1
|
Hivare P, Mujmer K, Swarup G, Gupta S, Bhatia D. Endocytic pathways of pathogenic protein aggregates in neurodegenerative diseases. Traffic 2023; 24:434-452. [PMID: 37392160 DOI: 10.1111/tra.12906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/14/2023] [Accepted: 06/11/2023] [Indexed: 07/03/2023]
Abstract
Endocytosis is the fundamental uptake process through which cells internalize extracellular materials and species. Neurodegenerative diseases (NDs) are characterized by a progressive accumulation of intrinsically disordered protein species, leading to neuronal death. Misfolding in many proteins leads to various NDs such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and other disorders. Despite the significance of disordered protein species in neurodegeneration, their spread between cells and the cellular uptake of extracellular species is not entirely understood. This review discusses the major internalization mechanisms of the different conformer species of these proteins and their endocytic mechanisms. We briefly introduce the broad types of endocytic mechanisms found in cells and then summarize what is known about the endocytosis of monomeric, oligomeric and aggregated conformations of tau, Aβ, α-Syn, Huntingtin, Prions, SOD1, TDP-43 and other proteins associated with neurodegeneration. We also highlight the key players involved in internalizing these disordered proteins and the several techniques and approaches to identify their endocytic mechanisms. Finally, we discuss the obstacles involved in studying the endocytosis of these protein species and the need to develop better techniques to elucidate the uptake mechanisms of a particular disordered protein species.
Collapse
Affiliation(s)
- Pravin Hivare
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Kratika Mujmer
- Center for Brain and Cognitive Sciences, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Gitanjali Swarup
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Sharad Gupta
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Dhiraj Bhatia
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| |
Collapse
|
2
|
Kapuganti RS, Hayat B, Padhy B, Mohanty PP, Alone DP. Dickkopf-1 and ROCK2 upregulation and associated protein aggregation in pseudoexfoliation syndrome and glaucoma. Life Sci 2023; 326:121797. [PMID: 37217133 DOI: 10.1016/j.lfs.2023.121797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/08/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
AIMS The etiology of pseudoexfoliation (PEX), a stress-induced fibrillopathy and a leading cause of secondary glaucoma worldwide, remains limited. This study aims to understand the role of the Wnt antagonist Dickkopf-related protein 1 (DKK1) in PEX pathophysiology and assess its candidature as a biomarker for PEX. MAIN METHODS Expression levels of DKK1 and Wnt signaling genes were assayed in the anterior ocular tissues of study subjects by qRT-PCR, Western blotting, and immunohistochemistry. Protein aggregation was studied through Proteostat staining. Role of DKK1 in protein aggregation and regulation of target Wnt signaling genes was elucidated through overexpression and knockdown studies in Human Lens Epithelial cells (HLEB3). Levels of DKK1 in circulating fluids were assayed through ELISA. KEY FINDINGS DKK1 upregulation was observed in lens capsule and conjunctiva tissues of PEX individuals compared to controls correlating with an upregulation of the Wnt signaling target, ROCK2. Proteostat staining showed increased protein aggregates in lens epithelial cells of PEX patients. HLE B-3 cells overexpressed with DKK1 showed increased protein aggregates along with upregulation of ROCK2, and knockdown of DKK1 in HLE B-3 cells demonstrated downregulation of ROCK2. Further, ROCK2 inhibition by Y-27632 in DKK1 overexpressed cells showed that DKK1 regulated protein aggregation via ROCK2. Also, increased levels of DKK1 were observed in patients' plasma and aqueous humor compared to controls. SIGNIFICANCE This study shows that DKK1 and ROCK2 might play a role in protein aggregation in PEX. Further, elevated levels of DKK1 in aqueous humor serve as a fair classifier of pseudoexfoliation glaucoma.
Collapse
Affiliation(s)
- Ramani Shyam Kapuganti
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Bushra Hayat
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Biswajit Padhy
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India
| | | | - Debasmita Pankaj Alone
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
3
|
Paoletti M, Caverzasi E, Mandelli ML, Brown JA, Henry RG, Miller BL, Rosen HJ, DeArmond SJ, Bastianello S, Seeley WW, Geschwind MD. Default Mode Network quantitative diffusion and resting-state functional magnetic resonance imaging correlates in sporadic Creutzfeldt-Jakob disease. Hum Brain Mapp 2022; 43:4158-4173. [PMID: 35662331 PMCID: PMC9374887 DOI: 10.1002/hbm.25945] [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: 01/22/2021] [Revised: 04/14/2022] [Accepted: 05/01/2022] [Indexed: 11/25/2022] Open
Abstract
Grey matter involvement is a well-known feature in sporadic Creutzfeldt-Jakob disease (sCJD), yet precise anatomy-based quantification of reduced diffusivity is still not fully understood. Default Mode Network (DMN) areas have been recently demonstrated as selectively involved in sCJD, and functional connectivity has never been investigated in prion diseases. We analyzed the grey matter involvement using a quantitatively multi-parametric MRI approach. Specifically, grey matter mean diffusivity of 37 subjects with sCJD was compared with that of 30 age-matched healthy controls with a group-wise approach. Differences in mean diffusivity were also examined between the cortical (MM(V)1, MM(V)2C, and VV1) and subcortical (VV2 and MV2K) subgroups of sCJD for those with autopsy data available (n = 27, 73%). We also assessed resting-state functional connectivity of both ventral and dorsal components of DMN in a subset of subject with a rs-fMRI dataset available (n = 17). Decreased diffusivity was predominantly present in posterior cortical regions of the DMN, but also outside of the DMN in temporal areas and in a few limbic and frontal areas, in addition to extensive deep nuclei involvement. Both subcortical and cortical sCJD subgroups showed decreased diffusivity subcortically, whereas only the cortical type expressed significantly decreased diffusivity cortically, mainly in parietal, occipital, and medial-inferior temporal cortices bilaterally. Interestingly, we found abnormally increased connectivity in both dorsal and ventral components of the DMN in sCJD subjects compared with healthy controls. The significance and possible utility of functional imaging as a biomarker for tracking disease progression in prion disease needs to be explored further.
Collapse
Affiliation(s)
- Matteo Paoletti
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeuroradiologyIRCCS Mondino FoundationPaviaItaly
| | - Eduardo Caverzasi
- Weill Institute for Neurosciences, Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | - Maria Luisa Mandelli
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jesse A. Brown
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Roland G. Henry
- Weill Institute for Neurosciences, Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Graduate Group in BioengineeringUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Howard J. Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Stefano Bastianello
- Department of NeuroradiologyIRCCS Mondino FoundationPaviaItaly
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Michael D. Geschwind
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
4
|
Lopes DM, Llewellyn SK, Harrison IF. Propagation of tau and α-synuclein in the brain: therapeutic potential of the glymphatic system. Transl Neurodegener 2022; 11:19. [PMID: 35314000 PMCID: PMC8935752 DOI: 10.1186/s40035-022-00293-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Many neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease, are characterised by the accumulation of misfolded protein deposits in the brain, leading to a progressive destabilisation of the neuronal network and neuronal death. Among the proteins that can abnormally accumulate are tau and α-synuclein, which can propagate in a prion-like manner and which upon aggregation, represent the most common intracellular proteinaceous lesions associated with neurodegeneration. For years it was thought that these intracellular proteins and their accumulation had no immediate relationship with extracellular homeostasis pathways such as the glymphatic clearance system; however, mounting evidence has now suggested that this is not the case. The involvement of the glymphatic system in neurodegenerative disease is yet to be fully defined; however, it is becoming increasingly clear that this pathway contributes to parenchymal solute clearance. Importantly, recent data show that proteins prone to intracellular accumulation are subject to glymphatic clearance, suggesting that this system plays a key role in many neurological disorders. In this review, we provide a background on the biology of tau and α-synuclein and discuss the latest findings on the cell-to-cell propagation mechanisms of these proteins. Importantly, we discuss recent data demonstrating that manipulation of the glymphatic system may have the potential to alleviate and reduce pathogenic accumulation of propagation-prone intracellular cytotoxic proteins. Furthermore, we will allude to the latest potential therapeutic opportunities targeting the glymphatic system that might have an impact as disease modifiers in neurodegenerative diseases.
Collapse
|
5
|
Awasthi P, Das S. Kinetics of protein aggregation at a temperature gradient condition. SOFT MATTER 2021; 17:9008-9013. [PMID: 34610083 DOI: 10.1039/d1sm00857a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The unconventional multi-sigmoidal kinetic behaviour of protein aggregation at a temperature gradient condition is reported in this study. To establish a feasible theory for protein aggregation kinetics at a temperature gradient condition, the spatial height of the protein solution is divided into hypothetical layers and the kinetic equations in those layers are solved. Furthermore, we endeavour to study numerically the effect of the temperature gradient on the kinetics of oligomer-mediated protein aggregation and protein inhibition.
Collapse
Affiliation(s)
- Prasoon Awasthi
- BioMEMS and Microfluidic Laboratory, School of Medical Science and Technology, IIT Kharagpur, 721302, India.
| | - Soumen Das
- BioMEMS and Microfluidic Laboratory, School of Medical Science and Technology, IIT Kharagpur, 721302, India.
| |
Collapse
|
6
|
Sheikh AM, Wada Y, Tabassum S, Inagaki S, Mitaki S, Yano S, Nagai A. Aggregation of Cystatin C Changes Its Inhibitory Functions on Protease Activities and Amyloid β Fibril Formation. Int J Mol Sci 2021; 22:ijms22189682. [PMID: 34575849 PMCID: PMC8465189 DOI: 10.3390/ijms22189682] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
Cystatin C (CST3) is an endogenous cysteine protease inhibitor, which is implicated in cerebral amyloid angiopathy (CAA). In CAA, CST3 is found to be aggregated. The purpose of this study is to investigate whether this aggregation could alter the activity of the protein relevant to the molecular pathology of CAA. A system of CST3 protein aggregation was established, and the aggregated protein was characterized. The results showed that CST3 aggregated both at 80 °C without agitation, and at 37 °C with agitation in a time-dependent manner. However, the levels of aggregation were high and appeared earlier at 80 °C. Dot-blot immunoassay for oligomers revealed that CST3 could make oligomeric aggregates at the 37 °C condition. Electron microscopy showed that CST3 could make short fibrillary aggregates at 37 °C. Cathepsin B activity assay demonstrated that aggregated CST3 inhibited the enzyme activity less efficiently at pH 5.5. At 7.4 pH, it lost the inhibitory properties almost completely. In addition, aggregated CST3 did not inhibit Aβ1-40 fibril formation, rather, it slightly increased it. CST3 immunocytochemistry showed that the protein was positive both in monomeric and aggregated CST3-treated neuronal culture. However, His6 immunocytochemistry revealed that the internalization of exogenous recombinant CST3 by an astrocytoma cell culture was higher when the protein was aggregated compared to its monomeric form. Finally, MTT cell viability assay showed that the aggregated form of CST3 was more toxic than the monomeric form. Thus, our results suggest that aggregation may result in a loss-of-function phenotype of CST3, which is toxic and responsible for cellular degeneration.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Yasuko Wada
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
| | - Shatera Tabassum
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Satoshi Inagaki
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
| | - Shingo Mitaki
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
| | - Shozo Yano
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Atsushi Nagai
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
- Correspondence: ; Tel./Fax: +81-0853-20-2198
| |
Collapse
|
7
|
Himalian R, Singh SK, Singh MP. Ameliorative Role of Nutraceuticals on Neurodegenerative Diseases Using the Drosophila melanogaster as a Discovery Model to Define Bioefficacy. J Am Coll Nutr 2021; 41:511-539. [PMID: 34125661 DOI: 10.1080/07315724.2021.1904305] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Neurodegeneration is the destruction of neurons, and once the neurons degenerate they can't revive. This is one of the most concerned health conditions among aged population, more than ∼70% of the elderly people are suffering from neurodegeneration. Among all of the neurodegenerative diseases, Alzheimer's disease (AD), Parkinson's disease (PD) and Poly-glutamine disease (Poly-Q) are the major one and affecting most of the people around the world and posing excessive burden on the society. In order to understand this disease in non-human animal models it is pertinent to examine in model organism and various animal model are being used for such diseases like rat, mice and non-vertebrate model like Drosophila. Drosophila melanogaster is one of the best animal proven by several eminent scientist and had received several Nobel prizes for uncovering mechanism of human related genes and highly efficient model for studying neurodegenerative diseases due to its great affinity with human disease-related genes. Another factor is also employed to act as therapeutic or preventive method that is nutraceuticals. Nutraceuticals are functional natural compounds with antioxidant properties and had extensively showed the neuroprotective effect in different organisms. These nutraceuticals having antioxidant properties act through scavenging free radicals or by increasing endogenous cellular antioxidant defense molecules. For the best benefit, we are trying to utilize these nutraceuticals, which will have no or negligible side effects. In this review, we are dealing with various types of such nutraceuticals which have potent value in the prevention and curing of the diseases related to neurodegeneration.HighlightsNeurodegeneration is the silently progressing disease which shows its symptoms when it is well rooted.Many chemical drugs (almost all) have only symptomatic relief with side effects.Potent mechanism of neurodegeneration and improvement effect by nutraceuticals is proposed.Based on the Indian Cuisine scientists are trying to find the medicine from the food or food components having antioxidant properties.The best model to study the neurodegenerative diseases is Drosophila melanogaster.Many nutraceuticals having antioxidant properties have been studied and attenuated various diseases are discussed.
Collapse
Affiliation(s)
- Ranjana Himalian
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology (ISET) Foundation, Lucknow, India
| | - Mahendra Pratap Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
8
|
Lo Cascio F, Garcia S, Montalbano M, Puangmalai N, McAllen S, Pace A, Palumbo Piccionello A, Kayed R. Modulating disease-relevant tau oligomeric strains by small molecules. J Biol Chem 2020; 295:14807-14825. [PMID: 32737202 PMCID: PMC7606668 DOI: 10.1074/jbc.ra120.014630] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
The pathological aggregation of tau plays an important role in Alzheimer's disease and many other related neurodegenerative diseases, collectively referred to as tauopathies. Recent evidence has demonstrated that tau oligomers, small and soluble prefibrillar aggregates, are highly toxic due to their strong ability to seed tau misfolding and propagate the pathology seen across different neurodegenerative diseases. We previously showed that novel curcumin derivatives affect preformed tau oligomer aggregation pathways by promoting the formation of more aggregated and nontoxic tau aggregates. To further investigate their therapeutic potential, we have extended our studies o disease-relevant brain-derived tau oligomers (BDTOs). Herein, using well-characterized BDTOs, isolated from brain tissues of different tauopathies, including Alzheimer's disease, progressive supranuclear palsy, and dementia with Lewy bodies, we found that curcumin derivatives modulate the aggregation state of BDTOs by reshaping them and rescue neurons from BDTO-associated toxicity. Interestingly, compound CL3 showed an effect on the aggregation pattern of BDTOs from different tauopathies, resulting in the formation of less neurotoxic larger tau aggregates with decreased hydrophobicity and seeding propensity. Our results lay the groundwork for potential investigations of the efficacy and beneficial effects of CL3 and other promising compounds for the treatment of tauopathies. Furthermore, CL3 may aid in the development of tau imaging agent for the detection of tau oligomeric strains and differential diagnosis of the tauopathies, thus enabling earlier interventions.
Collapse
Affiliation(s)
- Filippa Lo Cascio
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Stephanie Garcia
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Salome McAllen
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrea Pace
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies-STEBICEF, University of Palermo, Palermo, Italy
| | - Antonio Palumbo Piccionello
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies-STEBICEF, University of Palermo, Palermo, Italy
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
9
|
Stolerman LM, Getz M, Smith SGL, Holst M, Rangamani P. Stability Analysis of a Bulk-Surface Reaction Model for Membrane Protein Clustering. Bull Math Biol 2020; 82:30. [PMID: 32025918 DOI: 10.1007/s11538-020-00703-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022]
Abstract
Protein aggregation on the plasma membrane (PM) is of critical importance to many cellular processes such as cell adhesion, endocytosis, fibrillar conformation, and vesicle transport. Lateral diffusion of protein aggregates or clusters on the surface of the PM plays an important role in governing their heterogeneous surface distribution. However, the stability behavior of the surface distribution of protein aggregates remains poorly understood. Therefore, understanding the spatial patterns that can emerge on the PM solely through protein-protein interaction, lateral diffusion, and feedback is an important step toward a complete description of the mechanisms behind protein clustering on the cell surface. In this work, we investigate the pattern formation of a reaction-diffusion model that describes the dynamics of a system of ligand-receptor complexes. The purely diffusive ligand in the cytosol can bind receptors in the PM and the resultant ligand-receptor complexes not only diffuse laterally but can also form clusters resulting in different oligomers. Finally, the largest oligomers recruit ligands from the cytosol using positive feedback. From a methodological viewpoint, we provide theoretical estimates for diffusion-driven instabilities of the protein aggregates based on the Turing mechanism. Our main result is a threshold phenomenon, in which a sufficiently high recruitment of ligands promotes the input of new monomeric components and consequently drives the formation of a single-patch spatially heterogeneous steady state.
Collapse
Affiliation(s)
- Lucas M Stolerman
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093-0411, USA
| | - Michael Getz
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093-0411, USA
| | - Stefan G Llewellyn Smith
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093-0411, USA.,Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, 92093-0213, USA
| | - Michael Holst
- Department of Mathematics, University of California, San Diego, La Jolla, CA, 92093-0112, USA.,Department of Physics, University of California, San Diego, La Jolla, CA, 92093-0424, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093-0411, USA.
| |
Collapse
|
10
|
Ferreira JV, Rosa Soares A, Ramalho JS, Ribeiro-Rodrigues T, Máximo C, Zuzarte M, Girão H, Pereira P. Exosomes and STUB1/CHIP cooperate to maintain intracellular proteostasis. PLoS One 2019; 14:e0223790. [PMID: 31613922 PMCID: PMC6794069 DOI: 10.1371/journal.pone.0223790] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/27/2019] [Indexed: 01/08/2023] Open
Abstract
Deregulation of proteostasis is a main feature of many age-related diseases, often leading to the accumulation of toxic oligomers and insoluble protein aggregates that accumulate intracellularly or in the extracellular space. To understand the mechanisms whereby toxic or otherwise unwanted proteins are secreted to the extracellular space, we inactivated the quality-control and proteostasis regulator ubiquitin ligase STUB1/CHIP. Data indicated that STUB1 deficiency leads both to the intracellular accumulation of protein aggregates and to an increase in the secretion of small extracellular vesicles (sEVs), including exosomes. Secreted sEVs are enriched in ubiquitinated and/or undegraded proteins and protein oligomers. Data also indicates that oxidative stress induces an increase in the release of sEVs in cells depleted from STUB1. Overall, the results presented here suggest that cells use exosomes to dispose of damaged and/or undegraded proteins as a means to reduce intracellular accumulation of proteotoxic material.
Collapse
Affiliation(s)
- Joao Vasco Ferreira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Ana Rosa Soares
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - José S. Ramalho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Teresa Ribeiro-Rodrigues
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Catarina Máximo
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Mónica Zuzarte
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Henrique Girão
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Paulo Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
- * E-mail:
| |
Collapse
|
11
|
Despres C, Di J, Cantrelle FX, Li Z, Huvent I, Chambraud B, Zhao J, Chen J, Chen S, Lippens G, Zhang F, Linhardt R, Wang C, Klärner FG, Schrader T, Landrieu I, Bitan G, Smet-Nocca C. Major Differences between the Self-Assembly and Seeding Behavior of Heparin-Induced and in Vitro Phosphorylated Tau and Their Modulation by Potential Inhibitors. ACS Chem Biol 2019; 14:1363-1379. [PMID: 31046227 PMCID: PMC6636790 DOI: 10.1021/acschembio.9b00325] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
Self-assembly of
the microtubule-associated protein tau into neurotoxic
oligomers, fibrils, and paired helical filaments, and cell-to-cell
spreading of these pathological tau species are critical processes
underlying the pathogenesis of Alzheimer’s disease and other
tauopathies. Modulating the self-assembly process and inhibiting formation
and spreading of such toxic species are promising strategies for therapy
development. A challenge in investigating tau self-assembly in vitro
is that, unlike most amyloidogenic proteins, tau does not aggregate
in the absence of posttranslational modifications (PTM), aggregation
inducers, or preformed seeds. The most common induction method is
addition of polyanions, such as heparin; yet, this artificial system
may not represent adequately tau self-assembly in vivo, which is driven
by aberrant phosphorylation and other PTMs, potentially leading to
in vitro data that do not reflect the behavior of tau and its interaction
with modulators in vivo. To tackle these challenges, methods for in
vitro phosphorylation of tau to produce aggregation-competent forms
recently have been introduced (Despres
et al. (2017) Proc. Natl. Acad. Sci. U.S.A., 114, 9080−908528784767). However, the oligomerization, seeding, and interaction
with assembly modulators of the different forms of tau have not been
studied to date. To address these knowledge gaps, we compared here
side-by-side the self-assembly and seeding activity of heparin-induced
tau with two forms of in vitro phosphorylated tau and tested how the
molecular tweezer CLR01, a negatively charged compound, affected these
processes. Tau was phosphorylated by incubation either with activated
extracellular signal-regulated kinase 2 or with a whole rat brain
extract. Seeding activity was measured using a fluorescence-resonance
energy transfer-based biosensor-cell method. We also used solution-state
NMR to investigate the binding sites of CLR01 on tau and how they
were impacted by phosphorylation. Our systematic structure–activity
relationship study demonstrates that heparin-induced tau behaves differently
from in vitro phosphorylated tau. The aggregation rates of the different
forms are distinct as is the intracellular localization of the induced
aggregates, which resemble brain-derived tau strains suggesting that
heparin-induced tau and in vitro phosphorylated tau have different
conformations, properties, and activities. CLR01 inhibits aggregation
and seeding of both heparin-induced and in vitro phosphorylated tau
dose-dependently, although heparin induction interferes with the interaction
between CLR01 and tau.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jianle Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Shiguo Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Guy Lippens
- Lille University CNRS UMR 8576, UGSF, F-59000 Lille, France
| | | | - Robert Linhardt
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | | | - Frank-Gerrit Klärner
- Institute of Organic Chemistry, University of Duisburg-Essen, 45141 Essen, Germany
| | - Thomas Schrader
- Institute of Organic Chemistry, University of Duisburg-Essen, 45141 Essen, Germany
| | | | | | | |
Collapse
|
12
|
Cascarina SM, Ross ED. Aggregation and degradation scales for prion-like domains: sequence features and context weigh in. Curr Genet 2018; 65:387-392. [PMID: 30310993 DOI: 10.1007/s00294-018-0890-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 12/13/2022]
Abstract
Protein aggregation in vivo is generally combated by extensive proteostatic defenses. Many proteostasis factors specifically recognize aggregation-prone features and re-fold or degrade the targeted protein. However, protein aggregation is not uncommon, suggesting that some proteins employ evasive strategies to aggregate in spite of the proteostasis machinery. Therefore, in addition to understanding the inherent aggregation propensity of protein sequences, it is important to understand how these sequences affect proteostatic recognition and regulation in vivo. In a recent study, we used a genetic mutagenesis and screening approach to explore the aggregation or degradation promoting effects of the canonical amino acids in the context of G-rich and Q/N-rich prion-like domains (PrLDs). Our results indicate that aggregation propensity scales are strongly influenced by the interplay between specific PrLD features and proteostatic recognition. Here, we briefly review these results and expand upon their potential implications. In addition, a preliminary exploration of the yeast proteome suggests that these proteostatic regulation heuristics may influence the compositional features of native G-rich and Q/N-rich domains in yeast. These results improve our understanding of the features affecting the aggregation and proteostatic regulation of prion-like domains in a cellular context, and suggest that the sequence space for native prion-like domains may be shaped by proteostatic constraints.
Collapse
Affiliation(s)
- Sean M Cascarina
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, 80523, CO, USA
| | - Eric D Ross
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, 80523, CO, USA.
| |
Collapse
|
13
|
Zacco E, Martin SR, Thorogate R, Pastore A. The RNA-Recognition Motifs of TAR DNA-Binding Protein 43 May Play a Role in the Aberrant Self-Assembly of the Protein. Front Mol Neurosci 2018; 11:372. [PMID: 30356856 PMCID: PMC6190850 DOI: 10.3389/fnmol.2018.00372] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
The TAR DNA-binding protein 43 (TDP-43) is a nucleic acid-binding protein implicated in gene regulation and RNA processing and shuffling. It is a ribonuclear protein that carries out most of its functions by binding specific nucleic acid sequences with its two RNA-recognition motifs, RRM1 and RRM2. TDP-43 has been identified in toxic cytosolic inclusions in neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). The unstructured C-terminus has prion-like behavior and has been considered the driver of the aberrant self-assembly of TDP-43. In this work, we set out to test the hypothesis that the RNA-binding domains could also play a role in protein aggregation. This knowledge could be of important value for understanding TDP-43 aberrant, disease-leading behavior and, in the future, inform the design of small molecules that could prevent or slow down protein aggregation by exploiting the RNA-binding properties of the protein. We investigated the behavior of the two tandem RRM domains separately and linked together and studied their self-assembly properties and RNA-binding ability with a number of biophysical techniques. The picture that emerges from our study suggests that this region of the protein plays an important and so far unexplored role in the aggregation of this protein.
Collapse
Affiliation(s)
- Elsa Zacco
- UK Dementia Research Institute, King's College London, London, United Kingdom.,The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | | | - Richard Thorogate
- London Centre for Nanotechnology, Faculty of Mathematical and Physical Sciences, University College London, London, United Kingdom
| | - Annalisa Pastore
- UK Dementia Research Institute, King's College London, London, United Kingdom.,The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
14
|
Cascarina SM, Paul KR, Machihara S, Ross ED. Sequence features governing aggregation or degradation of prion-like proteins. PLoS Genet 2018; 14:e1007517. [PMID: 30005071 PMCID: PMC6059496 DOI: 10.1371/journal.pgen.1007517] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/25/2018] [Accepted: 06/26/2018] [Indexed: 01/12/2023] Open
Abstract
Enhanced protein aggregation and/or impaired clearance of aggregates can lead to neurodegenerative disorders such as Alzheimer's Disease, Huntington's Disease, and prion diseases. Therefore, many protein quality control factors specialize in recognizing and degrading aggregation-prone proteins. Prions, which generally result from self-propagating protein aggregates, must therefore evade or outcompete these quality control systems in order to form and propagate in a cellular context. We developed a genetic screen in yeast that allowed us to explore the sequence features that promote degradation versus aggregation of a model glutamine/asparagine (Q/N)-rich prion domain from the yeast prion protein, Sup35, and two model glycine (G)-rich prion-like domains from the human proteins hnRNPA1 and hnRNPA2. Unexpectedly, we found that aggregation propensity and degradation propensity could be uncoupled in multiple ways. First, only a subset of classically aggregation-promoting amino acids elicited a strong degradation response in the G-rich prion-like domains. Specifically, large aliphatic residues enhanced degradation of the prion-like domains, whereas aromatic residues promoted prion aggregation without enhancing degradation. Second, the degradation-promoting effect of aliphatic residues was suppressed in the context of the Q/N-rich prion domain, and instead led to a dose-dependent increase in the frequency of spontaneous prion formation. Degradation suppression correlated with Q/N content of the surrounding prion domain, potentially indicating an underappreciated activity for these residues in yeast prion domains. Collectively, these results provide key insights into how certain aggregation-prone proteins may evade protein quality control degradation systems.
Collapse
Affiliation(s)
- Sean M. Cascarina
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kacy R. Paul
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Satoshi Machihara
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Eric D. Ross
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
15
|
Abbott NJ, Pizzo ME, Preston JE, Janigro D, Thorne RG. The role of brain barriers in fluid movement in the CNS: is there a 'glymphatic' system? Acta Neuropathol 2018; 135:387-407. [PMID: 29428972 DOI: 10.1007/s00401-018-1812-4] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/16/2018] [Accepted: 01/24/2018] [Indexed: 12/15/2022]
Abstract
Brain fluids are rigidly regulated to provide stable environments for neuronal function, e.g., low K+, Ca2+, and protein to optimise signalling and minimise neurotoxicity. At the same time, neuronal and astroglial waste must be promptly removed. The interstitial fluid (ISF) of the brain tissue and the cerebrospinal fluid (CSF) bathing the CNS are integral to this homeostasis and the idea of a glia-lymph or 'glymphatic' system for waste clearance from brain has developed over the last 5 years. This links bulk (convective) flow of CSF into brain along the outside of penetrating arteries, glia-mediated convective transport of fluid and solutes through the brain extracellular space (ECS) involving the aquaporin-4 (AQP4) water channel, and finally delivery of fluid to venules for clearance along peri-venous spaces. However, recent evidence favours important amendments to the 'glymphatic' hypothesis, particularly concerning the role of glia and transfer of solutes within the ECS. This review discusses studies which question the role of AQP4 in ISF flow and the lack of evidence for its ability to transport solutes; summarizes attributes of brain ECS that strongly favour the diffusion of small and large molecules without ISF flow; discusses work on hydraulic conductivity and the nature of the extracellular matrix which may impede fluid movement; and reconsiders the roles of the perivascular space (PVS) in CSF-ISF exchange and drainage. We also consider the extent to which CSF-ISF exchange is possible and desirable, the impact of neuropathology on fluid drainage, and why using CSF as a proxy measure of brain components or drug delivery is problematic. We propose that new work and key historical studies both support the concept of a perivascular fluid system, whereby CSF enters the brain via PVS convective flow or dispersion along larger caliber arteries/arterioles, diffusion predominantly regulates CSF/ISF exchange at the level of the neurovascular unit associated with CNS microvessels, and, finally, a mixture of CSF/ISF/waste products is normally cleared along the PVS of venules/veins as well as other pathways; such a system may or may not constitute a true 'circulation', but, at the least, suggests a comprehensive re-evaluation of the previously proposed 'glymphatic' concepts in favour of a new system better taking into account basic cerebrovascular physiology and fluid transport considerations.
Collapse
Affiliation(s)
- N Joan Abbott
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building 3.82, 150 Stamford St, London, SE1 9NH, UK.
| | - Michelle E Pizzo
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison School of Pharmacy, Madison, WI, USA
- Clinical Neuroengineering Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Jane E Preston
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building 3.82, 150 Stamford St, London, SE1 9NH, UK
| | - Damir Janigro
- Flocel Inc., Cleveland, OH, USA
- Department of Physiology, Case Western Reserve University, Cleveland, OH, USA
| | - Robert G Thorne
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison School of Pharmacy, Madison, WI, USA.
- Clinical Neuroengineering Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Cellular and Molecular Pathology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI, USA.
- , 5113 Rennebohm Hall, 777 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
16
|
SOD1 Mutations Causing Familial Amyotrophic Lateral Sclerosis Induce Toxicity in Astrocytes: Evidence for Bystander Effects in a Continuum of Astrogliosis. Neurochem Res 2018; 43:166-179. [DOI: 10.1007/s11064-017-2385-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/18/2022]
|
17
|
Platelet phosphorylated TDP-43: an exploratory study for a peripheral surrogate biomarker development for Alzheimer's disease. Future Sci OA 2017; 3:FSO238. [PMID: 29134122 PMCID: PMC5674277 DOI: 10.4155/fsoa-2017-0090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Aim: Alzheimer's disease (AD) and other forms of dementia create a noncurable disease population in world's societies. To develop a blood-based biomarker is important so that the remedial or disease-altering therapeutic intervention for AD patients would be available at the early stage. Materials & methods: TDP-43 levels were analyzed in postmortem brain tissue and platelets of AD and control subjects. Results: We observed an increased TDP-43 (<60%) in postmortem AD brain regions and similar trends were also observed in patient's platelets. Conclusion: Platelet TDP-43 could be used as a surrogate biomarker that is measurable, reproducible and sensitive for screening the patients with some early clinical signs of AD and can be used to monitor disease prognosis. In this study, we explore to identify an Alzheimer's disease (AD)-selective phospho-specific antibody that recognizes the diseased form of TDP-43 protein in patient's blood-derived platelets. Our results suggest that selective antiphosphorylated TDP-43 antibody discriminates AD from non-demented controls and patients with amyotrophic lateral sclerosis. Therefore, platelet screening with a selective antibody could potentially be a useful tool for diagnostic purposes for AD.
Collapse
|
18
|
Mathis CA, Lopresti BJ, Ikonomovic MD, Klunk WE. Small-molecule PET Tracers for Imaging Proteinopathies. Semin Nucl Med 2017; 47:553-575. [PMID: 28826526 DOI: 10.1053/j.semnuclmed.2017.06.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this chapter, we provide a review of the challenges and advances in developing successful PET imaging agents for 3 major types of aggregated amyloid proteins: amyloid-beta (Aβ), tau, and alpha-synuclein (α-syn). These 3 amyloids are involved in the pathogenesis of a variety of neurodegenerative diseases, referred to as proteinopathies or proteopathies, that include Alzheimer disease, Lewy body dementias, multiple system atrophy, and frontotemporal dementias, among others. In the Introduction section, we briefly discuss the history of amyloid in neurodegenerative diseases and describe why progress in developing effective imaging agents has been hampered by the failure of crystallography to provide definitive ligand-protein interactions for rational radioligand design efforts. Instead, the field has relied on largely serendipitous, trial-and-error methods to achieve useful and specific PET amyloid imaging tracers for Aβ, tau, and α-syn deposits. Because many of the proteopathies involve more than 1 amyloid protein, it is important to develop selective PET tracers for the different amyloids to help assess the relative contribution of each to total amyloid burden. We use Pittsburgh compound B to illustrate some of the critical steps in developing a potent and selective Aβ PET imaging agent. Other selective Aβ and tau PET imaging compounds have followed similar pathways in their developmental processes. Success for selective α-syn PET imaging agents has not been realized yet, but work is ongoing in multiple laboratories throughout the world. In the tau sections, we provide background regarding 3-repeat (3R) and 4-repeat (4R) tau proteins and how they can affect the binding of tau radioligands in different tauopathies. We review the ongoing efforts to assess the properties of tau ligands, which are useful in 3R, 4R, or combined 3R-4R tauopathies. Finally, we describe in the α-syn sections recent attempts to develop selective tracers to image α-synucleinopathies.
Collapse
Affiliation(s)
- Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA.
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
19
|
Cremades N, Dobson CM. The contribution of biophysical and structural studies of protein self-assembly to the design of therapeutic strategies for amyloid diseases. Neurobiol Dis 2017; 109:178-190. [PMID: 28709995 DOI: 10.1016/j.nbd.2017.07.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/26/2017] [Accepted: 07/10/2017] [Indexed: 01/26/2023] Open
Abstract
Many neurodegenerative disorders, including Alzheimer's, Parkinson's and the prion diseases, are characterized by a conformational conversion of normally soluble proteins or peptides into pathological species, by a process of misfolding and self-assembly that leads ultimately to the formation of amyloid fibrils. Recent studies support the idea that multiple intermediate species with a wide variety of degrees of neuronal toxicity are generated during such processes. The development of a high level of knowledge of the nature and structure of the pathogenic amyloid species would significantly enhance efforts to underline the molecular origins of these disorders and also to develop both accurate diagnoses and effective therapeutic interventions for these types of conditions. In this review, we discuss recent biophysical and structural information concerning different types of amyloid aggregates and the way in which such information can guide rational therapeutic approaches designed to target specific pathogenic events that occur during the development of these highly debilitating and increasingly common diseases.
Collapse
Affiliation(s)
- Nunilo Cremades
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Unit BIFI-IQFR(CSIC), Universidad de Zaragoza, Zaragoza 50018, Spain.
| | - Christopher M Dobson
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
20
|
Abstract
Prion diseases are characterized by the deposition of amyloids, misfolded conformers of the prion protein. The misfolded conformation is self-replicating, by a mechanism solely enciphered in the conformation of the protein. Because of low solubility and heterogeneous aggregate sizes, the detailed atomic structure of the infectious isoform is still unknown. Progress has, however, been made, and has allowed insights into the structural and disease-related mechanisms of prions. Many structural models have been proposed, and a number of them support a consensus trimeric β-helical model, significantly more complex than simple amyloid models. There is evidence that such complexity may be a necessary property of prion structure. Knowledge of the structure of prions will provide a greater understanding of the protein isoform conversion mechanism, and could eventually lead to rationally designed intervention strategies.
Collapse
Affiliation(s)
- Gerald Stubbs
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 53723
| | - Jan Stöhr
- Institute for Neurodegenerative Diseases, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
21
|
Endocytic uptake of monomeric amyloid-β peptides is clathrin- and dynamin-independent and results in selective accumulation of Aβ(1-42) compared to Aβ(1-40). Sci Rep 2017; 7:2021. [PMID: 28515429 PMCID: PMC5435687 DOI: 10.1038/s41598-017-02227-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 04/10/2017] [Indexed: 01/05/2023] Open
Abstract
Intraneuronal accumulation of amyloid-β (Aβ) peptides represent an early pathological feature in Alzheimer’s disease. We have therefore utilized flow cytometry and confocal microscopy in combination with endocytosis inhibition to explore the internalisation efficiency and uptake mechanisms of Aβ(1–40) and Aβ(1–42) monomers in cultured SH-SY5Y cells. We find that both variants are constitutively internalised via endocytosis and that their uptake is proportional to cellular endocytic rate. Moreover, SH-SY5Y cells internalise consistently twice the amount of Aβ(1–42) compared to Aβ(1–40); an imaging-based quantification showed that cells treated with 1 µM peptide for 8 h contained 800,000 peptides of Aβ(1–42) and 400,000 of Aβ(1–40). Both variants co-localised to >90% with lysosomes or other acidic compartments. Dynasore and chlorpromazine endocytosis inhibitors were both found to reduce uptake, particularly of Aβ(1–42). Overexpression of the C-terminal of the clathrin-binding domain of AP180, dynamin2 K44A, or Arf6 Q67L did however not reduce uptake of the Aβ variants. By contrast, perturbation of actin polymerisation and inhibition of macropinocytosis reduced Aβ(1–40) and Aβ(1–42) uptake considerably. This study clarifies mechanisms of Aβ(1–40) and Aβ(1–42) uptake, pinpoints differences between the two variants and highlights a common and putative role of macropinocytosis in the early accumulation of intraneuronal Aβ in AD.
Collapse
|
22
|
Lazarev VF, Mikhaylova ER, Guzhova IV, Margulis BA. Possible Function of Molecular Chaperones in Diseases Caused by Propagating Amyloid Aggregates. Front Neurosci 2017; 11:277. [PMID: 28559794 PMCID: PMC5433261 DOI: 10.3389/fnins.2017.00277] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 04/30/2017] [Indexed: 12/14/2022] Open
Abstract
The vast majority of neurodegenerative pathologies stem from the formation of toxic oligomers and aggregates composed of wrongly folded proteins. These protein complexes can be released from pathogenic cells and enthralled by other cells, causing the formation of new aggregates in a prion-like manner. By this mechanism, migrating complexes can transmit a disorder to distant regions of the brain and promote gradually transmitting degenerative processes. Molecular chaperones can counteract the toxicity of misfolded proteins. In this review, we discuss recent data on the possible cytoprotective functions of chaperones in horizontally transmitting neurological disorders.
Collapse
Affiliation(s)
- Vladimir F Lazarev
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of SciencesSt. Petersburg, Russia
| | - Elena R Mikhaylova
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of SciencesSt. Petersburg, Russia
| | - Irina V Guzhova
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of SciencesSt. Petersburg, Russia
| | - Boris A Margulis
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of SciencesSt. Petersburg, Russia
| |
Collapse
|
23
|
Lazarev VF, Mikhaylova ER, Dutysheva EA, Suezov RV, Guzhova IV, Margulis BA. A hydrocortisone derivative binds to GAPDH and reduces the toxicity of extracellular polyglutamine-containing aggregates. Biochem Biophys Res Commun 2017; 487:723-727. [PMID: 28450110 DOI: 10.1016/j.bbrc.2017.04.125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 04/23/2017] [Indexed: 10/19/2022]
Abstract
Huntington's disease (HD) has been recently shown to have a horizontally transmitted, prion-like pathology. Thus, the migration of polyglutamine-containing aggregates to acceptor cells is important for the progression of HD. These aggregates contain glyceraldehyde-3-phosphate dehydrogenase (GAPDH), which increases their intracellular transport and their toxicity. Here, we show that RX624, a derivative of hydrocortisone that binds to GAPDH, prevents the formation of aggregates of GAPDH-polyglutamine excreted into the culture medium by PC-12 rat cells expressing mutant huntingtin. RX624 was previously shown to be unable to penetrate cells and, thus, its principal therapeutic action might be the inhibition of polyglutamine-GAPDH complex aggregation in the extracellular matrix. The administration of RX624 to SH-SY5Y acceptor cells that incubated in conditioned medium from PC-12 cells expressing mutant huntingtin caused an approximately 20% increase in survival. This suggests that RX624 might be useful as a drug against polyglutamine pathologies, and that is could be administered exogenously without affecting target cell physiology. This protective effect was validated by the similar effect of an anti-GAPDH specific antibody.
Collapse
Affiliation(s)
- Vladimir F Lazarev
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Pr., 4, St. Petersburg, 194064, Russia.
| | - Elena R Mikhaylova
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Pr., 4, St. Petersburg, 194064, Russia.
| | - Elizaveta A Dutysheva
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Pr., 4, St. Petersburg, 194064, Russia.
| | - Roman V Suezov
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Pr., 4, St. Petersburg, 194064, Russia.
| | - Irina V Guzhova
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Pr., 4, St. Petersburg, 194064, Russia.
| | - Boris A Margulis
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Pr., 4, St. Petersburg, 194064, Russia.
| |
Collapse
|
24
|
Hoss F, Rodriguez-Alcazar JF, Latz E. Assembly and regulation of ASC specks. Cell Mol Life Sci 2017; 74:1211-1229. [PMID: 27761594 PMCID: PMC11107573 DOI: 10.1007/s00018-016-2396-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022]
Abstract
The inflammasome adapter ASC links activated inflammasome sensors to the effector molecule pro-caspase-1. Recruitment of pro-caspase-1 to ASC promotes the autocatalytic activation of caspase-1, which leads to the release of pro-inflammatory cytokines, such as IL-1β. Upon triggering of inflammasome sensors, ASC assembles into large helical fibrils that interact with each other serving as a supramolecular signaling platform termed the ASC speck. Alternative splicing, post-translational modifications of ASC, as well as interaction with other proteins can perturb ASC function. In several inflammatory diseases, ASC specks can be found in the extracellular space and its presence correlates with poor prognosis. Here, we review the role of ASC in inflammation, and focus on the structural mechanisms that lead to ASC speck formation, the regulation of ASC function during inflammasome assembly, and the importance of ASC specks in disease.
Collapse
Affiliation(s)
- Florian Hoss
- Institute of Innate Immunity, University Hospitals, University of Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany
| | - Juan F Rodriguez-Alcazar
- Institute of Innate Immunity, University Hospitals, University of Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals, University of Bonn, Sigmund-Freud-Straße 25, 53127, Bonn, Germany.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
- German Center for Neurodegenerative Diseases, Bonn, Germany.
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
25
|
Adegbuyiro A, Sedighi F, Pilkington AW, Groover S, Legleiter J. Proteins Containing Expanded Polyglutamine Tracts and Neurodegenerative Disease. Biochemistry 2017; 56:1199-1217. [PMID: 28170216 DOI: 10.1021/acs.biochem.6b00936] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several hereditary neurological and neuromuscular diseases are caused by an abnormal expansion of trinucleotide repeats. To date, there have been 10 of these trinucleotide repeat disorders associated with an expansion of the codon CAG encoding glutamine (Q). For these polyglutamine (polyQ) diseases, there is a critical threshold length of the CAG repeat required for disease, and further expansion beyond this threshold is correlated with age of onset and symptom severity. PolyQ expansion in the translated proteins promotes their self-assembly into a variety of oligomeric and fibrillar aggregate species that accumulate into the hallmark proteinaceous inclusion bodies associated with each disease. Here, we review aggregation mechanisms of proteins with expanded polyQ-tracts, structural consequences of expanded polyQ ranging from monomers to fibrillar aggregates, the impact of protein context and post-translational modifications on aggregation, and a potential role for lipid membranes in aggregation. As the pathogenic mechanisms that underlie these disorders are often classified as either a gain of toxic function or loss of normal protein function, some toxic mechanisms associated with mutant polyQ tracts will also be discussed.
Collapse
Affiliation(s)
- Adewale Adegbuyiro
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Faezeh Sedighi
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Albert W Pilkington
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Sharon Groover
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Justin Legleiter
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States.,Blanchette Rockefeller Neurosciences Institute, Robert C. Byrd Health Sciences Center, P.O. Box 9304, West Virginia University , Morgantown, West Virginia 26506, United States.,NanoSAFE, P.O. Box 6223, West Virginia University , Morgantown, West Virginia 26506, United States
| |
Collapse
|
26
|
Furman JL, Diamond MI. FRET and Flow Cytometry Assays to Measure Proteopathic Seeding Activity in Biological Samples. Methods Mol Biol 2017; 1523:349-359. [PMID: 27975263 DOI: 10.1007/978-1-4939-6598-4_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Transcellular propagation of protein aggregates-or seeds-is increasingly implicated as a mechanism for disease progression in many neurodegenerative disorders, including Alzheimer's disease and the related tauopathies. While neuropathology generally originates in one discrete brain region, pathology progresses as disease severity advances, often along discrete neural networks. The stereotypical spread of tau pathology suggests that cell-to-cell transfer of toxic protein aggregates could underlie disease progression, and recent studies implicate seeding as a proximal marker of disease, as compared to standard histological and biochemical analyses. Commonly used metrics for protein aggregation detection, however, lack sensitivity, are not quantitative, and/or undergo subjective classification. Here, we describe a FRET and flow cytometry cell-based assay that allows for rapid and quantitative detection of protein aggregates from human and rodent biological specimens.
Collapse
Affiliation(s)
- Jennifer L Furman
- Center for Alzheimer's and Neurodegenerative Diseases, Dallas, TX, USA.
- University of Texas-Southwestern Medical Center, 6000 Harry Hilned Blvd., NL 10.120K, Dallas, TX, 75390, USA.
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Dallas, TX, USA
- University of Texas-Southwestern Medical Center, 6000 Harry Hilned Blvd., NL 10.120K, Dallas, TX, 75390, USA
| |
Collapse
|
27
|
Braak H, Del Tredici K. Potential Pathways of Abnormal Tau and α-Synuclein Dissemination in Sporadic Alzheimer's and Parkinson's Diseases. Cold Spring Harb Perspect Biol 2016; 8:a023630. [PMID: 27580631 PMCID: PMC5088528 DOI: 10.1101/cshperspect.a023630] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Experimental data indicate that transneuronal propagation of abnormal protein aggregates in neurodegenerative proteinopathies, such as sporadic Alzheimer's disease (AD) and Parkinson's disease (PD), is capable of a self-propagating process that leads to a progression of neurodegeneration and accumulation of prion-like particles. The mechanisms by which misfolded tau and α-synuclein possibly spread from one involved nerve cell to the next in the neuronal chain to induce abnormal aggregation are still unknown. Based on findings from studies of human autopsy cases, we review potential pathways and mechanisms related to axonal and transneuronal dissemination of tau (sporadic AD) and α-synuclein (sporadic PD) aggregates between anatomically interconnected regions.
Collapse
Affiliation(s)
- Heiko Braak
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| |
Collapse
|
28
|
Abstract
A central dogma of molecular biology is that the sequence of a protein dictates its particular fold and the fold dictates its function. Indeed, the sequence → structure → function hypothesis has been a guiding principle by which scientists approach molecular biology. Every student knows that the genome encodes information for the progression from primary sequence to secondary, tertiary, and ultimately quaternary structure. Yet with a growing number of proteins, a fifth level has been identified: rearrangement of existing structures into distinct forms. Recent observations indicate that replication of Ebola virus depends on this fifth level. We believe other viruses with compact genomes and rapid evolution under selective pressure will be a rich source of examples of polypeptides that rearrange to gain added functions. In this review, we describe mechanisms by which viral, prokaryotic, and eukaryotic polypeptides have adopted alternate structures to control or gain function.
Collapse
Affiliation(s)
- Hal Wasserman
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037;
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037; .,Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
29
|
Sanders DW, Kaufman SK, Holmes BB, Diamond MI. Prions and Protein Assemblies that Convey Biological Information in Health and Disease. Neuron 2016; 89:433-48. [PMID: 26844828 DOI: 10.1016/j.neuron.2016.01.026] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prions derived from the prion protein (PrP) were first characterized as infectious agents that transmit pathology between individuals. However, the majority of cases of neurodegeneration caused by PrP prions occur sporadically. Proteins that self-assemble as cross-beta sheet amyloids are a defining pathological feature of infectious prion disorders and all major age-associated neurodegenerative diseases. In fact, multiple non-infectious proteins exhibit properties of template-driven self-assembly that are strikingly similar to PrP. Evidence suggests that like PrP, many proteins form aggregates that propagate between cells and convert cognate monomer into ordered assemblies. We now recognize that numerous proteins assemble into macromolecular complexes as part of normal physiology, some of which are self-amplifying. This review highlights similarities among infectious and non-infectious neurodegenerative diseases associated with prions, emphasizing the normal and pathogenic roles of higher-order protein assemblies. We propose that studies of the structural and cellular biology of pathological versus physiological aggregates will be mutually informative.
Collapse
Affiliation(s)
- David W Sanders
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, TX 75390, USA; Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - Sarah K Kaufman
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, TX 75390, USA; Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - Brandon B Holmes
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, TX 75390, USA; Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
30
|
Wan W, Wille H, Stöhr J, Kendall A, Bian W, McDonald M, Tiggelaar S, Watts JC, Prusiner SB, Stubbs G. Structural studies of truncated forms of the prion protein PrP. Biophys J 2016; 108:1548-1554. [PMID: 25809267 DOI: 10.1016/j.bpj.2015.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 11/19/2022] Open
Abstract
Prions are proteins that adopt self-propagating aberrant folds. The self-propagating properties of prions are a direct consequence of their distinct structures, making the understanding of these structures and their biophysical interactions fundamental to understanding prions and their related diseases. The insolubility and inherent disorder of prions have made their structures difficult to study, particularly in the case of the infectious form of the mammalian prion protein PrP. Many investigators have therefore preferred to work with peptide fragments of PrP, suggesting that these peptides might serve as structural and functional models for biologically active prions. We have used x-ray fiber diffraction to compare a series of different-sized fragments of PrP, to determine the structural commonalities among the fragments and the biologically active, self-propagating prions. Although all of the peptides studied adopted amyloid conformations, only the larger fragments demonstrated a degree of structural complexity approaching that of PrP. Even these larger fragments did not adopt the prion structure itself with detailed fidelity, and in some cases their structures were radically different from that of pathogenic PrP(Sc).
Collapse
Affiliation(s)
- William Wan
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee
| | - Holger Wille
- Institute for Neurodegenerative Diseases and Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Jan Stöhr
- Institute for Neurodegenerative Diseases and Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Amy Kendall
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee
| | - Wen Bian
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee
| | - Michele McDonald
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee
| | - Sarah Tiggelaar
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee
| | - Joel C Watts
- Institute for Neurodegenerative Diseases and Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases and Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Gerald Stubbs
- Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
31
|
Adam P, Křížková S, Heger Z, Babula P, Pekařík V, Vaculovičoá M, Gomes CM, Kizek R, Adam V. Metallothioneins in Prion- and Amyloid-Related Diseases. J Alzheimers Dis 2016; 51:637-56. [DOI: 10.3233/jad-150984] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Pavlína Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| | - Soňa Křížková
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| | - Zbyněk Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice, Brno, Czech Republic
| | - Vladimír Pekařík
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| | - Markéta Vaculovičoá
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| | - Cláudio M. Gomes
- Faculdade de Ciências Universidade de Lisboa, Biosystems and Integrative Sciences Institute and Department of Chemistry and Biochemistry, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - René Kizek
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| | - Vojtěch Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Technicka, Brno, Czech Republic
| |
Collapse
|
32
|
Internalized Tau Oligomers Cause Neurodegeneration by Inducing Accumulation of Pathogenic Tau in Human Neurons Derived from Induced Pluripotent Stem Cells. J Neurosci 2016; 35:14234-50. [PMID: 26490863 DOI: 10.1523/jneurosci.1523-15.2015] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Neuronal inclusions of hyperphosphorylated and aggregated tau protein are a pathological hallmark of several neurodegenerative tauopathies, including Alzheimer's disease (AD). The hypothesis of tau transmission in AD has emerged from histopathological studies of the spatial and temporal progression of tau pathology in postmortem patient brains. Increasing evidence in cellular and animal models supports the phenomenon of intercellular spreading of tau. However, the molecular and cellular mechanisms of pathogenic tau transmission remain unknown. The studies described herein investigate tau pathology propagation using human neurons derived from induced pluripotent stem cells. Neurons were seeded with full-length human tau monomers and oligomers and chronic effects on neuronal viability and function were examined over time. Tau oligomer-treated neurons exhibited an increase in aggregated and phosphorylated pathological tau. These effects were associated with neurite retraction, loss of synapses, aberrant calcium homeostasis, and imbalanced neurotransmitter release. In contrast, tau monomer treatment did not produce any measureable changes. This work supports the hypothesis that tau oligomers are toxic species that can drive the spread of tau pathology and neurodegeneration. SIGNIFICANCE STATEMENT Several independent studies have implicated tau protein as central to Alzheimer's disease progression and cell-to-cell pathology propagation. In this study, we investigated the ability of different tau species to propagate pathology in human neurons derived from induced pluripotent stem cells, which to date has not been shown. We demonstrated that tau oligomers, but not monomers, induce accumulation of pathological, hyperphosphorylated tau. This effect was accompanied with neurite degeneration, loss of synapses, aberrant calcium homeostasis, imbalanced neurotransmitter release, and ultimately with neuronal death. This study bridges various tau pathological phenotypes into a single and relevant induced pluripotent stem cell neuronal model of human disease that can be applied to the discovery of the mechanisms of tau-induced neurodegeneration.
Collapse
|
33
|
Mikhaylova ER, Lazarev VF, Nikotina AD, Margulis BA, Guzhova IV. Glyceraldehyde 3-phosphate dehydrogenase augments the intercellular transmission and toxicity of polyglutamine aggregates in a cell model of Huntington disease. J Neurochem 2016; 136:1052-63. [PMID: 26662373 DOI: 10.1111/jnc.13463] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/03/2015] [Accepted: 11/30/2015] [Indexed: 12/23/2022]
Abstract
The common feature of Huntington disease is the accumulation of oligomers or aggregates of mutant huntingtin protein (mHTT), which causes the death of a subset of striatal neuronal populations. The cytotoxic species can leave neurons and migrate to other groups of cells penetrating and damaging them in a prion-like manner. We hypothesized that the glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH), previously shown to elevate the aggregation of mHTT, is associated with an increased efficiency of intercellular propagation of mHTT. GAPDH, on its own or together with polyglutamine species, was shown to be released into the extracellular milieu mainly from dying cells as assessed by a novel enzyme immunoassay, western blotting, and ultrafiltration. The conditioned medium of cells with growing GAPDH-polyQ aggregates was toxic to naïve cells, whereas depletion of the aggregates from the medium lowered this cytotoxicity. The GAPDH component of the aggregates was found to increase their toxicity by two-fold in comparison with polyQ alone. Furthermore, GAPDH-polyQ complexes were shown to penetrate acceptor cells and to increase the capacity of polyQ to prionize its intracellular homolog containing a repeat of 25 glutamine residues. Finally, inhibitors of intracellular transport showed that polyQ-GAPDH complexes, as well as GAPDH itself, penetrated cells using clathrin-mediated endocytosis. This suggested a pivotal role of the enzyme in the intercellular transmission of Huntington disease pathogenicity. In conclusion, GAPDH occurring in complexes with polyglutamine strengthens the prion-like activity and toxicity of the migrating aggregates. Aggregating polygluatmine tracts were shown to release from the cells over-expressing mutant huntingtin in a complex with glyceraldehyde 3-phosphate dehydrogenase (GAPDH). The enzyme enhances the intracellular transport of aggregates to healthy cells, prionization of normal cellular proteins and finally cell death, thus demonstrating the pivotal role of GAPDH in the horizontal transmission of neurodegeneration.
Collapse
Affiliation(s)
- Elena R Mikhaylova
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Vladimir F Lazarev
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Alina D Nikotina
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Boris A Margulis
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Irina V Guzhova
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
34
|
Lasagna-Reeves CA, Rousseaux MW, Guerrero-Munoz MJ, Vilanova-Velez L, Park J, See L, Jafar-Nejad P, Richman R, Orr HT, Kayed R, Zoghbi HY. Ataxin-1 oligomers induce local spread of pathology and decreasing them by passive immunization slows Spinocerebellar ataxia type 1 phenotypes. eLife 2015; 4. [PMID: 26673892 PMCID: PMC4821792 DOI: 10.7554/elife.10891] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/15/2015] [Indexed: 11/29/2022] Open
Abstract
Previously, we reported that ATXN1 oligomers are the primary drivers of toxicity in Spinocerebellar ataxia type 1 (SCA1; Lasagna-Reeves et al., 2015). Here we report that polyQ ATXN1 oligomers can propagate locally in vivo in mice predisposed to SCA1 following intracerebral oligomeric tissue inoculation. Our data also show that targeting these oligomers with passive immunotherapy leads to some improvement in motor coordination in SCA1 mice and to a modest increase in their life span. These findings provide evidence that oligomer propagation is regionally limited in SCA1 and that immunotherapy targeting extracellular oligomers can mildly modify disease phenotypes. DOI:http://dx.doi.org/10.7554/eLife.10891.001
Collapse
Affiliation(s)
- Cristian A Lasagna-Reeves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | - Maxime Wc Rousseaux
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | | | - Luis Vilanova-Velez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | - Jeehye Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | - Lauren See
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | - Paymaan Jafar-Nejad
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | - Ronald Richman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minnesota, United States
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, United States
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United states
| |
Collapse
|
35
|
Furman JL, Holmes BB, Diamond MI. Sensitive Detection of Proteopathic Seeding Activity with FRET Flow Cytometry. J Vis Exp 2015:e53205. [PMID: 26710240 DOI: 10.3791/53205] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Increasing evidence supports transcellular propagation of toxic protein aggregates, or proteopathic seeds, as a mechanism for the initiation and progression of pathology in several neurodegenerative diseases, including Alzheimer's disease and the related tauopathies. The potentially critical role of tau seeds in disease progression strongly supports the need for a sensitive assay that readily detects seeding activity in biological samples. By combining the specificity of fluorescence resonance energy transfer (FRET), the sensitivity of flow cytometry, and the stability of a monoclonal cell line, an ultra-sensitive seeding assay has been engineered and is compatible with seed detection from recombinant or biological samples, including human and mouse brain homogenates. The assay employs monoclonal HEK 293T cells that stably express the aggregation-prone repeat domain (RD) of tau harboring the disease-associated P301S mutation fused to either CFP or YFP, which produce a FRET signal upon protein aggregation. The uptake of proteopathic tau seeds (but not other proteins) into the biosensor cells stimulates aggregation of RD-CFP and RD-YFP, and flow cytometry sensitively and quantitatively monitors this aggregation-induced FRET. The assay detects femtomolar concentrations (monomer equivalent) of recombinant tau seeds, has a dynamic range spanning three orders of magnitude, and is compatible with brain homogenates from tauopathy transgenic mice and human tauopathy subjects. With slight modifications, the assay can also detect seeding activity of other proteopathic seeds, such as α-synuclein, and is also compatible with primary neuronal cultures. The ease, sensitivity, and broad applicability of FRET flow cytometry makes it useful to study a wide range of protein aggregation disorders.
Collapse
Affiliation(s)
- Jennifer L Furman
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center;
| | - Brandon B Holmes
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center
| |
Collapse
|
36
|
Ludolph AC, Brettschneider J. TDP-43 in amyotrophic lateral sclerosis - is it a prion disease? Eur J Neurol 2015; 22:753-61. [PMID: 25846565 DOI: 10.1111/ene.12706] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/06/2015] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis is a devastating disease characterized by rapidly progressive paresis. The neuropathological hallmark of most amyotrophic lateral sclerosis cases are neuronal and glial aggregates of phosphorylated 43-kDa TAR DNA-binding protein (pTDP-43). The accumulation of similar proteins into insoluble aggregates is now recognized as a common pathological hallmark of neurodegenerative diseases in general. Importantly, many of these proteins such as tau and amyloid-β in Alzheimer's disease and α-synuclein in Parkinson's show a stereotypical sequential distribution pattern with progressing disease. In this review, we discuss recent evidence that TDP-43 in ALS may propagate similarly to other neurodegenerative disease proteins. We furthermore delineate similarities and important differences of TDP-43 proteinopathies to prion diseases.
Collapse
Affiliation(s)
- A C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany
| | | |
Collapse
|
37
|
Abstract
Proteostasis is maintained by multiple cellular pathways, including protein synthesis, quality control, and degradation. An imbalance of neuronal proteostasis, associated with protein misfolding and aggregation, leads to proteinopathies or neurodegeneration. While genetic variations and protein modifications contribute to aggregate formation, components of the proteostasis network dictate the fate of protein aggregates. Here we provide an overview of proteostasis pathways and their interplay (particularly autophagy) with the metabolism of disease-related proteins. We review recent studies on neuronal activity-mediated regulation of proteostasis and transcellular propagation of protein aggregates in the nervous system. Targeting proteostasis pathways therapeutically remains an attractive but challenging task.
Collapse
Affiliation(s)
- Junghyun Lim
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
38
|
Spreading of pathology in neurodegenerative diseases: a focus on human studies. Nat Rev Neurosci 2015; 16:109-20. [PMID: 25588378 DOI: 10.1038/nrn3887] [Citation(s) in RCA: 572] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The progression of many neurodegenerative diseases is thought to be driven by the template-directed misfolding, seeded aggregation and cell-cell transmission of characteristic disease-related proteins, leading to the sequential dissemination of pathological protein aggregates. Recent evidence strongly suggests that the anatomical connections made by neurons - in addition to the intrinsic characteristics of neurons, such as morphology and gene expression profile - determine whether they are vulnerable to degeneration in these disorders. Notably, this common pathogenic principle opens up opportunities for pursuing novel targets for therapeutic interventions for these neurodegenerative disorders. We review recent evidence that supports the notion of neuron-neuron protein propagation, with a focus on neuropathological and positron emission tomography imaging studies in humans.
Collapse
|
39
|
Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan M, Zimmer S, Monks B, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmüller W, Latz E. The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation. Nat Immunol 2014; 15:727-37. [PMID: 24952505 PMCID: PMC4116676 DOI: 10.1038/ni.2913] [Citation(s) in RCA: 613] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/01/2014] [Indexed: 12/15/2022]
Abstract
Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1β (IL-1β) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1β. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1β in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.
Collapse
Affiliation(s)
- Bernardo S. Franklin
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
| | - Lukas Bossaller
- Department of Immunology and Rheumatology, Hannover Medical School, 30625 Hannover, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, USA
| | - Dominic De Nardo
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
| | - Jacqueline M. Ratter
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
| | - Andrea Stutz
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
| | - Gudrun Engels
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
| | | | - Mark Nordhoff
- German Center for Neurodegenerative Diseases, 53175, Bonn, Germany
| | | | - Ashraf Al-Amoudi
- German Center for Neurodegenerative Diseases, 53175, Bonn, Germany
| | - Matthew Mangan
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
- German Center for Neurodegenerative Diseases, 53175, Bonn, Germany
| | - Sebastian Zimmer
- Department of Medicine/Cardiology, University of Bonn, 53105, Bonn, Germany
| | - Brian Monks
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, USA
| | - Martin Fricke
- Department of Immunology and Rheumatology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold E. Schmidt
- Department of Immunology and Rheumatology, Hannover Medical School, 30625 Hannover, Germany
| | - Terje Espevik
- Center of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, NTNU, Trondheim 7491, Norway
| | - Bernadette Jones
- The University of Newcastle & Vaccines, Infection, Viruses & Asthma (VIVA), Hunter Medical Research Institute, Newcastle, Australia
| | - Andrew G. Jarnicki
- The University of Newcastle & Vaccines, Infection, Viruses & Asthma (VIVA), Hunter Medical Research Institute, Newcastle, Australia
| | - Philip M. Hansbro
- The University of Newcastle & Vaccines, Infection, Viruses & Asthma (VIVA), Hunter Medical Research Institute, Newcastle, Australia
| | - Patricia Busto
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, USA
| | - Ann Marshak-Rothstein
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, USA
| | - Simone Hornemann
- University Hospital Zürich, Institute of Neuropathology, CH–8091 Zürich, Switzerland
| | - Adriano Aguzzi
- University Hospital Zürich, Institute of Neuropathology, CH–8091 Zürich, Switzerland
| | - Wolfgang Kastenmüller
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals, University of Bonn, 53127, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, USA
- German Center for Neurodegenerative Diseases, 53175, Bonn, Germany
- Center of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, NTNU, Trondheim 7491, Norway
| |
Collapse
|
40
|
Sanders DW, Kaufman SK, DeVos SL, Sharma AM, Mirbaha H, Li A, Barker SJ, Foley AC, Thorpe JR, Serpell LC, Miller TM, Grinberg LT, Seeley WW, Diamond MI. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 2014; 82:1271-88. [PMID: 24857020 DOI: 10.1016/j.neuron.2014.04.047] [Citation(s) in RCA: 743] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2014] [Indexed: 12/13/2022]
Abstract
Prion-like propagation of tau aggregation might underlie the stereotyped progression of neurodegenerative tauopathies. True prions stably maintain unique conformations ("strains") in vivo that link structure to patterns of pathology. We now find that tau meets this criterion. Stably expressed tau repeat domain indefinitely propagates distinct amyloid conformations in a clonal fashion in culture. Reintroduction of tau from these lines into naive cells reestablishes identical clones. We produced two strains in vitro that induce distinct pathologies in vivo as determined by successive inoculations into three generations of transgenic mice. Immunopurified tau from these mice recreates the original strains in culture. We used the cell system to isolate tau strains from 29 patients with 5 different tauopathies, finding that different diseases are associated with different sets of strains. Tau thus demonstrates essential characteristics of a prion. This might explain the phenotypic diversity of tauopathies and could enable more effective diagnosis and therapy.
Collapse
Affiliation(s)
- David W Sanders
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Sarah K Kaufman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Sarah L DeVos
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Apurwa M Sharma
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Hilda Mirbaha
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Aimin Li
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Scarlett J Barker
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Alex C Foley
- School of Life Sciences, University of Sussex, Falmer BN1 9QG, UK
| | - Julian R Thorpe
- School of Life Sciences, University of Sussex, Falmer BN1 9QG, UK
| | - Louise C Serpell
- School of Life Sciences, University of Sussex, Falmer BN1 9QG, UK
| | - Timothy M Miller
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA
| | - Lea T Grinberg
- Department of Neurology and Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William W Seeley
- Department of Neurology and Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marc I Diamond
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63105, USA.
| |
Collapse
|
41
|
Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013; 501:45-51. [PMID: 24005412 DOI: 10.1038/nature12481] [Citation(s) in RCA: 1200] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/17/2013] [Indexed: 12/12/2022]
Abstract
For several decades scientists have speculated that the key to understanding age-related neurodegenerative disorders may be found in the unusual biology of the prion diseases. Recently, owing largely to the advent of new disease models, this hypothesis has gained experimental momentum. In a remarkable variety of diseases, specific proteins have been found to misfold and aggregate into seeds that structurally corrupt like proteins, causing them to aggregate and form pathogenic assemblies ranging from small oligomers to large masses of amyloid. Proteinaceous seeds can therefore serve as self-propagating agents for the instigation and progression of disease. Alzheimer's disease and other cerebral proteopathies seem to arise from the de novo misfolding and sustained corruption of endogenous proteins, whereas prion diseases can also be infectious in origin. However, the outcome in all cases is the functional compromise of the nervous system, because the aggregated proteins gain a toxic function and/or lose their normal function. As a unifying pathogenic principle, the prion paradigm suggests broadly relevant therapeutic directions for a large class of currently intractable diseases.
Collapse
Affiliation(s)
- Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076 Tübingen, Germany.
| | | |
Collapse
|
42
|
Propagation of tau pathology in Alzheimer's disease: identification of novel therapeutic targets. ALZHEIMERS RESEARCH & THERAPY 2013; 5:49. [PMID: 24152385 PMCID: PMC3978816 DOI: 10.1186/alzrt214] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Accumulation and aggregation of the microtubule-associated protein tau are a pathological hallmark of neurodegenerative disorders such as Alzheimer’s disease (AD). In AD, tau becomes abnormally phosphorylated and forms inclusions throughout the brain, starting in the entorhinal cortex and progressively affecting additional brain regions as the disease progresses. Formation of these inclusions is thought to lead to synapse loss and cell death. Tau is also found in the cerebrospinal fluid (CSF), and elevated levels are a biomarker for AD. Until recently, it was thought that the presence of tau in the CSF was due to the passive release of aggregated tau from dead or dying tangle-bearing neurons. However, accumulating evidence from different AD model systems suggests that tau is actively secreted and transferred between synaptically connected neurons. Transgenic mouse lines with localized expression of aggregating human tau in the entorhinal cortex have demonstrated that, as these animals age, tau becomes mislocalized from axons to cell bodies and dendrites and that human tau-positive aggregates form first in the entorhinal cortex and later in downstream projection targets. Numerous in vitro and in vivo studies have provided insight into the mechanisms by which tau may be released and internalized by neurons and have started to provide insight into how tau pathology may spread in AD. In this review, we discuss the evidence for regulated tau release and its specific uptake by neurons. Furthermore, we identify possible therapeutic targets for preventing the propagation of tau pathology, as inhibition of tau transfer may restrict development of tau tangles in a small subset of neurons affected in early stages of AD and therefore prevent widespread neuron loss and cognitive dysfunction associated with later stages of the disease.
Collapse
|
43
|
Xiao X, Cali I, Dong Z, Puoti G, Yuan J, Qing L, Wang H, Kong Q, Gambetti P, Zou WQ. Protease-sensitive prions with 144-bp insertion mutations. Aging (Albany NY) 2013; 5:155-73. [PMID: 23515139 PMCID: PMC3629288 DOI: 10.18632/aging.100543] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Insertion of 144-base pair (bp) containing six extra octapeptide repeats between residues 51 and 91 of prion protein (PrP) gene is associated with inherited prion diseases. Most cases linked to this insertion examined by Western blotting showed detectable proteinase K-resistant PrPSc (rPrPSc) resembling PrPSc type 1 and type 2 in sporadic Creutzfeldt-Jakob disease (sCJD), or PrP7-8 in Gerstmann-Sträussler-Scheinker disease. However, cases lacking detectable rPrPSc also have been reported. Which PrP conformer is associated with neuropathological changes in the cases without detectable rPrPSc remains to be determined. Here we report that while all six but one subjects with the 144-bp insertion mutations examined display the pathognomonic PrP patches in the cerebellum, one of them exhibits no detectable typical rPrPSc even in PrPSc-enriched preparations. Instead, a large amount of abnormal PrP is captured from this case by gene 5 protein and sodium phosphotungstate, reagents that have been proved to specifically capture abnormal PrP. All captured abnormal PrP from the cerebellum and other brain regions is virtually sensitive to PK-digestion (termed sPrPSc). The presence of the predominant sPrPSc but absence of rPrPSc in this 144-bp insertion-linked inherited CJD case suggests that mutant sPrPSc is the main component of the PrP deposit patches and sPrPSc is sufficient to cause neurotoxicity and prion disease.
Collapse
Affiliation(s)
- Xiangzhu Xiao
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wan W, Bian W, McDonald M, Kijac A, Wemmer DE, Stubbs G. Heterogeneous seeding of a prion structure by a generic amyloid form of the fungal prion-forming domain HET-s(218-289). J Biol Chem 2013; 288:29604-12. [PMID: 23986444 DOI: 10.1074/jbc.m113.505511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The fungal prion-forming domain HET-s(218-289) forms infectious amyloid fibrils at physiological pH that were shown by solid-state NMR to be assemblies of a two-rung β-solenoid structure. Under acidic conditions, HET-s(218-289) has been shown to form amyloid fibrils that have very low infectivity in vivo, but structural information about these fibrils has been very limited. We show by x-ray fiber diffraction that the HET-s(218-289) fibrils formed under acidic conditions have a stacked β-sheet architecture commonly found in short amyloidogenic peptides and denatured protein aggregates. At physiological pH, stacked β-sheet fibrils nucleate the formation of the infectious β-solenoid prions in a process of heterogeneous seeding, but do so with kinetic profiles distinct from those of spontaneous or homogeneous (seeded with infectious β-solenoid fibrils) fibrillization. Several serial passages of stacked β-sheet-seeded solutions lead to fibrillization kinetics similar to homogeneously seeded solutions. Our results directly show that structural mutation can occur between substantially different amyloid architectures, lending credence to the suggestion that the processes of strain adaptation and crossing species barriers are facilitated by structural mutation.
Collapse
Affiliation(s)
- William Wan
- From the Department of Biological Sciences and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37235-1634
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
The pathway leading from soluble and monomeric to hyperphosphorylated, insoluble and filamentous tau protein is at the centre of many human neurodegenerative diseases, collectively referred to as tauopathies. Dominantly inherited mutations in MAPT, the gene that encodes tau, cause forms of frontotemporal dementia and parkinsonism, proving that dysfunction of tau is sufficient to cause neurodegeneration and dementia. However, most cases of tauopathy are not inherited in a dominant manner. The first tau aggregates form in a few nerve cells in discrete brain areas. These become self propagating and spread to distant brain regions in a prion-like manner. The prevention of tau aggregation and propagation is the focus of attempts to develop mechanism-based treatments for tauopathies.
Collapse
Affiliation(s)
- Maria Grazia Spillantini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
46
|
Margulis BA, Vigont V, Lazarev VF, Kaznacheyeva EV, Guzhova IV. Pharmacological protein targets in polyglutamine diseases: mutant polypeptides and their interactors. FEBS Lett 2013; 587:1997-2007. [PMID: 23684638 DOI: 10.1016/j.febslet.2013.05.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 12/18/2022]
Abstract
Polyglutamine diseases are a group of pathologies affecting different parts of the brain and causing dysfunction and atrophy of certain neural cell populations. These diseases stem from mutations in various cellular genes that result in the synthesis of proteins with extended polyglutamine tracts. In particular, this concerns huntingtin, ataxins, and androgen receptor. These mutant proteins can form oligomers, aggregates, and, finally, aggresomes with distinct functions and different degrees of cytotoxicity. In this review, we analyze the effects of different forms of polyQ proteins on other proteins and their functions, which are considered as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Boris A Margulis
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky pr., 4, St. Petersburg 194064, Russia
| | | | | | | | | |
Collapse
|
47
|
Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proc Natl Acad Sci U S A 2013; 110:9535-40. [PMID: 23690619 DOI: 10.1073/pnas.1301175110] [Citation(s) in RCA: 575] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Filamentous inclusions made of hyperphosphorylated tau are characteristic of numerous human neurodegenerative diseases, including Alzheimer's disease, tangle-only dementia, Pick disease, argyrophilic grain disease (AGD), progressive supranuclear palsy, and corticobasal degeneration. In Alzheimer's disease and AGD, it has been shown that filamentous tau appears to spread in a stereotypic manner as the disease progresses. We previously demonstrated that the injection of brain extracts from human mutant P301S tau-expressing transgenic mice into the brains of mice transgenic for wild-type human tau (line ALZ17) resulted in the assembly of wild-type human tau into filaments and the spreading of tau inclusions from the injection sites to anatomically connected brain regions. Here we injected brain extracts from humans who had died with various tauopathies into the hippocampus and cerebral cortex of ALZ17 mice. Argyrophilic tau inclusions formed in all cases and following the injection of the corresponding brain extracts, we recapitulated the hallmark lesions of AGD, PSP and CBD. Similar inclusions also formed after intracerebral injection of brain homogenates from human tauopathies into nontransgenic mice. Moreover, the induced formation of tau aggregates could be propagated between mouse brains. These findings suggest that once tau aggregates have formed in discrete brain areas, they become self-propagating and spread in a prion-like manner.
Collapse
|
48
|
Tanaka K, Matsuda N. Proteostasis and neurodegeneration: the roles of proteasomal degradation and autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:197-204. [PMID: 23523933 DOI: 10.1016/j.bbamcr.2013.03.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/01/2013] [Accepted: 03/10/2013] [Indexed: 11/30/2022]
Abstract
All proteins in a cell continuously turn over, each at its own rate, contributing to a cell's development, differentiation, or aging. Of course, unnecessary protein(s), or those synthesized in excess, that hamper cellular homeostasis should be discarded rapidly. Furthermore, cells that have been subjected to various environmental stresses, e.g., reactive oxygen species (ROS) and UV irradiation, may incur various types of protein damage, which vitiate normal and homeostatic functions in the cell. Thereby, the prompt elimination of impaired proteins is essential for cell viability. This housekeeping is accomplished by two major catabolic routes-proteasomal digestion and autophagy. Strict maintenance of proteostasis is particularly important in non-proliferative cells, especially neurons, and it is plausible that its failure leads to a number of the neurodegenerative diseases becoming prominent in the growing elderly population. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
Affiliation(s)
- Keiji Tanaka
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo 156-8506, Japan.
| | | |
Collapse
|