit, decreased brain concentrations of inflammatory proteins, and less cerebral edema, although hematoma volume did not vary. The improved RR performance was maintained when peptide administration was delayed for up to 2 h after ICH. CONCLUSIONS COG1410 administered at a dose of 2 mg/kg within 2 h after injury improves functional recovery in a murine model of ICH.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Department of Medicine (Neurology), Multidisciplinary Neuroprotection Laboratories, Duke University, Durham, NC, USA>
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Anderson ES, Lin CH, Xiao X, Stoilov P, Burge CB, Black DL. The cardiotonic steroid digitoxin regulates alternative splicing through depletion of the splicing factors SRSF3 and TRA2B. RNA (NEW YORK, N.Y.) 2012; 18:1041-9. [PMID: 22456266 PMCID: PMC3334691 DOI: 10.1261/rna.032912.112] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 02/24/2012] [Indexed: 05/24/2023]
Abstract
Modulation of alternative pre-mRNA splicing is a potential approach to therapeutic targeting for a variety of human diseases. We investigated the mechanism by which digitoxin, a member of the cardiotonic steroid class of drugs, regulates alternative splicing. Transcriptome-wide analysis identified a large set of alternative splicing events that change after digitoxin treatment. Within and adjacent to these regulated exons, we identified enrichment of potential binding sites for the splicing factors SRp20 (SRSF3/SFRS3) and Tra2-β (SFRS10/TRA2B). We further find that both of these proteins are depleted from cells by digitoxin treatment. Characterization of SRp20 and Tra2-β splicing targets revealed that many, but not all, digitoxin-induced splicing changes can be attributed to the depletion of one or both of these factors. Re-expression of SRp20 or Tra2-β after digitoxin treatment restores normal splicing of their targets, indicating that the digitoxin effect is directly due to these factors. These results demonstrate that cardiotonic steroids, long prescribed in the clinical treatment of heart failure, have broad effects on the cellular transcriptome through these and likely other RNA binding proteins. The approach described here can be used to identify targets of other potential therapeutics that act as alternative splicing modulators.
Collapse
Affiliation(s)
- Erik S. Anderson
- Molecular Biology Interdepartmental Graduate Program
- Medical Scientist Training Program
| | - Chia-Ho Lin
- Microbiology, Immunology and Molecular Genetics
- Howard Hughes Medical Institute
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology
- Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| | - Peter Stoilov
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Christopher B. Burge
- Department of Biology, Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | - Douglas L. Black
- Microbiology, Immunology and Molecular Genetics
- Howard Hughes Medical Institute
- Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
35
|
Indraswari F, Wang H, Lei B, James ML, Kernagis D, Warner DS, Dawson HN, Laskowitz DT. Statins improve outcome in murine models of intracranial hemorrhage and traumatic brain injury: a translational approach. J Neurotrauma 2012; 29:1388-400. [PMID: 22233347 DOI: 10.1089/neu.2011.2117] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) and intracerebral hemorrhage (ICH) are leading causes of neurological mortality and disability in the U.S. However, therapeutic options are limited and clinical management remains largely supportive. HMG-CoA reductase inhibitors (statins) have pleiotropic mechanisms of action in the setting of acute brain injury, and have been demonstrated to improve outcomes in preclinical models of ICH and TBI. To facilitate translation to clinical practice, we now characterize the optimal statin and dosing paradigm in murine models of ICH and TBI. In a preclinical model of TBI, mice received vehicle, simvastatin, and rosuvastatin at doses of 1 mg/kg and 5 mg/kg for 5 days after the impact. Immunohistochemistry, differential gene expression, and functional outcomes (rotarod and Morris water maze testing) were assessed to gauge treatment response. Following TBI, administration of rosuvastatin 1 mg/kg was associated with the greatest improvement in functional outcomes. Rosuvastatin treatment was associated with histological evidence of reduced neuronal degeneration at 24 h post-TBI, reduced microgliosis at day 7 post-TBI, and preserved neuronal density in the CA3 region at 35 days post-injury. Administration of rosuvastatin following TBI was also associated with downregulation of inflammatory gene expression in the brain. Following ICH, treatment with simvastatin 1 mg/kg was associated with the greatest improvement in functional outcomes, an effect that was independent of hemorrhage volume. Clinically relevant models of acute brain injury may be used to define variables such as optimal statin and dosing paradigms to facilitate the rational design of pilot clinical trials.
Collapse
Affiliation(s)
- Fransisca Indraswari
- Multidisciplinary Neuroprotection Laboratories, Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Chiba S, Takada E, Tadokoro M, Taniguchi T, Kadoyama K, Takenokuchi M, Kato S, Suzuki N. Loss of dopaminoreceptive neuron causes L-dopa resistant parkinsonism in tauopathy. Neurobiol Aging 2011; 33:2491-505. [PMID: 22169201 DOI: 10.1016/j.neurobiolaging.2011.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 09/16/2011] [Accepted: 11/03/2011] [Indexed: 10/14/2022]
Abstract
Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) is a family of inherited dementias caused by tauopathy. A mutation in exon 10 of the tau gene, N279K, causes a particular kindred of FTDP-17, which is predominant for parkinsonism. The disease initially presents as L-dopa resistant parkinsonism which then rapidly progresses. The final pathological features reveal disappearing dopamine (DA) neurons, but the causes remain poorly understood. We previously established a transgenic mouse with human N279K mutant tau as a model for FTDP-17, which showed cognitive dysfunctions caused by the mutant. Here we analyze L-dopa resistant parkinsonism by several behavioral tests, and focus on the distributions and accumulations of the mutant tau in the DA system by immunohistochemistry and Western blot. Interestingly, dopaminoreceptive (DAr) neurons in the striatum showed neurofibrils degeneration and apoptosis through caspase-3 activation by mutant tau accumulation. The DAr neuron loss in the caudoputamen, the target of the nigrostriatal system occurred before DA neuron loss in young symptomatic mice. Residual DA neurons in the mouse functioned in DA transportation, whereas dysregulation of intracellular DA compartmentalization implied an excess level of DA caused by DAr neuron loss. In the final stages, both DAr and DA neurons decreased equally, unlike Parkinson's disease. Therefore, DAr neurons were fundamentally vulnerable to the mutation indicating a critical role for the L-dopa resistant parkinsonism in tauopathy.
Collapse
Affiliation(s)
- Shunmei Chiba
- Department of Pathology and Cell Biology, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ray P, Kar A, Fushimi K, Havlioglu N, Chen X, Wu JY. PSF suppresses tau exon 10 inclusion by interacting with a stem-loop structure downstream of exon 10. J Mol Neurosci 2011; 45:453-66. [PMID: 21881826 DOI: 10.1007/s12031-011-9634-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 08/17/2011] [Indexed: 01/24/2023]
Abstract
Microtubule binding protein Tau has been implicated in a wide range of neurodegenerative disorders collectively classified as tauopathies. Exon 10 of the human tau gene, which codes for a microtubule binding repeat region, is alternatively spliced to form Tau protein isoforms containing either four or three microtubule binding repeats, Tau4R and Tau3R, respectively. The levels of different Tau splicing isoforms are fine-tuned by alternative splicing with the ratio of Tau4R/Tau3R maintained approximately at one in adult neurons. Mutations that disrupt tau exon 10 splicing regulation cause an imbalance of different tau splicing isoforms and have been associated with tauopathy. To search for factors interacting with tau pre-messenger RNA (pre-mRNA) and regulating tau exon 10 alternative splicing, we performed a yeast RNA-protein interaction screen and identified polypyrimidine tract binding protein associated splicing factor (PSF) as a candidate tau exon 10 splicing regulator. UV crosslinking experiments show that PSF binds to the stem-loop structure at the 5' splice site downstream of tau exon 10. This PSF-interacting RNA element is distinct from known PSF binding sites previously identified in other genes. Overexpression of PSF promotes tau exon 10 exclusion, whereas down-regulation of the endogenous PSF facilitates exon 10 inclusion. Immunostaining shows that PSF is expressed in the human brain regions affected by tauopathy. Our data reveal a new player in tau exon 10 alternative splicing regulation and uncover a previously unknown mechanism of PSF in regulating tau pre-mRNA splicing.
Collapse
Affiliation(s)
- Payal Ray
- Department of Neurology, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
38
|
Moderate Environmental Enrichment Mitigates Tauopathy in a Neurofibrillary Tangle Mouse Model. J Neuropathol Exp Neurol 2011; 70:610-21. [DOI: 10.1097/nen.0b013e318221bfab] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
39
|
Ittner A, Ke YD, Eersel JV, Gladbach A, Götz J, Ittner LM. Brief update on different roles of tau in neurodegeneration. IUBMB Life 2011; 63:495-502. [DOI: 10.1002/iub.467] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Differential regional distribution of phosphorylated tau and synapse loss in the nucleus accumbens in tauopathy model mice. Neurobiol Dis 2011; 42:404-14. [DOI: 10.1016/j.nbd.2011.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 01/29/2011] [Accepted: 02/03/2011] [Indexed: 11/22/2022] Open
|
41
|
RNA helicase p68 (DDX5) regulates tau exon 10 splicing by modulating a stem-loop structure at the 5' splice site. Mol Cell Biol 2011; 31:1812-21. [PMID: 21343338 DOI: 10.1128/mcb.01149-10] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Regulation of tau exon 10 splicing plays an important role in tauopathy. One of the cis elements regulating tau alternative splicing is a stem-loop structure at the 5' splice site of tau exon 10. The RNA helicase(s) modulating this stem-loop structure was unknown. We searched for splicing regulators interacting with this stem-loop region using an RNA affinity pulldown-coupled mass spectrometry approach and identified DDX5/RNA helicase p68 as an activator of tau exon 10 splicing. The activity of p68 in stimulating tau exon 10 inclusion is dependent on RBM4, an intronic splicing activator. RNase H cleavage and U1 protection assays suggest that p68 promotes conformational change of the stem-loop structure, thereby increasing the access of U1snRNP to the 5' splice site of tau exon 10. This study reports the first RNA helicase interacting with a stem-loop structure at the splice site and regulating alternative splicing in a helicase-dependent manner. Our work uncovers a previously unknown function of p68 in regulating tau exon 10 splicing. Furthermore, our experiments reveal functional interaction between two splicing activators for tau exon 10, p68 binding at the stem-loop region and RBM4 interacting with the intronic splicing enhancer region.
Collapse
|
42
|
Abstract
AbstractThe H1 haplotype clade of the tau gene (MAPT) is associated with increased risk of the sporadic disorders, progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) and to a lesser extent, Parkinson’s disease (PD). The H1c sub-haplotype drives this association in PSP and CBD, and is also weakly associated with Alzheimer’s disease (AD), suggesting involvement in common pathogenic pathway(s). The rs242557 single-nucleotide polymorphism (SNP) that defines H1c resides in a highly conserved repressor domain in the MAPT promoter. Previously, in cellular reporter assays, we showed significant rs242557 allele-specific differences in transcriptional repression, with the H1c-specific rs242557/A allele contributing a significantly higher MAPT promoter activity compared to the non-H1c rs242557/G allele. With evidence of allele-specific differences in protein binding to this repressor domain, we set out to identify those proteins that bind to this region. Electrophoretic mobility shift assay (EMSA) analysis strongly suggested allele-specific differences in protein affinities. In order to identify nuclear proteins that differentially bind to this repressor domain, we carried out a promoter-trap assay and analysed the bound proteins by SDS-PAGE and HPLC ESI-QTOF mass spectrometry. We identified 37 proteins and used bioinformatic tools such as STRING and Reactome to analyse and stratify the results. These included U2AF65, hnRNPU, PTBP1, hnRNPD0, U5 snRNP 116, ALY, HMGB2, H1 and actin and provide the basis for further studies of the role of the MAPT repressor domain and the binding proteins in regulating MAPT gene transcription and splicing.
Collapse
|
43
|
Laskowitz DT, Song P, Wang H, Mace B, Sullivan PM, Vitek MP, Dawson HN. Traumatic Brain Injury Exacerbates Neurodegenerative Pathology: Improvement with an Apolipoprotein E-Based Therapeutic. J Neurotrauma 2010; 27:1983-95. [DOI: 10.1089/neu.2010.1396] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Daniel T. Laskowitz
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Pingping Song
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Haichen Wang
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Brian Mace
- Department of Medicine (Geriatrics), Duke University Medical Center, Durham, North Carolina
| | - Patrick M. Sullivan
- Department of Medicine (Geriatrics), Duke University Medical Center, Durham, North Carolina
| | - Michael P. Vitek
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
- Cognosci, Inc., Research Triangle Park, North Carolina
| | - Hana N. Dawson
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
44
|
Kimura T, Fukuda T, Sahara N, Yamashita S, Murayama M, Mizoroki T, Yoshiike Y, Lee B, Sotiropoulos I, Maeda S, Takashima A. Aggregation of detergent-insoluble tau is involved in neuronal loss but not in synaptic loss. J Biol Chem 2010; 285:38692-9. [PMID: 20921222 DOI: 10.1074/jbc.m110.136630] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurofibrillary tangles (NFTs), which consist of highly phosphorylated tau, are hallmarks of neurodegenerative diseases including Alzheimer disease (AD). In neurodegenerative diseases, neuronal dysfunction due to neuronal loss and synaptic loss accompanies NFT formation, suggesting that a process associated with NFT formation may be involved in neuronal dysfunction. To clarify the relationship between the tau aggregation process and synapse and neuronal loss, we compared two lines of mice expressing human tau with or without an aggregation-prone P301L mutation. P301L tau transgenic (Tg) mice exhibited neuronal loss and produced sarcosyl-insoluble tau in old age but did not exhibit synaptic loss and memory impairment. By contrast, wild-type tau Tg mice neither exhibited neuronal loss nor produced sarcosyl-insoluble tau but did exhibit synaptic loss and memory impairment. Moreover, P301L tau was less phosphorylated than wild-type tau, suggesting that the tau phosphorylation state is involved in synaptic loss, whereas the tau aggregation state is involved in neuronal loss. Finally, increasing concentrations of insoluble tau aggregates leads to the formation of fibrillar tau, which causes NFTs to form.
Collapse
Affiliation(s)
- Tetsuya Kimura
- Laboratory for Alzheimer's Disease, Brain Science Institute, Riken, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Loss of tau elicits axonal degeneration in a mouse model of Alzheimer's disease. Neuroscience 2010; 169:516-31. [PMID: 20434528 DOI: 10.1016/j.neuroscience.2010.04.037] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 04/01/2010] [Accepted: 04/16/2010] [Indexed: 01/20/2023]
Abstract
A central issue in the pathogenesis of tauopathy is the question of how tau protein dysfunction leads to neurodegeneration. We have previously demonstrated that the absence of tau protein is associated with destabilization of microtubules and impaired neurite outgrowth (Dawson et al., 2001; Rapoport et al., 2002). We now hypothesize that the absence of functional tau protein may render the central nervous system more vulnerable to secondary insults such as the overexpression of mutated beta amyloid precursor protein (APP) and traumatic brain injury. We therefore crossed tau knockout mice (Dawson et al., 2001) to mice overexpressing a mutated human APP (APP(670,671), A(sw)) (Hsiao et al., 1996) and created a mouse model (A(sw)/mTau(-/-)) that provides evidence that the loss of tau function causes degeneration of neuronal processes. The overexpression of APP(670,671) in tau knockout mice, elicits the extensive formation of axonal spheroids. While spheroids are only found associated with Abeta plaques in mice expressing APP(670,671) on an endogenous mouse tau background (Irizarry et al., 1997), A(sw)/mTau(-/-) mice have spheroids not only surrounding Abeta plaques but also in white matter tracks and in the neuropil. Plaque associated and neuropil dystrophic neurites and spheroids are prominent features of Alzheimer's disease (Masliah et al., 1993; Terry, 1996; Stokin et al., 2005), and our current data suggests that loss of tau function may lead to neurodegeneration.
Collapse
|
46
|
Abstract
In dementia research, animal models have become indispensable tools. They not only model aspects of the human condition, but also simulate processes that occur in humans and hence provide insight into how disease is initiated and propagated. The present review discusses two prominent human neurodegenerative disorders, Alzheimer's disease and frontotemporal dementia. It discusses what we would like to model in animals and highlights some of the more recent achievements using species as diverse as mice, fish, flies and worms. Advances in imaging and therapy are explored. We also discuss some anticipated new models and developments. These will reveal how key players in the pathogenesis of Alzheimer's disease and frontotemporal dementia, such as the peptide Aβ (amyloid β) and the protein tau, cause neuronal dysfunction and eventually, neuronal demise. Understanding these processes fully will lead to early diagnosis and therapy.
Collapse
|
47
|
Korenova M, Stozicka Z. Improved behavioral response as a valid biomarker for drug screening program in transgenic rodent models of tauopathies. Cell Mol Neurobiol 2009; 29:937-44. [PMID: 19283467 PMCID: PMC11505800 DOI: 10.1007/s10571-009-9378-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 02/24/2009] [Indexed: 11/25/2022]
Abstract
Neurodegenerative tauopathies are defined as a group of dementia and movement disorders characterized by prominent filamentous tau inclusions and degeneration located within certain brain regions. Their common sign is a presence of proteinaceous aggregates composed of hyperphosphorylated and truncated tau proteins. The molecular mechanisms of the disease still remain unresolved, therefore transgenic organisms displaying tau-related neurodegenerative cascade have been created to allow decoding of individual pathways involved in human pathological conditions. Moreover, use of transgenic model organisms enables the application of potential therapeutic approaches. The expression of mutated or misfolded tau as a transgene in vivo leads to significant alteration of neurobehavioral features of experimental animal, therefore detailed classification of behavioral phenotype become one of the first crucial analyses, while it functionally correlates with central nervous system impairment. Currently, two major types of behavioral impairment have been described in transgenic rodent models of tauopathies, (1) progressive motor impairment associated with muscular weakness and premature death and (2) age-related impairment of cognitive functions attended with unaffected motor status. Up to the present, only transgenic models displaying motor impairment were successfully applied into the drug trials targeting misfolded tau protein, despite their behavioral inconsistence with clinical profile of progressive human tauopathy. The aim of this study was, therefore, to summarize the pros and cons of used transgenic rodent models mimicking human tauopathies in connection with development of therapeutic strategies.
Collapse
Affiliation(s)
- Miroslava Korenova
- Institute of Neuroimmunology, Slovak Academy of Sciences, AD Centre, Bratislava 845 10, Slovak Republic.
| | | |
Collapse
|
48
|
Ludolph AC, Kassubek J, Landwehrmeyer BG, Mandelkow E, Mandelkow EM, Burn DJ, Caparros-Lefebvre D, Frey KA, de Yebenes JG, Gasser T, Heutink P, Höglinger G, Jamrozik Z, Jellinger KA, Kazantsev A, Kretzschmar H, Lang AE, Litvan I, Lucas JJ, McGeer PL, Melquist S, Oertel W, Otto M, Paviour D, Reum T, Saint-Raymond A, Steele JC, Tolnay M, Tumani H, van Swieten JC, Vanier MT, Vonsattel JP, Wagner S, Wszolek ZK. Tauopathies with parkinsonism: clinical spectrum, neuropathologic basis, biological markers, and treatment options. Eur J Neurol 2009; 16:297-309. [PMID: 19364361 DOI: 10.1111/j.1468-1331.2008.02513.x] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tauopathies with parkinsonism represent a spectrum of disease entities unified by the pathologic accumulation of hyperphosphorylated tau protein fragments within the central nervous system. These pathologic characteristics suggest shared pathogenetic pathways and possible molecular targets for disease-modifying therapeutic interventions. Natural history studies, for instance, in progressive supranuclear palsy, frontotemporal dementia with parkinsonism linked to chromosome 17, corticobasal degeneration, and Niemann-Pick disease type C as well as in amyotrophic lateral sclerosis/Parkinson-dementia complex permit clinical characterization of the disease phenotypes and are crucial to the development and validation of biological markers for differential diagnostics and disease monitoring, for example, by use of neuroimaging or proteomic approaches. The wide pathologic and clinical spectrum of the tauopathies with parkinsonism is reviewed in this article, and perspectives on future advances in the understanding of the pathogenesis are given, together with potential therapeutic strategies.
Collapse
Affiliation(s)
- A C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zilka N, Korenova M, Novak M. Misfolded tau protein and disease modifying pathways in transgenic rodent models of human tauopathies. Acta Neuropathol 2009; 118:71-86. [PMID: 19238406 DOI: 10.1007/s00401-009-0499-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 02/05/2009] [Accepted: 02/09/2009] [Indexed: 12/24/2022]
Abstract
Human tauopathies represent a heterogeneous group of neurodegenerative disorders such as Alzheimer's disease (AD) that are characterized by the presence of intracellular accumulations of abnormal filaments of protein tau. Presently, AD poses an increasing public health concern, because it affects nearly 2% of the population in industrialized countries and the number of patients is expected to increase threefold within the next 50 years. Therefore, the identification of disease modifying pathways that will lead to the development of novel therapeutic approaches targeting downstream molecular events of the tauopathy is of paramount importance. In order to identify factors that may exacerbate or inhibit the disease phenotype a number of genetically modified rodent models reproducing key clinical, histopathological and molecular hallmarks of human tauopathies were developed. Current tau transgenic rodent models express as a transgene either an individual or all six human wild-type tau isoforms, mutant tau linked to FTDP-17, or structurally modified tau species derived from AD. In this review we will provide an up-to-date account of various facets of the tau neurodegenerative cascade with a special emphasis on the evolution of neurofibrillary tangles, neuronal death and neuroinflammation.
Collapse
Affiliation(s)
- Norbert Zilka
- Centre of Excellence for Alzheimer's Disease and Related Disorders, Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska 9, 84510 Bratislava, Slovak Republic
| | | | | |
Collapse
|
50
|
Ludolph AC, Kassubek J, Landwehrmeyer BG, Mandelkow E, Mandelkow EM, Burn DJ, Caparros-Lefebvre D, Frey KA, de Yebenes JG, Gasser T, Heutink P, Höglinger G, Jamrozik Z, Jellinger KA, Kazantsev A, Kretzschmar H, Lang AE, Litvan I, Lucas JJ, McGeer PL, Melquist S, Oertel W, Otto M, Paviour D, Reum T, Saint-Raymond A, Steele JC, Tolnay M, Tumani H, van Swieten JC, Vanier MT, Vonsattel JP, Wagner S, Wszolek ZK. Tauopathies with parkinsonism: clinical spectrum, neuropathologic basis, biological markers, and treatment options. Eur J Neurol 2009. [PMID: 19364361 DOI: 10.1111/j.l468-1331.2008.02513.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tauopathies with parkinsonism represent a spectrum of disease entities unified by the pathologic accumulation of hyperphosphorylated tau protein fragments within the central nervous system. These pathologic characteristics suggest shared pathogenetic pathways and possible molecular targets for disease-modifying therapeutic interventions. Natural history studies, for instance, in progressive supranuclear palsy, frontotemporal dementia with parkinsonism linked to chromosome 17, corticobasal degeneration, and Niemann-Pick disease type C as well as in amyotrophic lateral sclerosis/Parkinson-dementia complex permit clinical characterization of the disease phenotypes and are crucial to the development and validation of biological markers for differential diagnostics and disease monitoring, for example, by use of neuroimaging or proteomic approaches. The wide pathologic and clinical spectrum of the tauopathies with parkinsonism is reviewed in this article, and perspectives on future advances in the understanding of the pathogenesis are given, together with potential therapeutic strategies.
Collapse
Affiliation(s)
- A C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|