1
|
Szegvari E, Holec SAM, Woerman AL. Limitations and Applications of Rodent Models in Tauopathy and Synucleinopathy Research. J Neurochem 2025; 169:e70021. [PMID: 40026260 PMCID: PMC11874209 DOI: 10.1111/jnc.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/01/2025] [Accepted: 02/08/2025] [Indexed: 03/05/2025]
Abstract
Rodent models that accurately recapitulate key aspects of human disease have long been fundamental to the successful development of clinical interventions. This is greatly underscored in the neurodegenerative disease field, where preclinical testing of anti-prion therapeutics against rodent-adapted prions resulted in the development of small molecules effective against rodent-adapted prions but not against human prions. These findings provided critical lessons for ongoing efforts to develop treatments for patients with neurodegenerative diseases caused by misfolding and accumulation of the proteins tau and α-synuclein, or tauopathies and synucleinopathies, respectively. To avoid the potential pitfalls previously identified in the prion field, this review focuses on rodent models currently available to study tau and α-synuclein disease pathogenesis, emphasizing the strengths and limitations of each with the particular goal of better supporting preclinical research.
Collapse
Affiliation(s)
- Emma Szegvari
- Department of Microbiology, Immunology, & Pathology and Prion Research CenterColorado State UniversityFort CollinsColoradoUSA
| | - Sara A. M. Holec
- Department of Microbiology, Immunology, & Pathology and Prion Research CenterColorado State UniversityFort CollinsColoradoUSA
| | - Amanda L. Woerman
- Department of Microbiology, Immunology, & Pathology and Prion Research CenterColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
2
|
Yao Y, Muench M, Alle T, Zhang B, Lucero B, Perez‐Tremble R, McGrosso D, Newman M, Gonzalez DJ, Lee VM, Ballatore C, Brunden KR. A small-molecule microtubule-stabilizing agent safely reduces Aβ plaque and tau pathology in transgenic mouse models of Alzheimer's disease. Alzheimers Dement 2024; 20:4540-4558. [PMID: 38884283 PMCID: PMC11247666 DOI: 10.1002/alz.13875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Intraneuronal inclusions composed of tau protein are found in Alzheimer's disease (AD) and other tauopathies. Tau normally binds microtubules (MTs), and its disengagement from MTs and misfolding in AD is thought to result in MT abnormalities. We previously identified triazolopyrimidine-containing MT-stabilizing compounds that provided benefit in AD mouse models and herein describe the characterization and efficacy testing of an optimized candidate, CNDR-51997. METHODS CNDR-51997 underwent pharmacokinetic, pharmacodynamic, safety pharmacology, and mouse tolerability testing. In addition, the compound was examined for efficacy in 5XFAD amyloid beta (Aβ) plaque mice and PS19 tauopathy mice. RESULTS CNDR-51997 significantly reduced Aβ plaques in 5XFAD mice and tau pathology in PS19 mice, with the latter also showing attenuated axonal dystrophy and gliosis. CNDR-51997 was well tolerated at doses that exceeded efficacy doses, with a good safety pharmacology profile. DISCUSSION CNDR-51997 may be a candidate for advancement as a potential therapeutic agent for AD and/or other tauopathies. Highlights There is evidence of microtubule alterations (MT) in Alzheimer's disease (AD) brain and in mouse models of AD pathology. Intermittent dosing with an optimized, brain-penetrant MT-stabilizing small-molecule, CNDR-51997, reduced both Aβ plaque and tau inclusion pathology in established mouse models of AD. CNDR-51997 attenuated axonal dystrophy and gliosis in a tauopathy mouse model, with a strong trend toward reduced hippocampal neuron loss. CNDR-51997 is well tolerated in mice at doses that are meaningfully greater than required for efficacy in AD mouse models, and the compound has a good safety pharmacology profile.
Collapse
Affiliation(s)
- Yuemang Yao
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Megan Muench
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Thibault Alle
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Bin Zhang
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Bobby Lucero
- Department of Chemistry and BiochemistryUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Roxanne Perez‐Tremble
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Dominic McGrosso
- Department of PharmacologyUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Mira Newman
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David J. Gonzalez
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
- Department of PharmacologyUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Virginia M.‐Y. Lee
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Carlo Ballatore
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Kurt R. Brunden
- Center for Neurodegenerative Disease ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
3
|
Kauwe G, Pareja-Navarro KA, Yao L, Chen JH, Wong I, Saloner R, Cifuentes H, Nana AL, Shah S, Li Y, Le D, Spina S, Grinberg LT, Seeley WW, Kramer JH, Sacktor TC, Schilling B, Gan L, Casaletto KB, Tracy TE. KIBRA repairs synaptic plasticity and promotes resilience to tauopathy-related memory loss. J Clin Invest 2024; 134:e169064. [PMID: 38299587 PMCID: PMC10836803 DOI: 10.1172/jci169064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/21/2023] [Indexed: 02/02/2024] Open
Abstract
Synaptic plasticity is obstructed by pathogenic tau in the brain, representing a key mechanism that underlies memory loss in Alzheimer's disease (AD) and related tauopathies. Here, we found that reduced levels of the memory-associated protein KIdney/BRAin (KIBRA) in the brain and increased KIBRA protein levels in cerebrospinal fluid are associated with cognitive impairment and pathological tau levels in disease. We next defined a mechanism for plasticity repair in vulnerable neurons using the C-terminus of the KIBRA protein (CT-KIBRA). We showed that CT-KIBRA restored plasticity and memory in transgenic mice expressing pathogenic human tau; however, CT-KIBRA did not alter tau levels or prevent tau-induced synapse loss. Instead, we found that CT-KIBRA stabilized the protein kinase Mζ (PKMζ) to maintain synaptic plasticity and memory despite tau-mediated pathogenesis. Thus, our results distinguished KIBRA both as a biomarker of synapse dysfunction and as the foundation for a synapse repair mechanism to reverse cognitive impairment in tauopathy.
Collapse
Affiliation(s)
- Grant Kauwe
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Lei Yao
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jackson H. Chen
- Buck Institute for Research on Aging, Novato, California, USA
| | - Ivy Wong
- Buck Institute for Research on Aging, Novato, California, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Helen Cifuentes
- Buck Institute for Research on Aging, Novato, California, USA
| | - Alissa L. Nana
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, California, USA
| | - Yaqiao Li
- Gladstone Institutes, San Francisco, Califoria, USA
| | - David Le
- Gladstone Institutes, San Francisco, Califoria, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Todd C. Sacktor
- The Robert F. Furchgott Center of Neural and Behavioral Science, Departments of Physiology and Pharmacology, Anesthesiology, and Neurology, State University of New York Health Sciences University, Brooklyn, New York, USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, California, USA
| |
Collapse
|
4
|
Canet G, Rocaboy E, Diego-Diàz S, Whittington RA, Julien C, Planel E. Methods for Biochemical Isolation of Insoluble Tau in Rodent Models of Tauopathies. Methods Mol Biol 2024; 2754:323-341. [PMID: 38512674 DOI: 10.1007/978-1-0716-3629-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
The intracellular accumulation of microtubule-associated protein tau is a characteristic feature of tauopathies, a group of neurodegenerative diseases including Alzheimer's disease. Formation of insoluble tau aggregates is initiated by the abnormal hyperphosphorylation and oligomerization of tau. Over the past decades, multiple transgenic rodent models mimicking tauopathies have been develop, showcasing this neuropathological hallmark. The biochemical analysis of insoluble tau in these models has served as a valuable tool to understand the progression of tau-related pathology. In this chapter, we provide a comprehensive review of the two primary methods for isolating insoluble tau, namely, sarkosyl and formic acid extraction (and their variants), which are employed for biochemical analysis in transgenic mouse models of tauopathy. We also analyze the strengths and limitations of these methods.
Collapse
Affiliation(s)
- Geoffrey Canet
- Faculté de Médecine, Département de Psychiatrie et Neurosciences, Université Laval, Quebec, QC, Canada
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Quebec, QC, Canada
| | - Emma Rocaboy
- Faculté de Médecine, Département de Psychiatrie et Neurosciences, Université Laval, Quebec, QC, Canada
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Quebec, QC, Canada
| | - Sofia Diego-Diàz
- Faculté de Médecine, Département de Psychiatrie et Neurosciences, Université Laval, Quebec, QC, Canada
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Quebec, QC, Canada
| | - Robert A Whittington
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Carl Julien
- Centre de Recherche en Sciences Animales de Deschambault, Deschambault, QC, Canada
- Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Quebec, QC, Canada
| | - Emmanuel Planel
- Faculté de Médecine, Département de Psychiatrie et Neurosciences, Université Laval, Quebec, QC, Canada.
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Quebec, QC, Canada.
| |
Collapse
|
5
|
Kauwe G, Pareja-Navarro KA, Yao L, Chen JH, Wong I, Saloner R, Cifuentes H, Nana AL, Shah S, Li Y, Le D, Spina S, Grinberg LT, Seeley WW, Kramer JH, Sacktor TC, Schilling B, Gan L, Casaletto KB, Tracy TE. KIBRA repairs synaptic plasticity and promotes resilience to tauopathy-related memory loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.543777. [PMID: 37398236 PMCID: PMC10312627 DOI: 10.1101/2023.06.12.543777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Synaptic plasticity is obstructed by pathogenic tau in the brain, representing a key mechanism that underlies memory loss in Alzheimer's disease (AD) and related tauopathies. Here, we define a mechanism for plasticity repair in vulnerable neurons using the C-terminus of the KIdney/BRAin (KIBRA) protein (CT-KIBRA). We show that CT-KIBRA restores plasticity and memory in transgenic mice expressing pathogenic human tau; however, CT-KIBRA did not alter tau levels or prevent tau-induced synapse loss. Instead, we find that CT-KIBRA binds to and stabilizes protein kinase Mζ (PKMζ) to maintain synaptic plasticity and memory despite tau-mediated pathogenesis. In humans we find that reduced KIBRA in brain and increased KIBRA in cerebrospinal fluid are associated with cognitive impairment and pathological tau levels in disease. Thus, our results distinguish KIBRA both as a novel biomarker of synapse dysfunction in AD and as the foundation for a synapse repair mechanism to reverse cognitive impairment in tauopathy.
Collapse
Affiliation(s)
- Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Lei Yao
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Ivy Wong
- Buck Institute for Research on Aging, Novato, CA USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | | | - Alissa L. Nana
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, CA USA
| | - Yaqiao Li
- Gladstone Institutes, San Francisco, CA USA
| | - David Le
- Gladstone Institutes, San Francisco, CA USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
- Weill Institute for Neurosciences, Department of Pathology, University of California, San Francisco USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
- Weill Institute for Neurosciences, Department of Pathology, University of California, San Francisco USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Todd C. Sacktor
- The Robert F. Furchgott Center of Neural and Behavioral Science, Departments of Physiology and Pharmacology, Anesthesiology, and Neurology, State University of New York Health Sciences University, Brooklyn, NY USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, CA USA
| |
Collapse
|
6
|
Han ZZ, Fleet A, Larrieu D. Can accelerated ageing models inform us on age-related tauopathies? Aging Cell 2023; 22:e13830. [PMID: 37013265 PMCID: PMC10186612 DOI: 10.1111/acel.13830] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Ageing is the greatest risk factor of late-onset neurodegenerative diseases. In the realm of sporadic tauopathies, modelling the process of biological ageing in experimental animals forms the foundation of searching for the molecular origin of pathogenic tau and developing potential therapeutic interventions. Although prior research into transgenic tau models offers valuable lessons for studying how tau mutations and overexpression can drive tau pathologies, the underlying mechanisms by which ageing leads to abnormal tau accumulation remains poorly understood. Mutations associated with human progeroid syndromes have been proposed to be able to mimic an aged environment in animal models. Here, we summarise recent attempts in modelling ageing in relation to tauopathies using animal models that carry mutations associated with human progeroid syndromes, or genetic elements unrelated to human progeroid syndromes, or have exceptional natural lifespans, or a remarkable resistance to ageing-related disorders.
Collapse
Affiliation(s)
- Zhuang Zhuang Han
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| | - Alex Fleet
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| | - Delphine Larrieu
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| |
Collapse
|
7
|
Nishioka C, Liang HF, Ong S, Sun SW. Axonal transport impairment and its relationship with diffusion tensor imaging metrics of a murine model of p301L tau induced tauopathy. Neuroscience 2022; 498:144-154. [PMID: 35753531 DOI: 10.1016/j.neuroscience.2022.06.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/15/2022] [Indexed: 11/15/2022]
Abstract
Diffusion Tensor Imaging (DTI) and Manganese Enhanced MRI (MEMRI) are noninvasive tools to characterize neural fiber microstructure and axonal transport. A combination of both may provide novel insights into the progress of neurodegeneration. To investigate the relationship of DTI and MEMRI in white matter of tauopathy, twelve optic nerves of 11-month-old p301L tau mice were imaged and finished with postmortem immunohistochemistry. MEMRI was used to quantify Mn2+ accumulation rates in the optic nerve (ON, termed ONAR) and the Superior Colliculus (SC, termed SCAR), the primary terminal site of ON in mice. We found that both ONAR and SCAR revealed a significant linear correlation with mean diffusion (mD) and radial diffusion (rD) but not with other DTI quantities. Immunohistochemistry findings showed that ONAR, mD, and rD are significantly correlated with the myelin content (Myelin Basic Protein, p < 0.05) but not with the axonal density (SMI-31), tubulin density, or tau aggregates (AT8 staining). In summary, slower axonal transport appeared to have less myelinated axons and thinner remaining axons, associated with reduced rD and mD of in vivo DTI. A combination of in vivo MEMRI and DTI can provide critical information to delineate the progress of white matter deficits in neurodegenerative diseases.
Collapse
Affiliation(s)
- Christopher Nishioka
- Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States; Neuroscience Graduate Program, University of California, Riverside, CA, United States
| | - Hsiao-Fang Liang
- Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Stephen Ong
- Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States; Robert Wood Johnson Barnabas Health (RWJBH) and Rutgers University, United States
| | - Shu-Wei Sun
- Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States; Neuroscience Graduate Program, University of California, Riverside, CA, United States.
| |
Collapse
|
8
|
Song L, Oseid DE, Wells EA, Coaston T, Robinson AS. Heparan Sulfate Proteoglycans (HSPGs) Serve as the Mediator Between Monomeric Tau and Its Subsequent Intracellular ERK1/2 Pathway Activation. J Mol Neurosci 2022; 72:772-791. [PMID: 35040015 PMCID: PMC8763444 DOI: 10.1007/s12031-021-01943-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/06/2021] [Indexed: 12/15/2022]
Abstract
The conversion of soluble tau protein to insoluble, hyperphosphorylated neurofibrillary tangles (NFTs) is a major hallmark leading to neuronal death observed in neurodegenerative tauopathies. Unlike NFTs, the involvement of monomeric tau in the progression of tau pathology has been less investigated. Using live-cell confocal microscopy and flow cytometry, we demonstrate that soluble 0N4R monomers were rapidly endocytosed by SH-SY5Y and C6 glioma cells via actin-dependent macropinocytosis. Further, cellular endocytosis of monomeric tau has been demonstrated to be HSPG-dependent, as shown in C6 glial cells with genetic knockouts of xylosyltransferase-1-a key enzyme in HSPG synthesis-with a reduced level of tau uptake. Tau internalization subsequently triggers ERK1/2 activation and therefore, the upregulation of IL-6 and IL-1β. The role of ERK1/2 in regulating the levels of pro-inflammatory gene transcripts was confirmed by inhibiting the MEK-ERK1/2 signaling pathway, which led to the attenuated IL-6 and IL-1β expressions but not that of TNF-α. Moreover, as a key regulator of tau internalization, LRP1 (low-density lipoprotein receptor-related protein 1) levels were downregulated in response to monomeric tau added to C6 cells, while it was upregulated in HSPG-deficient cells, suggesting that the involvement of LRP1 in tau uptake depends on the presence of HSPGs on the cell surface. The subsequent LRP1 knockdown experiment we performed shows that LRP1 deficiency leads to an attenuated propensity for tau uptake and further elevated IL-6 gene expression. Collectively, our data suggest that tau has multiple extracellular binding partners that mediate its internalization through distinct mechanisms. Additionally, this study demonstrates the important role of both HSPGs and LRP1 in regulating cellular immune responses to tau protein monomers, providing a novel target for alleviating the neuroinflammatory environment before the formation of neurofibrillary tangles.
Collapse
Affiliation(s)
- Liqing Song
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Daniel E Oseid
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA
| | - Evan A Wells
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Troy Coaston
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
9
|
Shah SH, Schiapparelli LM, Ma Y, Yokota S, Atkins M, Xia X, Cameron EG, Huang T, Saturday S, Sun CB, Knasel C, Blackshaw S, Yates Iii JR, Cline HT, Goldberg JL. Quantitative transportomics identifies Kif5a as a major regulator of neurodegeneration. eLife 2022; 11:68148. [PMID: 35259089 PMCID: PMC8947766 DOI: 10.7554/elife.68148] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/07/2022] [Indexed: 11/29/2022] Open
Abstract
Many neurons in the adult central nervous system, including retinal ganglion cells (RGCs), degenerate and die after injury. Early axon protein and organelle trafficking failure is a key component in many neurodegenerative disorders yet changes to axoplasmic transport in disease models have not been quantified. We analyzed early changes in the protein ‘transportome’ from RGC somas to their axons after optic nerve injury and identified transport failure of an anterograde motor protein Kif5a early in RGC degeneration. We demonstrated that manipulating Kif5a expression affects anterograde mitochondrial trafficking in RGCs and characterized axon transport in Kif5a knockout mice to identify proteins whose axon localization was Kif5a-dependent. Finally, we found that knockout of Kif5a in RGCs resulted in progressive RGC degeneration in the absence of injury. Together with expression data localizing Kif5a to human RGCs, these data identify Kif5a transport failure as a cause of RGC neurodegeneration and point to a mechanism for future therapeutics.
Collapse
Affiliation(s)
- Sahil H Shah
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | | | - Yuanhui Ma
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Satoshi Yokota
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Melissa Atkins
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Xin Xia
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Evan G Cameron
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Thanh Huang
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Sarah Saturday
- Neuroscience Department, The Scripps Research Institute, La Jolla, United States
| | - Catalin B Sun
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Cara Knasel
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - John R Yates Iii
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Hollis T Cline
- Neuroscience Department, The Scripps Research Institute, La Jolla, United States
| | - Jeffrey L Goldberg
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| |
Collapse
|
10
|
Ghosh A, Singh S. Regulation Of Microtubule: Current Concepts And Relevance To Neurodegenerative Diseases. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:656-679. [PMID: 34323203 DOI: 10.2174/1871527320666210728144043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 02/23/2021] [Indexed: 11/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are abnormalities linked to neuronal structure and irregularities associated with the proliferation of cells, transportation, and differentiation. NDD also involves synaptic circuitry and neural network alterations known as synaptopathies. Microtubules (MTs) and MTs-associated proteins help to maintain neuronal health as well as their development. The microtubular dynamic structure plays a crucial role in the division of cells and forms mitotic spindles, thus take part in initiating stages of differentiation and polarization for various types of cells. The MTs also take part in the cellular death but MT-based cellular degenerations are not yet well excavated. In the last few years, studies have provided the protagonist activity of MTs in neuronal degeneration. In this review, we largely engrossed our discussion on the change of MT cytoskeleton structure, describing their organization, dynamics, transportation, and their failure causing NDDs. At end of this review, we are targeting the therapeutic neuroprotective strategies on clinical priority and also try to discuss the clues for the development of new MT-based therapy as a new pharmacological intervention. This will be a new potential site to block not only neurodegeneration but also promotes the regeneration of neurons.
Collapse
Affiliation(s)
- Anirban Ghosh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| |
Collapse
|
11
|
Axonal TAU Sorting Requires the C-terminus of TAU but is Independent of ANKG and TRIM46 Enrichment at the AIS. Neuroscience 2021; 461:155-171. [PMID: 33556457 DOI: 10.1016/j.neuroscience.2021.01.041] [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: 09/25/2020] [Revised: 01/26/2021] [Accepted: 01/31/2021] [Indexed: 01/06/2023]
Abstract
Somatodendritic missorting of the axonal protein TAU is a hallmark of Alzheimer's disease and related tauopathies. Rodent primary neurons and iPSC-derived neurons are used for studying mechanisms of neuronal polarity, including TAU trafficking. However, these models are expensive, time-consuming, and/or require the killing of animals. In this study, we tested four differentiation procedures to generate mature neuron cultures from human SH-SY5Y neuroblastoma cells and assessed the TAU sorting capacity. We show that SH-SY5Y-derived neurons, differentiated with sequential RA/BDNF treatment, are suitable for investigating axonal TAU sorting. These human neurons show pronounced neuronal polarity, axodendritic outgrowth, expression of the neuronal maturation markers TAU and MAP2, and, importantly, efficient axonal sorting of endogenous and transfected human wild-type TAU, similar to mouse primary neurons. We demonstrate that the N-terminal half of TAU is not sufficient for axonal targeting, as a C-terminus-lacking construct (N-term-TAUHA) is not axonally enriched in both neuronal cell models. Importantly, SH-SY5Y-derived neurons do not show the formation of a classical axon initial segment (AIS), indicated by the lack of ankyrin G (ANKG) and tripartite motif-containing protein 46 (TRIM46) at the proximal axon, which suggests that successful axonal TAU sorting is independent of classical AIS formation. Taken together, our results provide evidence that (i) SH-SY5Y-derived neurons are a valuable human neuronal cell model for studying TAU sorting readily accessible at low cost and without animal need, and that (ii) efficient axonal TAU targeting is independent of ANKG or TRIM46 enrichment at the proximal axon in these neurons.
Collapse
|
12
|
Zareba-Paslawska J, Patra K, Kluzer L, Revesz T, Svenningsson P. Tau Isoform-Driven CBD Pathology Transmission in Oligodendrocytes in Humanized Tau Mice. Front Neurol 2021; 11:589471. [PMID: 33519674 PMCID: PMC7845573 DOI: 10.3389/fneur.2020.589471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
The aggregation of abnormally phosphorylated tau protein in neurons and glia is a neuropathological hallmark of several neurodegenerative disorders, collectively known as tauopathies. They are further subclassified based on the preferential pathological aggregation of three carboxyl-terminal repeat domains (3R) and/or 4R tau. Corticobasal degeneration (CBD) is a rare neurodegenerative disorder classified as a 4R tauopathy. In the present study, we extend analysis of CBD-tau cell-type specific pathology transmission with 3R and 4R tau isoform distinguishable changes. We use a humanized tau (hTau) mouse line, which overexpress all six human tau isoforms in a murine tau knockout background and perform intrastriatal inoculation of control and CBD-tau enriched human brain homogenate. We show that CBD-tau causes hyperphosphorylation of tau at Ser202 predominantly in oligodendrocytes. Next, we demonstrate the spread of tau pathology from striatum to the overlaying corpus callosum and further to the contralateral side. Finally, we demonstrate that the almost exclusive oligodendrocyte-based transmission of hyperphosphorylated tau is reflected in the endogenous 4R tau isoform expression and corresponds to subclassification of CBD as a 4R tauopathy. Additionally, we identify functional changes in oligodendrocytes reflected by myelin basic protein abnormalities upon CBD-tau inoculation. These changes are not observed in murine tau knockout mice lacking both human and murine tau. Our study presents not only in vivo tau isoform–driven region- and cell-specific tau pathology, but also underlines that tau pathology seeding and transmission might be oligodendrocyte-based. These results, which need to be extended to more cases, give new insights into why tauopathies might vary greatly in both histopathological and neuroanatomical patterns.
Collapse
Affiliation(s)
- Justyna Zareba-Paslawska
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kalicharan Patra
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Luca Kluzer
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Tamas Revesz
- Queen Square Brain Bank, Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Per Svenningsson
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
13
|
d'Errico P, Meyer-Luehmann M. Mechanisms of Pathogenic Tau and Aβ Protein Spreading in Alzheimer's Disease. Front Aging Neurosci 2020; 12:265. [PMID: 33061903 PMCID: PMC7481386 DOI: 10.3389/fnagi.2020.00265] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is pathologically defined by extracellular accumulation of amyloid-β (Aβ) peptides generated by the cleavage of amyloid precursor protein (APP), strings of hyperphosphorylated Tau proteins accumulating inside neurons known as neurofibrillary tangles (NFTs) and neuronal loss. The association between the two hallmarks and cognitive decline has been known since the beginning of the 20th century when the first description of the disease was carried out by Alois Alzheimer. Today, more than 40 million people worldwide are affected by AD that represents the most common cause of dementia and there is still no effective treatment available to cure the disease. In general, the aggregation of Aβ is considered an essential trigger in AD pathogenesis that gives rise to NFTs, neuronal dysfunction and dementia. During the process leading to AD, tau and Aβ first misfold and form aggregates in one brain region, from where they spread to interconnected areas of the brain thereby inducing its gradual morphological and functional deterioration. In this mini-review article, we present an overview of the current literature on the spreading mechanisms of Aβ and tau pathology in AD since a more profound understanding is necessary to design therapeutic approaches aimed at preventing or halting disease progression.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Ashrafizadeh M, Zarrabi A, Najafi M, Samarghandian S, Mohammadinejad R, Ahn KS. Resveratrol targeting tau proteins, amyloid-beta aggregations, and their adverse effects: An updated review. Phytother Res 2020; 34:2867-2888. [PMID: 32491273 DOI: 10.1002/ptr.6732] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/18/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Resveratrol (Res) is a non-flavonoid compound with pharmacological actions such as antioxidant, antiinflammatory, hepatoprotective, antidiabetes, and antitumor. This plant-derived chemical has a long history usage in treatment of diseases. The excellent therapeutic impacts of Res and its capability in penetration into blood-brain barrier have made it an appropriate candidate in the treatment of neurological disorders (NDs). Tau protein aggregations and amyloid-beta (Aβ) deposits are responsible for the induction of NDs. A variety of studies have elucidated the role of these aggregations in NDs and the underlying molecular pathways in their development. In the present review, based on the recently published articles, we describe that how Res administration could inhibit amyloidogenic pathway and stimulate processes such as autophagy to degrade Aβ aggregations. Besides, we demonstrate that Res supplementation is beneficial in dephosphorylation of tau proteins and suppressing their aggregations. Then, we discuss molecular pathways and relate them to the treatment of NDs.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Turkey
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Brunello CA, Merezhko M, Uronen RL, Huttunen HJ. Mechanisms of secretion and spreading of pathological tau protein. Cell Mol Life Sci 2020; 77:1721-1744. [PMID: 31667556 PMCID: PMC7190606 DOI: 10.1007/s00018-019-03349-1] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of misfolded and aggregated forms of tau protein in the brain is a neuropathological hallmark of tauopathies, such as Alzheimer's disease and frontotemporal lobar degeneration. Tau aggregates have the ability to transfer from one cell to another and to induce templated misfolding and aggregation of healthy tau molecules in previously healthy cells, thereby propagating tau pathology across different brain areas in a prion-like manner. The molecular mechanisms involved in cell-to-cell transfer of tau aggregates are diverse, not mutually exclusive and only partially understood. Intracellular accumulation of misfolded tau induces several mechanisms that aim to reduce the cellular burden of aggregated proteins and also promote secretion of tau aggregates. However, tau may also be released from cells physiologically unrelated to protein aggregation. Tau secretion involves multiple vesicular and non-vesicle-mediated pathways, including secretion directly through the plasma membrane. Consequently, extracellular tau can be found in various forms, both as a free protein and in vesicles, such as exosomes and ectosomes. Once in the extracellular space, tau aggregates can be internalized by neighboring cells, both neurons and glial cells, via endocytic, pinocytic and phagocytic mechanisms. Importantly, accumulating evidence suggests that prion-like propagation of misfolding protein pathology could provide a general mechanism for disease progression in tauopathies and other related neurodegenerative diseases. Here, we review the recent literature on cellular mechanisms involved in cell-to-cell transfer of tau, with a particular focus in tau secretion.
Collapse
Affiliation(s)
- Cecilia A Brunello
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Riikka-Liisa Uronen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland.
| |
Collapse
|
16
|
Gao Y, He Z, He X, Zhang H, Weng J, Yang X, Meng F, Luo L, Tang BZ. Dual-Color Emissive AIEgen for Specific and Label-Free Double-Stranded DNA Recognition and Single-Nucleotide Polymorphisms Detection. J Am Chem Soc 2019; 141:20097-20106. [DOI: 10.1021/jacs.9b09239] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yuting Gao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhenyan He
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xuewen He
- Department of Chemistry and Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, People’s Republic of China
| | - Haoke Zhang
- Department of Chemistry and Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, People’s Republic of China
| | - Jun Weng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ben Zhong Tang
- Department of Chemistry and Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, People’s Republic of China
| |
Collapse
|
17
|
Kokubun H, Jin H, Aoe T. Pathogenic Effects of Impaired Retrieval between the Endoplasmic Reticulum and Golgi Complex. Int J Mol Sci 2019; 20:ijms20225614. [PMID: 31717602 PMCID: PMC6888596 DOI: 10.3390/ijms20225614] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular activities, such as growth and secretion, are dependent on correct protein folding and intracellular protein transport. Injury, like ischemia, malnutrition, and invasion of toxic substances, affect the folding environment in the endoplasmic reticulum (ER). The ER senses this information, following which cells adapt their response to varied situations through the unfolded protein response. Activation of the KDEL receptor, resulting from the secretion from the ER of chaperones containing the KDEL sequence, plays an important role in this adaptation. The KDEL receptor was initially shown to be necessary for the retention of KDEL sequence-containing proteins in the ER. However, it has become clear that the activated KDEL receptor also regulates bidirectional transport between the ER and the Golgi complex, as well as from the Golgi to the secretory pathway. In addition, it has been suggested that the signal for KDEL receptor activation may also affect several other cellular activities. In this review, we discuss KDEL receptor-mediated bidirectional transport and signaling and describe disease models and human diseases related to KDEL receptor dysfunction.
Collapse
Affiliation(s)
- Hiroshi Kokubun
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
- Correspondence: ; Tel.: +81-436-62-1211
| |
Collapse
|
18
|
Nakamura M, Shiozawa S, Tsuboi D, Amano M, Watanabe H, Maeda S, Kimura T, Yoshimatsu S, Kisa F, Karch CM, Miyasaka T, Takashima A, Sahara N, Hisanaga SI, Ikeuchi T, Kaibuchi K, Okano H. Pathological Progression Induced by the Frontotemporal Dementia-Associated R406W Tau Mutation in Patient-Derived iPSCs. Stem Cell Reports 2019; 13:684-699. [PMID: 31543469 PMCID: PMC6829766 DOI: 10.1016/j.stemcr.2019.08.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/22/2022] Open
Abstract
Mutations in the microtubule-associated protein tau (MAPT) gene are known to cause familial frontotemporal dementia (FTD). The R406W tau mutation is a unique missense mutation whose patients have been reported to exhibit Alzheimer's disease (AD)-like phenotypes rather than the more typical FTD phenotypes. In this study, we established patient-derived induced pluripotent stem cell (iPSC) models to investigate the disease pathology induced by the R406W mutation. We generated iPSCs from patients and established isogenic lines using CRISPR/Cas9. The iPSCs were induced into cerebral organoids, which were dissociated into cortical neurons with high purity. In this neuronal culture, the mutant tau protein exhibited reduced phosphorylation levels and was increasingly fragmented by calpain. Furthermore, the mutant tau protein was mislocalized and the axons of the patient-derived neurons displayed morphological and functional abnormalities, which were rescued by microtubule stabilization. The findings of our study provide mechanistic insight into tau pathology and a potential for therapeutic intervention.
Collapse
Affiliation(s)
- Mari Nakamura
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Department of Biomedical Chemistry, School of International Health, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Seiji Shiozawa
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Daisuke Tsuboi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa, Nagoya, Aichi 466-8550, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa, Nagoya, Aichi 466-8550, Japan
| | - Hirotaka Watanabe
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Sumihiro Maeda
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Taeko Kimura
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage, Chiba 266-8555, Japan
| | - Sho Yoshimatsu
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Fumihiko Kisa
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Celeste M Karch
- Department of Psychiatry and Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Tomohiro Miyasaka
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe-shi, Kyoto 610-0394, Japan
| | - Akihiko Takashima
- Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage, Chiba 266-8555, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachidori, Chuo-ku, Niigata 951-8585, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsurumai, Showa, Nagoya, Aichi 466-8550, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
19
|
Regulation of Tau Homeostasis and Toxicity by Acetylation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:47-55. [PMID: 32096027 DOI: 10.1007/978-981-32-9358-8_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiple neurodegenerative conditions including Alzheimer's disease and frontotemporal dementia are characterized by the accumulation of tau in the brain, associated with synapse loss and cognitive decline. Currently, the molecular events that lead to tau aggregation, and the pathological effects of the tau protein, are incompletely understood. Recent work has highlighted aberrant acetylation of tau as a key to understanding the pathophysiological roles of this protein. Specific acetylation sites regulate the formation of tau aggregates, synaptic signaling and long-term potentiation. Unraveling the details of this emerging story may offer novel insights into potential therapeutic approaches for devastating neurodegenerative diseases.
Collapse
|
20
|
Jiang S, Wen N, Li Z, Dube U, Del Aguila J, Budde J, Martinez R, Hsu S, Fernandez MV, Cairns NJ, Harari O, Cruchaga C, Karch CM. Integrative system biology analyses of CRISPR-edited iPSC-derived neurons and human brains reveal deficiencies of presynaptic signaling in FTLD and PSP. Transl Psychiatry 2018; 8:265. [PMID: 30546007 PMCID: PMC6293323 DOI: 10.1038/s41398-018-0319-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/13/2018] [Indexed: 01/12/2023] Open
Abstract
Mutations in the microtubule-associated protein tau (MAPT) gene cause autosomal dominant frontotemporal lobar degeneration with tau inclusions (FTLD-tau). MAPT p.R406W carriers present clinically with progressive memory loss and neuropathologically with neuronal and glial tauopathy. However, the pathogenic events triggered by the expression of the mutant tau protein remain poorly understood. To identify the genes and pathways that are dysregulated in FTLD-tau, we performed transcriptomic analyses in induced pluripotent stem cell (iPSC)-derived neurons carrying MAPT p.R406W and CRISPR/Cas9-corrected isogenic controls. We found that the expression of the MAPT p.R406W mutation was sufficient to create a significantly different transcriptomic profile compared with that of the isogeneic controls and to cause the differential expression of 328 genes. Sixty-one of these genes were also differentially expressed in the same direction between MAPT p.R406W carriers and pathology-free human control brains. We found that genes differentially expressed in the stem cell models and human brains were enriched for pathways involving gamma-aminobutyric acid (GABA) receptors and pre-synaptic function. The expression of GABA receptor genes, including GABRB2 and GABRG2, were consistently reduced in iPSC-derived neurons and brains from MAPT p.R406W carriers. Interestingly, we found that GABA receptor genes, including GABRB2 and GABRG2, are significantly lower in symptomatic mouse models of tauopathy, as well as in brains with progressive supranuclear palsy. Genome wide association analyses reveal that common variants within GABRB2 are associated with increased risk for frontotemporal dementia (P < 1 × 10-3). Thus, our systems biology approach, which leverages molecular data from stem cells, animal models, and human brain tissue can reveal novel disease mechanisms. Here, we demonstrate that MAPT p.R406W is sufficient to induce changes in GABA-mediated signaling and synaptic function, which may contribute to the pathogenesis of FTLD-tau and other primary tauopathies.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Natalie Wen
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Zeran Li
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Umber Dube
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Jorge Del Aguila
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Rita Martinez
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Simon Hsu
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Maria V Fernandez
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Nigel J Cairns
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, 660S. Euclid Ave, Campus Box 8118, Saint Louis, MO, 63110, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA.
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA.
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA.
| |
Collapse
|
21
|
Jin H, Komita M, Aoe T. Decreased Protein Quality Control Promotes the Cognitive Dysfunction Associated With Aging and Environmental Insults. Front Neurosci 2018; 12:753. [PMID: 30443201 PMCID: PMC6221900 DOI: 10.3389/fnins.2018.00753] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 10/01/2018] [Indexed: 11/29/2022] Open
Abstract
Background: Most neurodegenerative diseases are sporadic and develop with age. Degenerative neural tissues often contain intra- and extracellular protein aggregates, suggesting that the proteostasis network that combats protein misfolding could be dysfunctional in the setting of neurodegenerative disease. Binding immunoglobulin protein (BiP) is an endoplasmic reticulum (ER) chaperone that is crucial for protein folding and modulating the adaptive response in early secretory pathways. The interaction between BiP and unfolded proteins is mediated by the substrate-binding domain and nucleotide-binding domain with ATPase activity. The interaction facilitates protein folding and maturation. BiP has a recovery motif at the carboxyl terminus. The aim of this study is to examine cognitive function in model mice with an impaired proteostasis network by expressing a mutant form of BiP lacking the recovery motif. We also investigated if impairments of cognitive function were exacerbated by exposure to environmental insults, such as inhaled anesthetics. Methods: We examined cognitive function by performing radial maze testing with mutant BiP mice and assessed the additional impact of general anesthesia in the context of proteostasis dysfunction. Testing over 8 days was performed 10 weeks, 6 months, and 1 year after birth. Results: Age-related cognitive decline occurred in both forms of mice. The mutant BiP and anesthetic exposure promoted cognitive dysfunction prior to the senile period. After senescence, when mice were tested at 6 months of age and at 1 year old, there were no significant differences between the two genotypes in terms of the radial maze testing; furthermore, there was no significant difference when tested with and without anesthetic exposure. Conclusion: Our data suggest that aging was the predominant factor underlying the impairment of cognitive function in this study. Impairment of the proteostasis network may promote age-related neurodegeneration, and this is exacerbated by external insults.
Collapse
Affiliation(s)
- Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Mari Komita
- Department of Anesthesiology, Chiba Rosai Hospital, Ichihara, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara, Japan
| |
Collapse
|
22
|
Dumbacher M, Van Dooren T, Princen K, De Witte K, Farinelli M, Lievens S, Tavernier J, Dehaen W, Wera S, Winderickx J, Allasia S, Kilonda A, Spieser S, Marchand A, Chaltin P, Hoogenraad CC, Griffioen G. Modifying Rap1-signalling by targeting Pde6δ is neuroprotective in models of Alzheimer's disease. Mol Neurodegener 2018; 13:50. [PMID: 30257685 PMCID: PMC6158915 DOI: 10.1186/s13024-018-0283-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 09/13/2018] [Indexed: 01/06/2023] Open
Abstract
Background Neuronal Ca2+ dyshomeostasis and hyperactivity play a central role in Alzheimer’s disease pathology and progression. Amyloid-beta together with non-genetic risk-factors of Alzheimer’s disease contributes to increased Ca2+ influx and aberrant neuronal activity, which accelerates neurodegeneration in a feed-forward fashion. As such, identifying new targets and drugs to modulate excessive Ca2+ signalling and neuronal hyperactivity, without overly suppressing them, has promising therapeutic potential. Methods Here we show, using biochemical, electrophysiological, imaging, and behavioural tools, that pharmacological modulation of Rap1 signalling by inhibiting its interaction with Pde6δ normalises disease associated Ca2+ aberrations and neuronal activity, conferring neuroprotection in models of Alzheimer’s disease. Results The newly identified inhibitors of the Rap1-Pde6δ interaction counteract AD phenotypes, by reconfiguring Rap1 signalling underlying synaptic efficacy, Ca2+ influx, and neuronal repolarisation, without adverse effects in-cellulo or in-vivo. Thus, modulation of Rap1 by Pde6δ accommodates key mechanisms underlying neuronal activity, and therefore represents a promising new drug target for early or late intervention in neurodegenerative disorders. Conclusion Targeting the Pde6δ-Rap1 interaction has promising therapeutic potential for disorders characterised by neuronal hyperactivity, such as Alzheimer’s disease. Electronic supplementary material The online version of this article (10.1186/s13024-018-0283-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael Dumbacher
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium.,Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH, Utrecht, The Netherlands
| | - Tom Van Dooren
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium
| | - Katrien Princen
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium
| | - Koen De Witte
- reMYND NV, Gaston Geenslaan 1, Leuven-Heverlee, 3001, Belgium
| | - Mélissa Farinelli
- E-Phy-Science, IPMC, 660 route des Lucioles, 06560, Sophia Antipolis, France
| | - Sam Lievens
- Orionis Biosciences, Technologiepark 12B, Zwijnaarde-Ghent, 9052, Belgium.,Cytokine Receptor Lab, Flanders Institute of Biotechnology, Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Lab, Flanders Institute of Biotechnology, Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Wim Dehaen
- Department of Chemistry, KU Leuven, Celestijnenlaan 200f - box 2404, Leuven-Heverlee, 3001, Belgium
| | - Stefaan Wera
- ViroVet NV, Ambachtenlaan 1, Leuven-Heverlee, 3001, Belgium
| | - Joris Winderickx
- Department of Biology, Functional Biology, KU Leuven, Kasteelpark Arenberg 31 box 2433, Leuven-Heverlee, 3001, Belgium
| | - Sara Allasia
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Amuri Kilonda
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Stéphane Spieser
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Arnaud Marchand
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium
| | - Patrick Chaltin
- Cistim Leuven vzw, Gaston Geenslaan 2, Leuven-Heverlee, 3001, Belgium.,Center for Drug Design and Development (CD3), KU Leuven, Waaistraat 6, 3000, Leuven, Belgium
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH, Utrecht, The Netherlands
| | | |
Collapse
|
23
|
Shah SH, Goldberg JL. The Role of Axon Transport in Neuroprotection and Regeneration. Dev Neurobiol 2018; 78:998-1010. [PMID: 30027690 DOI: 10.1002/dneu.22630] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Retinal ganglion cells and other central nervous system neurons fail to regenerate after injury. Understanding the obstacles to survival and regeneration, and overcoming them, is key to preserving and restoring function. While comparisons in the cellular changes seen in these non-regenerative cells with those that do have intrinsic regenerative ability has yielded many candidate genes for regenerative therapies, complete visual recovery has not yet been achieved. Insights gained from neurodegenerative diseases, like glaucoma, underscore the importance of axonal transport of organelles, mRNA, and effector proteins in injury and disease. Targeting molecular motor networks, and their cargoes, may be necessary for realizing complete axonal regeneration and vision restoration.
Collapse
Affiliation(s)
- Sahil H Shah
- Byers Eye Institute, Stanford University, Palo Alto, California.,Neurosciences Graduate Program, University of California, San Diego, California.,Medical Scientist Training Program, University of California, San Diego, California
| | | |
Collapse
|
24
|
Rao SS, Adlard PA. Untangling Tau and Iron: Exploring the Interaction Between Iron and Tau in Neurodegeneration. Front Mol Neurosci 2018; 11:276. [PMID: 30174587 PMCID: PMC6108061 DOI: 10.3389/fnmol.2018.00276] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/20/2018] [Indexed: 11/16/2022] Open
Abstract
There is an emerging link between the accumulation of iron in the brain and abnormal tau pathology in a number of neurodegenerative disorders, such as Alzheimer’s disease (AD). Studies have demonstrated that iron can regulate tau phosphorylation by inducing the activity of multiple kinases that promote tau hyperphosphorylation and potentially also by impacting protein phosphatase 2A activity. Iron is also reported to induce the aggregation of hyperphosphorylated tau, possibly through a direct interaction via a putative iron binding motif in the tau protein, facilitating the formation of neurofibrillary tangles (NFTs). Furthermore, in human studies high levels of iron have been reported to co-localize with tau in NFT-bearing neurons. These data, together with our own work showing that tau has a role in mediating cellular iron efflux, provide evidence supporting a critical tau:iron interaction that may impact both the symptomatic presentation and the progression of disease. Importantly, this may also have relevance for therapeutic directions, and indeed, the use of iron chelators such as deferiprone and deferoxamine have been reported to alleviate the phenotypes, reduce phosphorylated tau levels and stabilize iron regulation in various animal models. As these compounds are also moving towards clinical translation, then it is imperative that we understand the intersection between iron and tau in neurodegeneration. In this article, we provide an overview of the key pathological and biochemical interactions between tau and iron. We also review the role of iron and tau in disease pathology and the potential of metal-based therapies for tauopathies.
Collapse
Affiliation(s)
- Shalini S Rao
- Division of Mental Health, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Anthony Adlard
- Division of Mental Health, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
25
|
Moszczynski AJ, Gopaul J, McCunn P, Volkening K, Harvey M, Bartha R, Schmid S, Strong MJ. Somatic Gene Transfer Using a Recombinant Adenoviral Vector (rAAV9) Encoding Pseudophosphorylated Human Thr175 Tau in Adult Rat Hippocampus Induces Tau Pathology. J Neuropathol Exp Neurol 2018; 77:685-695. [DOI: 10.1093/jnen/nly044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Alexander J Moszczynski
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | | | | | - Kathryn Volkening
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | - Madeline Harvey
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | | | | | - Michael J Strong
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
- Department of Clinical Neurological Sciences, University Hospital, University of Western Ontario, Ontario, Canada
| |
Collapse
|
26
|
Tracy TE, Gan L. Tau-mediated synaptic and neuronal dysfunction in neurodegenerative disease. Curr Opin Neurobiol 2018; 51:134-138. [PMID: 29753269 DOI: 10.1016/j.conb.2018.04.027] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/28/2018] [Accepted: 04/25/2018] [Indexed: 10/16/2022]
Abstract
The accumulation of pathological tau in the brain is associated with neuronal deterioration and cognitive impairments in tauopathies including Alzheimer's disease. Tau, while primarily localized in the axons of healthy neurons, accumulates in the soma and dendrites of neurons under pathogenic conditions. Tau is found in both presynaptic and postsynaptic compartments of neurons in Alzheimer's disease. New research supports that soluble forms of tau trigger pathophysiology in the brain by altering properties of synaptic and neuronal function at the early stages of disease progression, before neurons die. Here we review the current understanding of how tau-mediated synaptic and neuronal dysfunction contributes to cognitive decline. Delineating the mechanisms by which pathogenic tau alters synapses, dendrites and axons will help lay the foundation for new strategies that can restore neuronal function in tauopathy.
Collapse
Affiliation(s)
- Tara E Tracy
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, Weill Institute of Neuroscience, University of California, San Francisco, CA 91458, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, Weill Institute of Neuroscience, University of California, San Francisco, CA 91458, USA; Neuroscience Graduate Program, University of California, San Francisco, CA 91458, USA.
| |
Collapse
|
27
|
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
|
28
|
Wang X, Smith K, Pearson M, Hughes A, Cosden ML, Marcus J, Hess JF, Savage MJ, Rosahl T, Smith SM, Schachter JB, Uslaner JM. Early intervention of tau pathology prevents behavioral changes in the rTg4510 mouse model of tauopathy. PLoS One 2018; 13:e0195486. [PMID: 29624602 PMCID: PMC5889169 DOI: 10.1371/journal.pone.0195486] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/24/2018] [Indexed: 11/19/2022] Open
Abstract
Although tau pathology, behavioral deficits, and neuronal loss are observed in patients with tauopathies, the relationship between these endpoints has not been clearly established. Here we found that rTg4510 mice, which overexpress human mutant tau in the forebrain, develop progressive age-dependent increases in locomotor activity (LMA), which correlates with neurofibrillary tangle (NFT) pathology, hyperphosphorylated tau levels, and brain atrophy. To further clarify the relationship between these endpoints, we treated the rTg4510 mice with either doxycycline to reduce mutant tau expression or an O-GlcNAcase inhibitor Thiamet G, which has been shown to ameliorate tau pathology in animal models. We found that both doxycycline and Thiamet G treatments starting at 2 months of age prevented the progression of hyperactivity, slowed brain atrophy, and reduced brain hyperphosphorylated tau. In contrast, initiating doxycycline treatment at 4 months reduced neither brain hyperphosphorylated tau nor hyperactivity, further confirming the relationship between these measures. Collectively, our results demonstrate a unique behavioral phenotype in the rTg4510 mouse model of tauopathy that strongly correlates with disease progression, and that early interventions which reduce tau pathology ameliorate the progression of the locomotor dysfunction. These findings suggest that better understanding the relationship between locomotor deficits and tau pathology in the rTg4510 model may improve our understanding of the mechanisms underlying behavioral disturbances in patients with tauopathies.
Collapse
Affiliation(s)
- Xiaohai Wang
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
- * E-mail:
| | - Karen Smith
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Michelle Pearson
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Anna Hughes
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Mali L. Cosden
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Jacob Marcus
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - J. Fred Hess
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Mary J. Savage
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Thomas Rosahl
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Sean M. Smith
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Joel B. Schachter
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Jason M. Uslaner
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| |
Collapse
|
29
|
Adalbert R, Milde S, Durrant C, Ando K, Stygelbout V, Yilmaz Z, Gould S, Brion JP, Coleman MP. Interaction between a MAPT variant causing frontotemporal dementia and mutant APP affects axonal transport. Neurobiol Aging 2018; 68:68-75. [PMID: 29729423 PMCID: PMC5998378 DOI: 10.1016/j.neurobiolaging.2018.03.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/27/2018] [Accepted: 03/29/2018] [Indexed: 01/28/2023]
Abstract
In Alzheimer's disease, many indicators point to a central role for poor axonal transport, but the potential for stimulating axonal transport to alleviate the disease remains largely untested. Previously, we reported enhanced anterograde axonal transport of mitochondria in 8- to 11-month-old MAPTP301L knockin mice, a genetic model of frontotemporal dementia with parkinsonism-17T. In this study, we further characterized the axonal transport of mitochondria in younger MAPTP301L mice crossed with the familial Alzheimer's disease model, TgCRND8, aiming to test whether boosting axonal transport in young TgCRND8 mice can alleviate axonal swelling. We successfully replicated the enhancement of anterograde axonal transport in young MAPTP301L/P301L knockin animals. Surprisingly, we found that in the presence of the amyloid precursor protein mutations, MAPTP301L/P3101L impaired anterograde axonal transport. The numbers of plaque-associated axonal swellings or amyloid plaques in TgCRND8 brains were unaltered. These findings suggest that amyloid-β promotes an action of mutant tau that impairs axonal transport. As amyloid-β levels increase with age even without amyloid precursor protein mutation, we suggest that this rise could contribute to age-related decline in frontotemporal dementia.
Collapse
Affiliation(s)
- Robert Adalbert
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK; John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Stefan Milde
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Claire Durrant
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK; John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussells, Belgium
| | - Virginie Stygelbout
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussells, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussells, Belgium
| | - Stacey Gould
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK; John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussells, Belgium
| | - Michael P Coleman
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK; John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Bertrand A, Baron M, Hoang DM, Hill LK, Mendoza SL, Sigurdsson EM, Wadghiri YZ. In Vivo Evaluation of Neuronal Transport in Murine Models of Neurodegeneration Using Manganese-Enhanced MRI. Methods Mol Biol 2018; 1779:527-541. [PMID: 29886555 PMCID: PMC11837276 DOI: 10.1007/978-1-4939-7816-8_33] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Manganese-enhanced MRI (MRI) is a technique that allows for a noninvasive in vivo estimation of neuronal transport. It relies on the physicochemical properties of manganese, which is both a calcium analogue being transported along neurons by active transport, and a paramagnetic compound that can be detected on conventional T1-weighted images. Here, we report a multi-session MEMRI protocol that helps establish time-dependent curves relating to neuronal transport along the olfactory tract over several days. The characterization of these curves via unbiased fitting enables us to infer objectively a set of three parameters (the rate of manganese transport from the maximum slope, the peak intensity, and the time to peak intensity). These parameters, measured previously in wild type mice during normal aging, have served as a baseline to demonstrate their significant sensitivity to pathogenic processes associated with Tau pathology. Importantly, the evaluation of these three parameters and their use as indicators can be extended to monitor any normal and pathogenic processes where neuronal transport is altered. This approach can be applied to characterize and quantify the effect of any neurological disease conditions on neuronal transport in animal models, together with the efficacy of potential therapies.
Collapse
Affiliation(s)
- Anne Bertrand
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, NY, USA
- Institut du Cerveau et la Moelle (ICM), AP-HP-Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Paris, France
- INRIA Paris, Aramis Project-Team, Paris, France
| | - Maria Baron
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, NY, USA
| | - Dung M Hoang
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, NY, USA
| | - Lindsay K Hill
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, NY, USA
- Biomedical Engineering, SUNY Downstate Medical Center, Brooklyn, New York, NY, USA
| | - Sebastian L Mendoza
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, NYU School of Medicine and NYU Langone Health, New York, NY, USA
- Department of Psychiatry, NYU School of Medicine and NYU Langone Health, New York, NY, USA
| | - Youssef Z Wadghiri
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, NY, USA.
| |
Collapse
|
31
|
Guo J, Cheng J, North BJ, Wei W. Functional analyses of major cancer-related signaling pathways in Alzheimer's disease etiology. Biochim Biophys Acta Rev Cancer 2017; 1868:341-358. [PMID: 28694093 PMCID: PMC5675793 DOI: 10.1016/j.bbcan.2017.07.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease and accounts for majority of human dementia. The hyper-phosphorylated tau-mediated intracellular neurofibrillary tangle and amyloid β-mediated extracellular senile plaque are characterized as major pathological lesions of AD. Different from the dysregulated growth control and ample genetic mutations associated with human cancers, AD displays damage and death of brain neurons in the absence of genomic alterations. Although various biological processes predominately governing tumorigenesis such as inflammation, metabolic alteration, oxidative stress and insulin resistance have been associated with AD genesis, the mechanistic connection of these biological processes and signaling pathways including mTOR, MAPK, SIRT, HIF, and the FOXO pathway controlling aging and the pathological lesions of AD are not well recapitulated. Hence, we performed a thorough review by summarizing the physiological roles of these key cancer-related signaling pathways in AD pathogenesis, comprising of the crosstalk of these pathways with neurofibrillary tangle and senile plaque formation to impact AD phenotypes. Importantly, the pharmaceutical investigations of anti-aging and AD relevant medications have also been highlighted. In summary, in this review, we discuss the potential role that cancer-related signaling pathways may play in governing the pathogenesis of AD, as well as their potential as future targeted strategies to delay or prevent aging-related diseases and combating AD.
Collapse
Affiliation(s)
- Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ji Cheng
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Brian J North
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
32
|
De Vos KJ, Hafezparast M. Neurobiology of axonal transport defects in motor neuron diseases: Opportunities for translational research? Neurobiol Dis 2017; 105:283-299. [PMID: 28235672 PMCID: PMC5536153 DOI: 10.1016/j.nbd.2017.02.004] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/20/2017] [Indexed: 12/12/2022] Open
Abstract
Intracellular trafficking of cargoes is an essential process to maintain the structure and function of all mammalian cell types, but especially of neurons because of their extreme axon/dendrite polarisation. Axonal transport mediates the movement of cargoes such as proteins, mRNA, lipids, membrane-bound vesicles and organelles that are mostly synthesised in the cell body and in doing so is responsible for their correct spatiotemporal distribution in the axon, for example at specialised sites such as nodes of Ranvier and synaptic terminals. In addition, axonal transport maintains the essential long-distance communication between the cell body and synaptic terminals that allows neurons to react to their surroundings via trafficking of for example signalling endosomes. Axonal transport defects are a common observation in a variety of neurodegenerative diseases, and mutations in components of the axonal transport machinery have unequivocally shown that impaired axonal transport can cause neurodegeneration (reviewed in El-Kadi et al., 2007, De Vos et al., 2008; Millecamps and Julien, 2013). Here we review our current understanding of axonal transport defects and the role they play in motor neuron diseases (MNDs) with a specific focus on the most common form of MND, amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
- Kurt J De Vos
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Majid Hafezparast
- Neuroscience, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK.
| |
Collapse
|
33
|
Holt DE, Brown DC, Henthorn PS. Evaluation of the dynactin 1 gene in Leonbergers and Labrador Retrievers with laryngeal paralysis. Am J Vet Res 2017; 77:1114-20. [PMID: 27668583 DOI: 10.2460/ajvr.77.10.1114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To sequence exons and splice consensus sites of the dynactin subunit 1 (DCTN1) gene in Leonbergers and Labrador Retrievers with clinical laryngeal paralysis. ANIMALS 5 unrelated Leonbergers with laryngeal paralysis, 2 clinically normal Leonbergers, 7 unrelated Labrador Retrievers with laryngeal paralysis, and 2 clinically normal Labrador Retrievers. PROCEDURES Primers were designed for the entire coding regions of the DCTN1 gene, a noncoding exon at the 5´ end of the gene, and a 900-bp single-nucleotide polymorphism (SNP)-rich region located 17 kb upstream of the DCTN1 gene by use of the CanFam3 assembly of the canine genome sequence. Sequences were generated and compared between clinically normal and affected dogs. The SNPs flanking the DCTN1 gene as well as a previously identified nonsynonymous SNP in exon 32 were genotyped in affected and clinically normal Leonbergers and Labrador Retrievers. RESULTS None of the affected dogs were homozygous for any mutation affecting coding regions or splicing consensus sequences. Of the 16 dogs tested for the missense SNP in exon 32, all were homozygous for the reference allele, except for 2 affected and 1 clinically normal Labrador Retriever and 1 clinically normal Leonberger. The DCTN1 gene sequences (5 dogs) and haplotypes of polymorphic markers surrounding the DCTN1 gene (all dogs) were not consistent with the hypothesis that laryngeal paralysis was associated with inheritance of the same DCTN1 disease-causing allele within all Labrador Retrievers or Leonbergers evaluated. CONCLUSIONS AND CLINICAL RELEVANCE Mutations in the DCTN1 gene did not appear to cause laryngeal paralysis in Leonbergers or Labrador Retrievers.
Collapse
|
34
|
Calcium dysregulation contributes to neurodegeneration in FTLD patient iPSC-derived neurons. Sci Rep 2016; 6:34904. [PMID: 27721502 PMCID: PMC5056519 DOI: 10.1038/srep34904] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/20/2016] [Indexed: 12/23/2022] Open
Abstract
Mutations in the gene MAPT encoding tau, a microtubules-associated protein, cause a subtype of familial neurodegenerative disorder, known as frontotemporal lobar degeneration tauopathy (FTLD-Tau), which presents with dementia and is characterized by atrophy in the frontal and temporal lobes of the brain. Although induced pluripotent stem cell (iPSC) technology has facilitated the investigation of phenotypes of FTLD-Tau patient neuronal cells in vitro, it remains unclear how FTLD-Tau patient neurons degenerate. Here, we established neuronal models of FTLD-Tau by Neurogenin2-induced direct neuronal differentiation from FTLD-Tau patient iPSCs. We found that FTLD-Tau neurons, either with an intronic MAPT mutation or with an exonic mutation, developed accumulation and extracellular release of misfolded tau followed by neuronal death, which we confirmed by correction of the intronic mutation with CRISPR/Cas9. FTLD-Tau neurons showed dysregulation of the augmentation of Ca2+ transients evoked by electrical stimulation. Chemogenetic or pharmacological control of neuronal activity-relevant Ca2+ influx by the introduction of designer receptors exclusively activated by designer drugs (DREADDs) or by the treatment with glutamate receptor blockers attenuated misfolded tau accumulation and neuronal death. These data suggest that neuronal activity may regulate neurodegeneration in tauopathy. This FTLD-Tau model provides mechanistic insights into tauopathy pathogenesis and potential avenues for treatments.
Collapse
|
35
|
Rottscholl R, Haegele M, Jainsch B, Xu H, Respondek G, Höllerhage M, Rösler TW, Bony E, Le Ven J, Guérineau V, Schmitz-Afonso I, Champy P, Oertel WH, Yamada ES, Höglinger GU. Chronic consumption ofAnnona muricatajuice triggers and aggravates cerebral tau phosphorylation in wild-type andMAPTtransgenic mice. J Neurochem 2016; 139:624-639. [DOI: 10.1111/jnc.13835] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022]
Affiliation(s)
| | - Marlen Haegele
- Experimental Neurology; University of Marburg; Marburg Germany
| | - Britta Jainsch
- Experimental Neurology; University of Marburg; Marburg Germany
| | - Hong Xu
- Experimental Neurology; University of Marburg; Marburg Germany
- German Center for Neurodegenerative Diseases (DZNE); Munich Germany
| | - Gesine Respondek
- Experimental Neurology; University of Marburg; Marburg Germany
- German Center for Neurodegenerative Diseases (DZNE); Munich Germany
- Department of Neurology; Technical University Munich; Munich Germany
| | - Matthias Höllerhage
- Experimental Neurology; University of Marburg; Marburg Germany
- German Center for Neurodegenerative Diseases (DZNE); Munich Germany
- Department of Neurology; Technical University Munich; Munich Germany
| | - Thomas W. Rösler
- Experimental Neurology; University of Marburg; Marburg Germany
- German Center for Neurodegenerative Diseases (DZNE); Munich Germany
| | - Emilie Bony
- Laboratoire de Pharmacognosie; BioCIS; Univ. Paris-Sud; CNRS; Université Paris-Saclay; UFR Pharmacie; Châtenay-Malabry France
| | - Jessica Le Ven
- Laboratoire de Pharmacognosie; BioCIS; Univ. Paris-Sud; CNRS; Université Paris-Saclay; UFR Pharmacie; Châtenay-Malabry France
| | - Vincent Guérineau
- Centre de recherche de Gif; Institut de Chimie des Substances Naturelles; CNRS; Gif-sur-Yvette France
| | - Isabelle Schmitz-Afonso
- Centre de recherche de Gif; Institut de Chimie des Substances Naturelles; CNRS; Gif-sur-Yvette France
- Normandie Université; COBRA; UMR 6014 et FR3038; Université de Rouen; INSA de Rouen; CNRS; IRCOF; Mont-Saint-Aignan Cedex France
| | - Pierre Champy
- Laboratoire de Pharmacognosie; BioCIS; Univ. Paris-Sud; CNRS; Université Paris-Saclay; UFR Pharmacie; Châtenay-Malabry France
| | | | - Elizabeth S. Yamada
- Experimental Neurology; University of Marburg; Marburg Germany
- Laboratory of Experimental Neuropathology-ICB; João de Barros Barreto University Hospital; Federal University of Pará; Belém Brazil
| | - Günter U. Höglinger
- Experimental Neurology; University of Marburg; Marburg Germany
- German Center for Neurodegenerative Diseases (DZNE); Munich Germany
- Department of Neurology; Technical University Munich; Munich Germany
| |
Collapse
|
36
|
Intraneuronal aggregation of the β-CTF fragment of APP (C99) induces Aβ-independent lysosomal-autophagic pathology. Acta Neuropathol 2016; 132:257-276. [PMID: 27138984 PMCID: PMC4947121 DOI: 10.1007/s00401-016-1577-6] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/24/2022]
Abstract
Endosomal-autophagic-lysosomal (EAL) dysfunction is an early and prominent neuropathological feature of Alzheimers’s disease, yet the exact molecular mechanisms contributing to this pathology remain undefined. By combined biochemical, immunohistochemical and ultrastructural approaches, we demonstrate a link between EAL pathology and the intraneuronal accumulation of the β-secretase-derived βAPP fragment (C99) in two in vivo models, 3xTgAD mice and adeno-associated viral-mediated C99-infected mice. We present a pathological loop in which the accumulation of C99 is both the effect and causality of impaired lysosomal-autophagic function. The deleterious effect of C99 was found to be linked to its aggregation within EAL-vesicle membranes leading to disrupted lysosomal proteolysis and autophagic impairment. This effect was Aβ independent and was even exacerbated when γ-secretase was pharmacologically inhibited. No effect was observed in inhibitor-treated wild-type animals suggesting that lysosomal dysfunction was indeed directly linked to C99 accumulation. In some brain areas, strong C99 expression also led to inflammatory responses and synaptic dysfunction. Taken together, this work demonstrates a toxic effect of C99 which could underlie some of the early-stage anatomical hallmarks of Alzheimer’s disease pathology. Our work also proposes molecular mechanisms likely explaining some of the unfavorable side-effects associated with γ-secretase inhibitor-directed therapies.
Collapse
|
37
|
Bodea L, Eckert A, Ittner LM, Piguet O, Götz J. Tau physiology and pathomechanisms in frontotemporal lobar degeneration. J Neurochem 2016; 138 Suppl 1:71-94. [PMID: 27306859 PMCID: PMC5094566 DOI: 10.1111/jnc.13600] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/31/2016] [Accepted: 02/24/2016] [Indexed: 12/27/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) has been associated with toxic intracellular aggregates of hyperphosphorylated tau (FTLD-tau). Moreover, genetic studies identified mutations in the MAPT gene encoding tau in familial cases of the disease. In this review, we cover a range of aspects of tau function, both in the healthy and diseased brain, discussing several in vitro and in vivo models. Tau structure and function in the healthy brain is presented, accentuating its distinct compartmentalization in neurons and its role in microtubule stabilization and axonal transport. Furthermore, tau-driven pathology is discussed, introducing current concepts and the underlying experimental evidence. Different aspects of pathological tau phosphorylation, the protein's genomic and domain organization as well as its spreading in disease, together with MAPT-associated mutations and their respective models are presented. Dysfunction related to other post-transcriptional modifications and their effect on normal neuronal functions such as cell cycle, epigenetics and synapse dynamics are also discussed, providing a mechanistic explanation for the observations made in FTLD-tau cases, with the possibility for therapeutic intervention. In this review, we cover aspects of tau function, both in the healthy and diseased brain, referring to different in vitro and in vivo models. In healthy neurons, tau is compartmentalized, with higher concentrations found in the distal part of the axon. Cargo molecules are sensitive to this gradient. A disturbed tau distribution, as found in frontotemporal lobar degeneration (FTLD-tau), has severe consequences for cellular physiology: tau accumulates in the neuronal soma and dendrites, leading among others to microtubule depolymerization and impaired axonal transport. Tau forms insoluble aggregates that sequester additional molecules stalling cellular physiology. Neuronal communication is gradually lost as toxic tau accumulates in dendritic spines with subsequent degeneration of synapses and synaptic loss. Thus, by providing a mechanistic explanation for the observations made in FTLD-tau cases, arises a possibility for therapeutic interventions. This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Liviu‐Gabriel Bodea
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Anne Eckert
- Neurobiology LaboratoryPsychiatric University Clinics BaselUniversity of BaselBaselSwitzerland
| | - Lars Matthias Ittner
- Dementia Research UnitSchool of Medical SciencesFaculty of MedicineUniversity of New South WalesSydneyNSWAustralia
| | | | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
38
|
Maeda S, Djukic B, Taneja P, Yu GQ, Lo I, Davis A, Craft R, Guo W, Wang X, Kim D, Ponnusamy R, Gill TM, Masliah E, Mucke L. Expression of A152T human tau causes age-dependent neuronal dysfunction and loss in transgenic mice. EMBO Rep 2016; 17:530-51. [PMID: 26931567 PMCID: PMC4818780 DOI: 10.15252/embr.201541438] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/01/2015] [Accepted: 01/13/2016] [Indexed: 11/24/2022] Open
Abstract
A152T-variant human tau (hTau-A152T) increases risk for tauopathies, including Alzheimer's disease. Comparing mice with regulatable expression of hTau-A152T or wild-type hTau (hTau-WT), we find age-dependent neuronal loss, cognitive impairments, and spontaneous nonconvulsive epileptiform activity primarily in hTau-A152T mice. However, overexpression of either hTau species enhances neuronal responses to electrical stimulation of synaptic inputs and to an epileptogenic chemical. hTau-A152T mice have higher hTau protein/mRNA ratios in brain, suggesting that A152T increases production or decreases clearance of hTau protein. Despite their functional abnormalities, aging hTau-A152T mice show no evidence for accumulation of insoluble tau aggregates, suggesting that their dysfunctions are caused by soluble tau. In human amyloid precursor protein (hAPP) transgenic mice, co-expression of hTau-A152T enhances risk of early death and epileptic activity, suggesting copathogenic interactions between hTau-A152T and amyloid-β peptides or other hAPP metabolites. Thus, the A152T substitution may augment risk for neurodegenerative diseases by increasing hTau protein levels, promoting network hyperexcitability, and synergizing with the adverse effects of other pathogenic factors.
Collapse
Affiliation(s)
- Sumihiro Maeda
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Biljana Djukic
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Praveen Taneja
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Allyson Davis
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Ryan Craft
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Weikun Guo
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Xin Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Daniel Kim
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - T Michael Gill
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
39
|
Koss DJ, Robinson L, Drever BD, Plucińska K, Stoppelkamp S, Veselcic P, Riedel G, Platt B. Mutant Tau knock-in mice display frontotemporal dementia relevant behaviour and histopathology. Neurobiol Dis 2016; 91:105-23. [PMID: 26949217 DOI: 10.1016/j.nbd.2016.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/26/2016] [Accepted: 03/02/2016] [Indexed: 10/22/2022] Open
Abstract
Models of Tau pathology related to frontotemporal dementia (FTD) are essential to determine underlying neurodegenerative pathologies and resulting tauopathy relevant behavioural changes. However, existing models are often limited in their translational value due to Tau overexpression, and the frequent occurrence of motor deficits which prevent comprehensive behavioural assessments. In order to address these limitations, a forebrain-specific (CaMKIIα promoter), human mutated Tau (hTauP301L+R406W) knock-in mouse was generated out of the previously characterised PLB1Triple mouse, and named PLB2Tau. After confirmation of an additional hTau species (~60kDa) in forebrain samples, we identified age-dependent progressive Tau phosphorylation which coincided with the emergence of FTD relevant behavioural traits. In line with the non-cognitive symptomatology of FTD, PLB2Tau mice demonstrated early emerging (~6months) phenotypes of heightened anxiety in the elevated plus maze, depressive/apathetic behaviour in a sucrose preference test and generally reduced exploratory activity in the absence of motor impairments. Investigations of cognitive performance indicated prominent dysfunctions in semantic memory, as assessed by social transmission of food preference, and in behavioural flexibility during spatial reversal learning in a home cage corner-learning task. Spatial learning was only mildly affected and task-specific, with impairments at 12months of age in the corner learning but not in the water maze task. Electroencephalographic (EEG) investigations indicated a vigilance-stage specific loss of alpha power during wakefulness at both parietal and prefrontal recording sites, and site-specific EEG changes during non-rapid eye movement sleep (prefrontal) and rapid eye movement sleep (parietal). Further investigation of hippocampal electrophysiology conducted in slice preparations indicated a modest reduction in efficacy of synaptic transmission in the absence of altered synaptic plasticity. Together, our data demonstrate that the transgenic PLB2Tau mouse model presents with a striking behavioural and physiological face validity relevant for FTD, driven by the low level expression of mutant FTD hTau.
Collapse
Affiliation(s)
- David J Koss
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Lianne Robinson
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Benjamin D Drever
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Kaja Plucińska
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Sandra Stoppelkamp
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter Veselcic
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Gernot Riedel
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | - Bettina Platt
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
40
|
Hannan SB, Dräger NM, Rasse TM, Voigt A, Jahn TR. Cellular and molecular modifier pathways in tauopathies: the big picture from screening invertebrate models. J Neurochem 2016; 137:12-25. [PMID: 26756400 DOI: 10.1111/jnc.13532] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/14/2015] [Accepted: 01/04/2016] [Indexed: 12/16/2022]
Abstract
Abnormal tau accumulations were observed and documented in post-mortem brains of patients affected by Alzheimer's disease (AD) long before the identification of mutations in the Microtubule-associated protein tau (MAPT) gene, encoding the tau protein, in a different neurodegenerative disease called Frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17). The discovery of mutations in the MAPT gene associated with FTDP-17 highlighted that dysfunctions in tau alone are sufficient to cause neurodegeneration. Invertebrate models have been diligently utilized in investigating tauopathies, contributing to the understanding of cellular and molecular pathways involved in disease etiology. An important discovery came with the demonstration that over-expression of human tau in Drosophila leads to premature mortality and neuronal dysfunction including neurodegeneration, recapitulating some key neuropathological features of the human disease. The simplicity of handling invertebrate models combined with the availability of a diverse range of experimental resources make these models, in particular Drosophila a powerful invertebrate screening tool. Consequently, several large-scale screens have been performed using Drosophila, to identify modifiers of tau toxicity. The screens have revealed not only common cellular and molecular pathways, but in some instances the same modifier has been independently identified in two or more screens suggesting a possible role for these modifiers in regulating tau toxicity. The purpose of this review is to discuss the genetic modifier screens on tauopathies performed in Drosophila and C. elegans models, and to highlight the common cellular and molecular pathways that have emerged from these studies. Here, we summarize results of tau toxicity screens providing mechanistic insights into pathological alterations in tauopathies. Key pathways or modifiers that have been identified are associated with a broad range of processes including, but not limited to, phosphorylation, cytoskeleton organization, axonal transport, regulation of cellular proteostasis, transcription, RNA metabolism, cell cycle regulation, and apoptosis. We discuss the utility and application of invertebrate models in elucidating the cellular and molecular functions of novel and uncharacterized disease modifiers identified in large-scale screens as well as for investigating the function of genes identified as risk factors in genome-wide association studies from human patients in the post-genomic era. In this review, we combined and summarized several large-scale modifier screens performed in invertebrate models to identify modifiers of tau toxicity. A summary of the screens show that diverse cellular processes are implicated in the modification of tau toxicity. Kinases and phosphatases are the most predominant class of modifiers followed by components required for cellular proteostasis and axonal transport and cytoskeleton elements.
Collapse
Affiliation(s)
- Shabab B Hannan
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany.,Graduate School of Cellular and Molecular Neuroscience, Graduate Training Center of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Nina M Dräger
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany
| | - Tobias M Rasse
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany
| | - Aaron Voigt
- Department of Neurology at University Clinic Aachen, RWTH Aachen, Germany
| | - Thomas R Jahn
- Schaller Research Group at the University of Heidelberg and DKFZ, Proteostasis in Neurodegenerative Disease (B180), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
41
|
Huang Y, Wu Z, Zhou B. Behind the curtain of tauopathy: a show of multiple players orchestrating tau toxicity. Cell Mol Life Sci 2016; 73:1-21. [PMID: 26403791 PMCID: PMC11108533 DOI: 10.1007/s00018-015-2042-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/22/2015] [Accepted: 09/08/2015] [Indexed: 12/24/2022]
Abstract
tau, a microtubule-associated protein, directly binds with microtubules to dynamically regulate the organization of cellular cytoskeletons, and is especially abundant in neurons of the central nervous system. Under disease conditions such as Pick's disease, progressive supranuclear palsy, frontotemporal dementia, parkinsonism linked to chromosome 17 and Alzheimer's disease, tau proteins can self-assemble to paired helical filaments progressing to neurofibrillary tangles. In these diseases, collectively referred to as "tauopathies", alterations of diverse tau modifications including phosphorylation, metal ion binding, glycosylation, as well as structural changes of tau proteins have all been observed, indicating the complexity and variability of factors in the regulation of tau toxicity. Here, we review our current knowledge and hypotheses from relevant studies on tau toxicity, emphasizing the roles of phosphorylations, metal ions, folding and clearance control underlining tau etiology and their regulations. A summary of clinical efforts and associated findings of drug candidates under development is also presented. It is hoped that a more comprehensive understanding of tau regulation will provide us with a better blueprint of tau networking in neuronal cells and offer hints for the design of more efficient strategies to tackle tau-related diseases in the future.
Collapse
Affiliation(s)
- Yunpeng Huang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhihao Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Bing Zhou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
42
|
Sengupta U, Guerrero-Muñoz MJ, Castillo-Carranza DL, Lasagna-Reeves CA, Gerson JE, Paulucci-Holthauzen AA, Krishnamurthy S, Farhed M, Jackson GR, Kayed R. Pathological interface between oligomeric alpha-synuclein and tau in synucleinopathies. Biol Psychiatry 2015; 78:672-83. [PMID: 25676491 DOI: 10.1016/j.biopsych.2014.12.019] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Aberrant accumulation of α-synuclein constitutes inclusion bodies that are considered a characteristic feature of a group of neurological disorders described as synucleinopathies. Often, multiple disease-causing proteins overlap within a given disease pathology. An emerging body of research focuses on the oligomeric populations of various pathogenic proteins, considering them as the culprits causing neuronal damage and degeneration. To this end, the use of conformation-specific antibodies has proven to be an effective tool. Previous work from our laboratory and others has shown that oligomeric entities of α-synuclein and tau accumulate in their respective diseases, but their interrelationship at this higher order has yet to be shown in synucleinopathies. METHODS Here, we used two novel conformation-specific antibodies, F8H7 and Syn33, which recognize α-synuclein oligomers and were developed in our laboratory. We investigated brain tissue from five of each Parkinson's disease and dementia with Lewy bodies patients by performing biophysical and biochemical assays using these antibodies, in addition to the previously characterized anti-tau oligomer antibody T22. RESULTS We demonstrate that in addition to the deposition of oligomeric α-synuclein, tau oligomers accumulate in these diseased brains compared with control brains. Moreover, we observed that oligomers of tau and α-synuclein exist in the same aggregates, forming hybrid oligomers in these patients' brains. CONCLUSIONS In addition to the deposition of tau oligomers, our results also provide compelling evidence of co-occurrence of α-synuclein and tau into their most toxic forms, i.e., oligomers suggesting that these species interact and influence each other's aggregation via an interface in synucleinopathies.
Collapse
Affiliation(s)
- Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Marcos J Guerrero-Muñoz
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Diana L Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Cristian A Lasagna-Reeves
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Julia E Gerson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | | | - Shashirekha Krishnamurthy
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Malika Farhed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - George R Jackson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology and Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
43
|
Vossel KA, Xu JC, Fomenko V, Miyamoto T, Suberbielle E, Knox JA, Ho K, Kim DH, Yu GQ, Mucke L. Tau reduction prevents Aβ-induced axonal transport deficits by blocking activation of GSK3β. ACTA ACUST UNITED AC 2015; 209:419-33. [PMID: 25963821 PMCID: PMC4427789 DOI: 10.1083/jcb.201407065] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tau ablation, knockdown, and reconstitution studies in primary mouse neurons show that tau enables amyloid β oligomers to inhibit axonal transport through activation of GSK3β and through functions of tau that do not depend on its microtubule binding activity. Axonal transport deficits in Alzheimer’s disease (AD) are attributed to amyloid β (Aβ) peptides and pathological forms of the microtubule-associated protein tau. Genetic ablation of tau prevents neuronal overexcitation and axonal transport deficits caused by recombinant Aβ oligomers. Relevance of these findings to naturally secreted Aβ and mechanisms underlying tau’s enabling effect are unknown. Here we demonstrate deficits in anterograde axonal transport of mitochondria in primary neurons from transgenic mice expressing familial AD-linked forms of human amyloid precursor protein. We show that these deficits depend on Aβ1–42 production and are prevented by tau reduction. The copathogenic effect of tau did not depend on its microtubule binding, interactions with Fyn, or potential role in neuronal development. Inhibition of neuronal activity, N-methyl-d-aspartate receptor function, or glycogen synthase kinase 3β (GSK3β) activity or expression also abolished Aβ-induced transport deficits. Tau ablation prevented Aβ-induced GSK3β activation. Thus, tau allows Aβ oligomers to inhibit axonal transport through activation of GSK3β, possibly by facilitating aberrant neuronal activity.
Collapse
Affiliation(s)
- Keith A Vossel
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| | - Jordan C Xu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Vira Fomenko
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Takashi Miyamoto
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| | - Elsa Suberbielle
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| | - Joseph A Knox
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Daniel H Kim
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
44
|
Butzlaff M, Hannan SB, Karsten P, Lenz S, Ng J, Voßfeldt H, Prüßing K, Pflanz R, Schulz JB, Rasse T, Voigt A. Impaired retrograde transport by the Dynein/Dynactin complex contributes to Tau-induced toxicity. Hum Mol Genet 2015; 24:3623-37. [DOI: 10.1093/hmg/ddv107] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/17/2015] [Indexed: 11/12/2022] Open
|
45
|
Fontaine SN, Sabbagh JJ, Baker J, Martinez-Licha CR, Darling A, Dickey CA. Cellular factors modulating the mechanism of tau protein aggregation. Cell Mol Life Sci 2015; 72:1863-79. [PMID: 25666877 DOI: 10.1007/s00018-015-1839-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/18/2014] [Accepted: 01/13/2015] [Indexed: 01/12/2023]
Abstract
Pathological accumulation of the microtubule-associated protein tau, in the form of neurofibrillary tangles, is a major hallmark of Alzheimer's disease, the most prevalent neurodegenerative condition worldwide. In addition to Alzheimer's disease, a number of neurodegenerative diseases, called tauopathies, are characterized by the accumulation of aggregated tau in a variety of brain regions. While tau normally plays an important role in stabilizing the microtubule network of the cytoskeleton, its dissociation from microtubules and eventual aggregation into pathological deposits is an area of intense focus for therapeutic development. Here we discuss the known cellular factors that affect tau aggregation, from post-translational modifications to molecular chaperones.
Collapse
Affiliation(s)
- Sarah N Fontaine
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
| | | | | | | | | | | |
Collapse
|
46
|
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
|
47
|
Ren Y, Sahara N, Giasson B, Lewis J. Tauopathy Mouse Models. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00055-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
48
|
Scholz T, Mandelkow E. Transport and diffusion of Tau protein in neurons. Cell Mol Life Sci 2014; 71:3139-50. [PMID: 24687422 PMCID: PMC11113808 DOI: 10.1007/s00018-014-1610-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/20/2014] [Accepted: 03/13/2014] [Indexed: 12/11/2022]
Abstract
In highly polarized and elongated cells such as neurons, Tau protein must enter and move down the axon to fulfill its biological task of stabilizing axonal microtubules. Therefore, cellular systems for distributing Tau molecules are needed. This review discusses different mechanisms that have been proposed to contribute to the dispersion of Tau molecules in neurons. They include (1) directed transport along microtubules as cargo of tubulin complexes and/or motor proteins, (2) diffusion, either through the cytosolic space or along microtubules, and (3) mRNA-based mechanisms such as transport of Tau mRNA into axons and local translation. Diffusion along the microtubule lattice or through the cytosol appear to be the major mechanisms for axonal distribution of Tau protein in the short-to-intermediate range over distances of up to a millimetre. The high diffusion coefficients ensure that Tau can distribute evenly throughout the axonal volume as well as along microtubules. Motor protein-dependent transport of Tau dominates over longer distances and time scales. At low near-physiological levels, Tau is co-transported along with short microtubules from cell bodies into axons by cytoplasmic dynein and kinesin family members at rates of slow axonal transport.
Collapse
Affiliation(s)
- Tim Scholz
- Institute for Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany,
| | | |
Collapse
|
49
|
Axonal Transport Defects in Alzheimer’s Disease. Mol Neurobiol 2014; 51:1309-21. [DOI: 10.1007/s12035-014-8810-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/10/2014] [Indexed: 10/25/2022]
|
50
|
Yuan A, Kumar A, Sasaki T, Duff K, Nixon RA. Global axonal transport rates are unaltered in htau mice in vivo. J Alzheimers Dis 2014; 37:579-86. [PMID: 23948900 DOI: 10.3233/jad-130671] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Microtubule-based axonal transport is believed to become globally disrupted in Alzheimer's disease in part due to alterations of tau expression or phosphorylation. We previously showed that axonal transport rates along retinal ganglion axons are unaffected by deletion of normal mouse tau or by overexpression of wild-type human tau. Here, we report that htau mice expressing 3-fold higher levels of human tau in the absence of mouse tau also display normal fast and slow transport kinetics despite the presence of abnormally hyperphosphorylated tau in some neurons. In addition, markers of slow transport (neurofilament light subunit) and fast transport (snap25) exhibit normal distributions along optic axons of these mice. These studies demonstrate that human tau overexpression, even when associated with a limited degree of tau pathology, does not necessarily impair general axonal transport function in vivo.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA Department of Psychiatry, New York University School of Medicine, NY, USA
| | | | | | | | | |
Collapse
|