1
|
Esquivel AR, Hill SE, Blair LJ. DnaJs are enriched in tau regulators. Int J Biol Macromol 2023; 253:127486. [PMID: 37852393 PMCID: PMC10842427 DOI: 10.1016/j.ijbiomac.2023.127486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
The aberrant accumulation of tau protein is implicated as a pathogenic factor in many neurodegenerative diseases. Tau seeding may underlie its predictable spread in these diseases. Molecular chaperones can modulate tau pathology, but their effects have mainly been studied in isolation. This study employed a semi-high throughput assay to identify molecular chaperones influencing tau seeding using Tau RD P301S FRET Biosensor cells, which express a portion of tau containing the frontotemporal dementia-related P301S tau mutation fused to a FRET biosensor. Approximately fifty chaperones from five major families were screened using live cell imaging to monitor FRET-positive tau seeding. Among the tested chaperones, five exhibited significant effects on tau in the primary screen. Notably, three of these were from the DnaJ family. In subsequent studies, overexpression of DnaJA2, DnaJB1, and DnaJB6b resulted in significant reductions in tau levels. Knockdown experiments by shRNA revealed an inverse correlation between DnaJB1 and DnaJB6b with tau levels. DnaJB6b overexpression, specifically, reduced total tau levels in a cellular model with a pre-existing pool of tau, partially through enhanced proteasomal degradation. Further, DnaJB6b interacted with tau complexes. These findings highlight the potent chaperone activity within the DnaJ family, particularly DnaJB6b, towards tau.
Collapse
Affiliation(s)
- Abigail R Esquivel
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Shannon E Hill
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Laura J Blair
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA; Research Service, James A Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA.
| |
Collapse
|
2
|
Nadel CM, Thwin AC, Callahan M, Lee K, Connelly E, Craik CS, Southworth DR, Gestwicki JE. The E3 Ubiquitin Ligase, CHIP/STUB1, Inhibits Aggregation of Phosphorylated Proteoforms of Microtubule-associated Protein Tau (MAPT). J Mol Biol 2023; 435:168026. [PMID: 37330289 PMCID: PMC10491737 DOI: 10.1016/j.jmb.2023.168026] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 06/19/2023]
Abstract
Hyper-phosphorylated tau accumulates as insoluble fibrils in Alzheimer's disease (AD) and related dementias. The strong correlation between phosphorylated tau and disease has led to an interest in understanding how cellular factors discriminate it from normal tau. Here, we screen a panel of chaperones containing tetratricopeptide repeat (TPR) domains to identify those that might selectively interact with phosphorylated tau. We find that the E3 ubiquitin ligase, CHIP/STUB1, binds 10-fold more strongly to phosphorylated tau than unmodified tau. The presence of even sub-stoichiometric concentrations of CHIP strongly suppresses aggregation and seeding of phosphorylated tau. We also find that CHIP promotes rapid ubiquitination of phosphorylated tau, but not unmodified tau, in vitro. Binding to phosphorylated tau requires CHIP's TPR domain, but the binding mode is partially distinct from the canonical one. In cells, CHIP restricts seeding by phosphorylated tau, suggesting that it could be an important barrier in cell-to-cell spreading. Together, these findings show that CHIP recognizes a phosphorylation-dependent degron on tau, establishing a pathway for regulating the solubility and turnover of this pathological proteoform.
Collapse
Affiliation(s)
- Cory M Nadel
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Aye C Thwin
- Biochemistry & Biophysics and the University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Matthew Callahan
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Kanghyun Lee
- Biochemistry & Biophysics and the University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA
| | - Emily Connelly
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA
| | - Charles S Craik
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA
| | - Daniel R Southworth
- Biochemistry & Biophysics and the University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA.
| | - Jason E Gestwicki
- Departments of Pharmaceutical Chemistry and University of California San Francisco, San Francisco, CA 94508, USA; Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA 94508, USA.
| |
Collapse
|
3
|
Hill SE, Beaulieu-Abdelahad D, Lemus A, Webster JM, Ospina SR, Darling AL, Martin MD, Patel S, Bridenstine L, Swonger R, Paul S, Blackburn R, Calcul L, Dickey CA, Leahy JW, Blair LJ. Benzothiazole Substitution Analogs of Rhodacyanine Hsp70 Inhibitors Modulate Tau Accumulation. ACS Chem Biol 2023; 18:1124-1135. [PMID: 37144894 PMCID: PMC10443619 DOI: 10.1021/acschembio.2c00919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The accumulation and aggregation of the microtubule-associated protein tau (tau) into intracellular neuronal tangles are a hallmark of a range of progressive neurodegenerative tauopathies, including Alzheimer's disease (AD), frontotemporal dementia, Pick's disease, and progressive supranuclear palsy. The aberrant phosphorylation of tau is associated with tau aggregates in AD. Members of the heat shock protein 70 kDa (Hsp70) family of chaperones bind directly to tau and modulate tau clearance and aggregation. Small molecules that inhibit the Hsp70 family of chaperones have been shown to reduce the accumulation of tau, including phosphorylated tau. Here, eight analogs of the rhodacyanine inhibitor, JG-98, were synthesized and evaluated. Like JG-98, many of the compounds inhibited ATPase activity of the cytosolic heat shock cognate 70 protein (Hsc70) and reduced total, aggregated, and phosphorylated tau accumulation in cultured cells. Three compounds, representing divergent clogP values, were evaluated for in vivo blood-brain barrier penetration and tau reduction in an ex vivo brain slice model. AL69, the compound with the lowest clogP and the lowest membrane retention in a parallel artificial membrane permeability assay (PAMPA), reduced phosphorylated tau accumulation. Our results suggest that benzothiazole substitutions of JG-98 that increase hydrophilicity may increase the efficacy of these Hsp70 inhibitors to reduce phosphorylated tau.
Collapse
Affiliation(s)
- Shannon E. Hill
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - David Beaulieu-Abdelahad
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Andrea Lemus
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, USA
| | - Jack M. Webster
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Santiago Rodriguez Ospina
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - April L. Darling
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Mackenzie D. Martin
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Shreya Patel
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, USA
| | - Liznair Bridenstine
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, USA
| | - Ronald Swonger
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, USA
| | - Steven Paul
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Roy Blackburn
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Laurent Calcul
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, USA
| | - Chad A. Dickey
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
- Research Service, James A Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - James W. Leahy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, USA
- Center for Drug Discovery and Innovation, University of South Florida, 3720 Spectrum Boulevard, Suite 303, Tampa, Florida 33612, USA
| | - Laura J. Blair
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
- Research Service, James A Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
4
|
Rai S, Tapadia MG. Hsc70-4 aggravates PolyQ-mediated neurodegeneration by modulating NF-κB mediated immune response in Drosophila. Front Mol Neurosci 2022; 15:857257. [PMID: 36425218 PMCID: PMC9678916 DOI: 10.3389/fnmol.2022.857257] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 10/11/2022] [Indexed: 10/06/2023] Open
Abstract
Huntington's disease occurs when the stretch of CAG repeats in exon 1 of the huntingtin (htt) gene crosses the permissible limit, causing the mutated protein (mHtt) to form insoluble aggregates or inclusion bodies. These aggregates are non-typically associated with various essential proteins in the cells, thus disrupting cellular homeostasis. The cells try to bring back normalcy by synthesizing evolutionary conserved cellular chaperones, and Hsp70 is one of the families of heat shock proteins that has a significant part in this, which comprises of heat-inducible and cognate forms. Here, we demonstrate that the heat shock cognate (Hsc70) isoform, Hsc70-4/HSPA8, has a distinct role in polyglutamate (PolyQ)-mediated pathogenicity, and its expression is enhanced in the polyQ conditions in Drosophila. Downregulation of hsc70-4 rescues PolyQ pathogenicity with a notable improvement in the ommatidia arrangement and near-normal restoration of optic neurons leading to improvement in phototaxis response. Reduced hsc70-4 also attenuates the augmented immune response by decreasing the expression of NF-κB and the antimicrobial peptides, along with that JNK overactivation is also restored. These lead to the rescue of the photoreceptor cells, indicating a decrease in the caspase activity, thus reverting the PolyQ pathogenicity. At the molecular level, we show the interaction between Hsc70-4, Polyglutamine aggregates, and NF-κB, which may be responsible for the dysregulation of signaling molecules in polyQ conditions. Thus, the present data provides a functional link between Hsc70-4 and NF-κB under polyQ conditions.
Collapse
Affiliation(s)
| | - Madhu G. Tapadia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
5
|
Parcerisas A, Ortega-Gascó A, Pujadas L, Soriano E. The Hidden Side of NCAM Family: NCAM2, a Key Cytoskeleton Organization Molecule Regulating Multiple Neural Functions. Int J Mol Sci 2021; 22:10021. [PMID: 34576185 PMCID: PMC8471948 DOI: 10.3390/ijms221810021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023] Open
Abstract
Although it has been over 20 years since Neural Cell Adhesion Molecule 2 (NCAM2) was identified as the second member of the NCAM family with a high expression in the nervous system, the knowledge of NCAM2 is still eclipsed by NCAM1. The first studies with NCAM2 focused on the olfactory bulb, where this protein has a key role in axonal projection and axonal/dendritic compartmentalization. In contrast to NCAM1, NCAM2's functions and partners in the brain during development and adulthood have remained largely unknown until not long ago. Recent studies have revealed the importance of NCAM2 in nervous system development. NCAM2 governs neuronal morphogenesis and axodendritic architecture, and controls important neuron-specific processes such as neuronal differentiation, synaptogenesis and memory formation. In the adult brain, NCAM2 is highly expressed in dendritic spines, and it regulates synaptic plasticity and learning processes. NCAM2's functions are related to its ability to adapt to the external inputs of the cell and to modify the cytoskeleton accordingly. Different studies show that NCAM2 interacts with proteins involved in cytoskeleton stability and proteins that regulate calcium influx, which could also modify the cytoskeleton. In this review, we examine the evidence that points to NCAM2 as a crucial cytoskeleton regulation protein during brain development and adulthood. This key function of NCAM2 may offer promising new therapeutic approaches for the treatment of neurodevelopmental diseases and neurodegenerative disorders.
Collapse
Affiliation(s)
- Antoni Parcerisas
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Basic Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Spain
| | - Alba Ortega-Gascó
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Lluís Pujadas
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
6
|
Irwin R, Faust O, Petrovic I, Wolf SG, Hofmann H, Rosenzweig R. Hsp40s play complementary roles in the prevention of tau amyloid formation. eLife 2021; 10:69601. [PMID: 34369377 PMCID: PMC8437434 DOI: 10.7554/elife.69601] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/08/2021] [Indexed: 11/21/2022] Open
Abstract
The microtubule-associated protein, tau, is the major subunit of neurofibrillary tangles associated with neurodegenerative conditions, such as Alzheimer's disease. In the cell, however, tau aggregation can be prevented by a class of proteins known as molecular chaperones. While numerous chaperones are known to interact with tau, though, little is known regarding the mechanisms by which these prevent tau aggregation. Here, we describe the effects of ATP-independent Hsp40 chaperones, DNAJA2 and DNAJB1, on tau amyloid-fiber formation and compare these to the small heat shock protein HSPB1. We find that the chaperones play complementary roles, with each preventing tau aggregation differently and interacting with distinct sets of tau species. Whereas HSPB1 only binds tau monomers, DNAJB1 and DNAJA2 recognize aggregation-prone conformers and even mature fibers. In addition, we find that both Hsp40s bind tau seeds and fibers via their C-terminal domain II (CTDII), with DNAJA2 being further capable of recognizing tau monomers by a second, distinct site in CTDI. These results lay out the mechanisms by which the diverse members of the Hsp40 family counteract the formation and propagation of toxic tau aggregates and highlight the fact that chaperones from different families/classes play distinct, yet complementary roles in preventing pathological protein aggregation. Several neurological conditions, such as Alzheimer’s and Parkinson’s disease, are characterized by the build-up of protein clumps known as aggregates. In the case of Alzheimer’s disease, a key protein, called tau, aggregates to form fibers that are harmful to neuronal cells in the brain. One of the ways our cells can prevent this from occurring is through the action of proteins known as molecular chaperones, which can bind to tau proteins and prevent them from sticking together. Tau can take on many forms. For example, a single tau protein on its own, known as a monomer, is unstructured. In patients with Alzheimer’s, these monomers join together into small clusters, known as seeds, that rapidly aggregate and accumulate into rigid, structured fibers. One chaperone, HSPB1, is known to bind to tau monomers and prevent them from being incorporated into fibers. Recently, another group of chaperones, called J-domain proteins, was also found to interact with tau. However, it was unclear how these chaperones prevent aggregation and whether they bind to tau in a similar manner as HSPB1. To help answer this question, Irwin, Faust et al. studied the effect of two J-domain proteins, as well as the chaperone HSBP1, on tau aggregation. This revealed that, unlike HSBP1, the two J-domain proteins can bind to multiple forms of tau, including when it has already aggregated in to seeds and fibers. This suggests that these chaperones can stop the accumulation of fibers at several different stages of the aggregation process. Further experiments examining which sections of the J-domain proteins bind to tau, showed that both attach to fibers via the same region. However, the two J-domain proteins are not identical in their interaction with tau. While one of them uses a distinct region to bind to tau monomers, the other does not bind to single tau proteins at all. These results demonstrate how different cellular chaperones can complement one another in order to inhibit harmful protein aggregation. Further studies will be needed to understand the full role of J-domain proteins in preventing tau from accumulating into fibers, as well as their potential as drug targets for developing new treatments.
Collapse
Affiliation(s)
- Rose Irwin
- Weizmann Institute of Science, Rehovot, Israel
| | - Ofrah Faust
- Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | |
Collapse
|
7
|
Raj K, Akundi RS. Mutant Ataxin-3-Containing Aggregates (MATAGGs) in Spinocerebellar Ataxia Type 3: Dynamics of the Disorder. Mol Neurobiol 2021; 58:3095-3118. [PMID: 33629274 DOI: 10.1007/s12035-021-02314-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/25/2021] [Indexed: 11/25/2022]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is the most common type of SCA worldwide caused by abnormal polyglutamine expansion in the coding region of the ataxin-3 gene. Ataxin-3 is a multi-faceted protein involved in various cellular processes such as deubiquitination, cytoskeletal organisation, and transcriptional regulation. The presence of an expanded poly(Q) stretch leads to altered processing and misfolding of the protein culminating in the production of insoluble protein aggregates in the cell. Various post-translational modifications affect ataxin-3 fibrillation and aggregation. This review provides an exhaustive assessment of the various pathogenic mechanisms undertaken by the mutant ataxin-3-containing aggregates (MATAGGs) for disease induction and neurodegeneration. This includes in-depth discussion on MATAGG dynamics including their formation, role in neuronal pathogenesis, and the debate over the toxic v/s protective nature of the MATAGGs in disease progression. Additionally, the currently available therapeutic strategies against SCA3 have been reviewed. The shift in the focus of such strategies, from targeting the steps that lead to or reduce aggregate formation to targeting the expression of mutant ataxin-3 itself via RNA-based therapeutics, has also been presented. We also discuss the intriguing promise that various growth and neurotrophic factors, especially the insulin pathway, hold in the modulation of SCA3 progression. These emerging areas show the newer directions through which SCA3 can be targeted including various preclinical and clinical trials. All these advances made in the last three decades since the discovery of the ataxin-3 gene have been critically reviewed here.
Collapse
Affiliation(s)
- Kritika Raj
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, Chanakyapuri, New Delhi, 110021, India
| | - Ravi Shankar Akundi
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, Chanakyapuri, New Delhi, 110021, India.
| |
Collapse
|
8
|
Shao H, Li X, Hayashi S, Bertron JL, Schwarz DMC, Tang BC, Gestwicki JE. Inhibitors of heat shock protein 70 (Hsp70) with enhanced metabolic stability reduce tau levels. Bioorg Med Chem Lett 2021; 41:128025. [PMID: 33839251 DOI: 10.1016/j.bmcl.2021.128025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/30/2021] [Accepted: 04/04/2021] [Indexed: 11/30/2022]
Abstract
The molecular chaperone, Heat Shock Protein 70 (Hsp70), is an emerging drug target for neurodegenerative diseases, because of its ability to promote degradation of microtubule-associated protein tau (MAPT/tau). Recently, we reported YM-08 as a brain penetrant, allosteric Hsp70 inhibitor, which reduces tau levels. However, the benzothiazole moiety of YM-08 is vulnerable to metabolism by CYP3A4, limiting its further application as a chemical probe. In this manuscript, we designed and synthesized seventeen YM-08 derivatives by systematically introducing halogen atoms to the benzothiazole ring and shifting the position of the heteroatom in a distal pyridine. In microsome assays, we found that compound JG-23 has 12-fold better metabolic stability and it retained the ability to reduce tau levels in two cell-based models. These chemical probes of Hsp70 are expected to be useful tools for studying tau homeostasis.
Collapse
Affiliation(s)
- Hao Shao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Xiaokai Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States; Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA 94158, United States
| | - Shigenari Hayashi
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA 94158, United States
| | - Jeanette L Bertron
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States; Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA 94158, United States
| | - Daniel M C Schwarz
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States; Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA 94158, United States
| | - Benjamin C Tang
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA 94158, United States
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States; Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, CA 94158, United States.
| |
Collapse
|
9
|
Sandusky-Beltran LA, Kovalenko A, Placides DS, Ratnasamy K, Ma C, Hunt JB, Liang H, Calahatian JIT, Michalski C, Fahnestock M, Blair LJ, Darling AL, Baker JD, Fontaine SN, Dickey CA, Gamsby JJ, Nash KR, Abner E, Selenica MLB, Lee DC. Aberrant AZIN2 and polyamine metabolism precipitates tau neuropathology. J Clin Invest 2021; 131:126299. [PMID: 33586680 PMCID: PMC7880423 DOI: 10.1172/jci126299] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/16/2020] [Indexed: 01/14/2023] Open
Abstract
Tauopathies display a spectrum of phenotypes from cognitive to affective behavioral impairments; however, mechanisms promoting tau pathology and how tau elicits behavioral impairment remain unclear. We report a unique interaction between polyamine metabolism, behavioral impairment, and tau fate. Polyamines are ubiquitous aliphatic molecules that support neuronal function, axonal integrity, and cognitive processing. Transient increases in polyamine metabolism hallmark the cell's response to various insults, known as the polyamine stress response (PSR). Dysregulation of gene transcripts associated with polyamine metabolism in Alzheimer's disease (AD) brains were observed, and we found that ornithine decarboxylase antizyme inhibitor 2 (AZIN2) increased to the greatest extent. We showed that sustained AZIN2 overexpression elicited a maladaptive PSR in mice with underlying tauopathy (MAPT P301S; PS19). AZIN2 also increased acetylpolyamines, augmented tau deposition, and promoted cognitive and affective behavioral impairments. Higher-order polyamines displaced microtubule-associated tau to facilitate polymerization but also decreased tau seeding and oligomerization. Conversely, acetylpolyamines promoted tau seeding and oligomers. These data suggest that tauopathies launch an altered enzymatic signature that endorses a feed-forward cycle of disease progression. Taken together, the tau-induced PSR affects behavior and disease continuance, but may also position the polyamine pathway as a potential entry point for plausible targets and treatments of tauopathy, including AD.
Collapse
Affiliation(s)
- Leslie A. Sandusky-Beltran
- Byrd Alzheimer’s Institute and
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida, USA
| | - Andrii Kovalenko
- Byrd Alzheimer’s Institute and
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida, USA
| | - Devon S. Placides
- Byrd Alzheimer’s Institute and
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida, USA
| | - Kevin Ratnasamy
- Byrd Alzheimer’s Institute and
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida, USA
| | - Chao Ma
- Byrd Alzheimer’s Institute and
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
- Sanders-Brown Center on Aging
| | - Jerry B. Hunt
- Sanders-Brown Center on Aging
- Department of Neuroscience
| | - Huimin Liang
- Sanders-Brown Center on Aging
- Department of Neuroscience
| | - John Ivan T. Calahatian
- Byrd Alzheimer’s Institute and
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida, USA
| | - Camilla Michalski
- Department of Psychiatry & Behavioral Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Margaret Fahnestock
- Department of Psychiatry & Behavioral Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Laura J. Blair
- Byrd Alzheimer’s Institute and
- Department of Molecular Medicine and
| | - April L. Darling
- Byrd Alzheimer’s Institute and
- Department of Molecular Medicine and
| | - Jeremy D. Baker
- Byrd Alzheimer’s Institute and
- Department of Molecular Medicine and
| | | | - Chad A. Dickey
- Byrd Alzheimer’s Institute and
- Department of Molecular Medicine and
| | - Joshua J. Gamsby
- Byrd Alzheimer’s Institute and
- Department of Molecular Medicine and
| | - Kevin R. Nash
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Erin Abner
- Sanders-Brown Center on Aging
- Department of Epidemiology, and
| | - Maj-Linda B. Selenica
- Sanders-Brown Center on Aging
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Daniel C. Lee
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, Florida, USA
- Sanders-Brown Center on Aging
- Department of Neuroscience
| |
Collapse
|
10
|
Johnson SL, Ranxhi B, Libohova K, Tsou WL, Todi SV. Ubiquitin-interacting motifs of ataxin-3 regulate its polyglutamine toxicity through Hsc70-4-dependent aggregation. eLife 2020; 9:60742. [PMID: 32955441 PMCID: PMC7505662 DOI: 10.7554/elife.60742] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2020] [Indexed: 12/17/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) belongs to the family of polyglutamine neurodegenerations. Each disorder stems from the abnormal lengthening of a glutamine repeat in a different protein. Although caused by a similar mutation, polyglutamine disorders are distinct, implicating non-polyglutamine regions of disease proteins as regulators of pathogenesis. SCA3 is caused by polyglutamine expansion in ataxin-3. To determine the role of ataxin-3’s non-polyglutamine domains in disease, we utilized a new, allelic series of Drosophila melanogaster. We found that ataxin-3 pathogenicity is saliently controlled by polyglutamine-adjacent ubiquitin-interacting motifs (UIMs) that enhance aggregation and toxicity. UIMs function by interacting with the heat shock protein, Hsc70-4, whose reduction diminishes ataxin-3 toxicity in a UIM-dependent manner. Hsc70-4 also enhances pathogenicity of other polyglutamine proteins. Our studies provide a unique insight into the impact of ataxin-3 domains in SCA3, identify Hsc70-4 as a SCA3 enhancer, and indicate pleiotropic effects from HSP70 chaperones, which are generally thought to suppress polyglutamine degeneration.
Collapse
Affiliation(s)
- Sean L Johnson
- Department of Pharmacology, Wayne State University, Detroit, United States
| | - Bedri Ranxhi
- Department of Pharmacology, Wayne State University, Detroit, United States
| | - Kozeta Libohova
- Department of Pharmacology, Wayne State University, Detroit, United States
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University, Detroit, United States
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University, Detroit, United States.,Department of Neurology, Wayne State University, Detroit, United States
| |
Collapse
|
11
|
De La-Rocque S, Moretto E, Butnaru I, Schiavo G. Knockin' on heaven's door: Molecular mechanisms of neuronal tau uptake. J Neurochem 2020; 156:563-588. [PMID: 32770783 PMCID: PMC8432157 DOI: 10.1111/jnc.15144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
Since aggregates of the microtubule‐binding protein tau were found to be the main component of neurofibrillary tangles more than 30 years ago, their contribution to neurodegeneration in Alzheimer's disease (AD) and tauopathies has become well established. Recent work shows that both tau load and its distribution in the brain of AD patients correlate with cognitive decline more closely compared to amyloid plaque deposition. In addition, the amyloid cascade hypothesis has been recently challenged because of disappointing results of clinical trials designed to treat AD by reducing beta‐amyloid levels, thus fuelling a renewed interest in tau. There is now robust evidence to indicate that tau pathology can spread within the central nervous system via a prion‐like mechanism following a stereotypical pattern, which can be explained by the trans‐synaptic inter‐neuronal transfer of pathological tau. In the receiving neuron, tau has been shown to take multiple routes of internalisation, which are partially dependent on its conformation and aggregation status. Here, we review the emerging mechanisms proposed for the uptake of extracellular tau in neurons and the requirements for the propagation of its pathological conformers, addressing how they gain access to physiological tau monomers in the cytosol. Furthermore, we highlight some of the key mechanistic gaps of the field, which urgently need to be addressed to expand our understanding of tau propagation and lead to the identification of new therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Samantha De La-Rocque
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edoardo Moretto
- UK Dementia Research Institute, University College London, London, UK
| | - Ioana Butnaru
- UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
12
|
Jung HY, Kim W, Hahn KR, Kwon HJ, Nam SM, Chung JY, Yoon YS, Kim DW, Yoo DY, Hwang IK. Effects of Pyridoxine Deficiency on Hippocampal Function and Its Possible Association with V-Type Proton ATPase Subunit B2 and Heat Shock Cognate Protein 70. Cells 2020; 9:cells9051067. [PMID: 32344819 PMCID: PMC7290376 DOI: 10.3390/cells9051067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Pyridoxine, one of the vitamin B6 vitamers, plays a crucial role in amino acid metabolism and synthesis of monoamines as a cofactor. In the present study, we observed the effects of pyridoxine deficiency on novel object recognition memory. In addition, we examined the levels of 5-hydroxytryptamine (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), 3,4-dihydroxyphenethylamine (DA), 3,4-dihydroxyphenylacetic acid, and homovanillic acid and the number of proliferating cells and neuroblasts in the hippocampus. We also examined the effects of pyridoxine deficiency on protein profiles applying a proteomic study. Five-week-old mice fed pyridoxine-deficient diets for 8 weeks and showed a significant decrease in the serum and brain (cerebral cortex, hippocampus, and thalamus) levels of pyridoxal 5′-phosphate, a catalytically active form of vitamin-B6, and decline in 5-HT and DA levels in the hippocampus compared to controls fed a normal chow. In addition, pyridoxine deficiency significantly decreased Ki67-positive proliferating cells and differentiated neuroblasts in the dentate gyrus compared to controls. A proteomic study demonstrated that a total of 41 spots were increased or decreased more than two-fold. Among the detected proteins, V-type proton ATPase subunit B2 (ATP6V1B2) and heat shock cognate protein 70 (HSC70) showed coverage and matching peptide scores. Validation by Western blot analysis showed that ATP6V1B2 and HSC70 levels were significantly decreased and increased, respectively, in pyridoxine-deficient mice compared to controls. These results suggest that pyridoxine is an important element of novel object recognition memory, monoamine levels, and hippocampal neurogenesis. Pyridoxine deficiency causes cognitive impairments and reduction in 5-HT and DA levels, which may be associated with a reduction of ATP6V1B2 and elevation of HSC70 levels in the hippocampus.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
| | - Woosuk Kim
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea;
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Sung Min Nam
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul 05030, Korea;
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Dae Young Yoo
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
- Correspondence: (D.Y.Y.); (I.K.H.)
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
- Correspondence: (D.Y.Y.); (I.K.H.)
| |
Collapse
|
13
|
Arginine π-stacking drives binding to fibrils of the Alzheimer protein Tau. Nat Commun 2020; 11:571. [PMID: 31996674 PMCID: PMC6989696 DOI: 10.1038/s41467-019-13745-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/15/2019] [Indexed: 01/26/2023] Open
Abstract
Aggregation of the Tau protein into fibrils defines progression of neurodegenerative diseases, including Alzheimer’s Disease. The molecular basis for potentially toxic reactions of Tau aggregates is poorly understood. Here we show that π-stacking by Arginine side-chains drives protein binding to Tau fibrils. We mapped an aggregation-dependent interaction pattern of Tau. Fibrils recruit specifically aberrant interactors characterised by intrinsically disordered regions of atypical sequence features. Arginine residues are key to initiate these aberrant interactions. Crucial for scavenging is the guanidinium group of its side chain, not its charge, indicating a key role of π-stacking chemistry for driving aberrant fibril interactions. Remarkably, despite the non-hydrophobic interaction mode, the molecular chaperone Hsp90 can modulate aberrant fibril binding. Together, our data present a molecular mode of action for derailment of protein-protein interaction by neurotoxic fibrils. Tau fibril formation is a hallmark of Alzheimer’s disease. Here the authors reveal an aggregation-dependent protein interaction pattern of Tau and further show that π-stacking of the arginine side-chains drives aberrant protein binding to Tau fibrils.
Collapse
|
14
|
Ajit D, Trzeciakiewicz H, Tseng JH, Wander CM, Chen Y, Ajit A, King DP, Cohen TJ. A unique tau conformation generated by an acetylation-mimic substitution modulates P301S-dependent tau pathology and hyperphosphorylation. J Biol Chem 2019; 294:16698-16711. [PMID: 31543505 DOI: 10.1074/jbc.ra119.009674] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/12/2019] [Indexed: 01/10/2023] Open
Abstract
Abnormal intracellular accumulation of aggregated tau is a hallmark feature of Alzheimer's disease and other tauopathies. Pathological tau can undergo a range of post-translational modifications (PTMs) that are implicated as triggers of disease pathology. Recent studies now indicate that tau acetylation, in particular, controls both microtubule binding and tau aggregation, thereby acting as a central regulator of tau's biochemical properties and providing avenues to exploit for potential therapies. Here, using cell-based assays and tau transgenic mice harboring an acetylation-mimic mutation at residue Lys-280 (K280Q), we evaluated whether this substitution modifies the neurodegenerative disease pathology associated with the aggregate-prone tau P301S variant. Strikingly, the addition of a K280Q-substituted variant altered P301S-mediated tau conformation and reduced tau hyperphosphorylation. We further evaluated neurodegeneration markers in K280Q acetylation-mimic mice and observed reduced neuroinflammation as well as restored levels of N-methyl-d-aspartate receptors and post-synaptic markers compared with the parental mice. Thus, substituting a single lysine residue in the context of a P301S disease-linked mutation produces a unique tau species that abrogates some of the cardinal features of tauopathy. The findings of our study indicate that a complex tau PTM code likely regulates tau pathogenesis, highlighting the potential utility of manipulating and detoxifying tau strains through site-specific tau-targeting approaches.
Collapse
Affiliation(s)
- Deepa Ajit
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Hanna Trzeciakiewicz
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Jui-Heng Tseng
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Connor M Wander
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Youjun Chen
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Aditi Ajit
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Diamond P King
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Todd J Cohen
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|
15
|
Abstract
The accumulation of tau filaments in neurons is a pathological hallmark of various neurodegenerative diseases, including Alzheimer's disease. However, it is not the filamentous aggregates themselves, but non-filamentous tau species, tau oligomer, that is thought to be the culprit in tau-mediated neurodegeneration. The definition of and methodology for isolating tau oligomers vary among researchers. Here we describe how tau oligomers are identified, summarize the differences of tau oligomers among research groups, and discuss their hypothesized functions.
Collapse
|
16
|
Baughman HER, Clouser AF, Klevit RE, Nath A. HspB1 and Hsc70 chaperones engage distinct tau species and have different inhibitory effects on amyloid formation. J Biol Chem 2018; 293:2687-2700. [PMID: 29298892 DOI: 10.1074/jbc.m117.803411] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 12/15/2017] [Indexed: 11/06/2022] Open
Abstract
The microtubule-associated protein tau forms insoluble, amyloid-type aggregates in various dementias, most notably Alzheimer's disease. Cellular chaperone proteins play important roles in maintaining protein solubility and preventing aggregation in the crowded cellular environment. Although tau is known to interact with numerous chaperones, it remains unclear how these chaperones function mechanistically to prevent tau aggregation and how chaperones from different classes compare in terms of mechanism. Here, we focused on the small heat shock protein HspB1 (also known as Hsp27) and the constitutive chaperone Hsc70 (also known as HspA8) and report how each chaperone interacts with tau to prevent its fibril formation. Using fluorescence and NMR spectroscopy, we show that the two chaperones inhibit tau fibril formation by distinct mechanisms. HspB1 delayed tau fibril formation by weakly interacting with early species in the aggregation process, whereas Hsc70 was highly efficient at preventing tau fibril elongation, possibly by capping the ends of tau fibrils. Both chaperones recognized aggregation-prone motifs within the microtubule-binding repeat region of tau. However, HspB1 binding remained transient in both aggregation-promoting and non-aggregating conditions, whereas Hsc70 binding was significantly tighter under aggregation-promoting conditions. These differences highlight the fact that chaperones from different families play distinct but complementary roles in the prevention of pathological protein aggregation.
Collapse
Affiliation(s)
- Hannah E R Baughman
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195-7610
| | - Amanda F Clouser
- Department of Biochemistry, University of Washington, Seattle, Washington 98195-7350
| | - Rachel E Klevit
- Department of Biochemistry, University of Washington, Seattle, Washington 98195-7350.
| | - Abhinav Nath
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195-7610.
| |
Collapse
|
17
|
Young ZT, Mok SA, Gestwicki JE. Therapeutic Strategies for Restoring Tau Homeostasis. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a024612. [PMID: 28159830 DOI: 10.1101/cshperspect.a024612] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Normal tau homeostasis is achieved when the synthesis, processing, and degradation of the protein is balanced. Together, the pathways that regulate tau homeostasis ensure that the protein is at the proper levels and that its posttranslational modifications and subcellular localization are appropriately controlled. These pathways include the enzymes responsible for posttranslational modifications, those systems that regulate mRNA splicing, and the molecular chaperones that control tau turnover and its binding to microtubules. In tauopathies, this delicate balance is disturbed. Tau becomes abnormally modified by posttranslational modification, it loses affinity for microtubules, and it accumulates in proteotoxic aggregates. How and why does this imbalance occur? In this review, we discuss how molecular chaperones and other components of the protein homeostasis (e.g., proteostasis) network normally govern tau quality control. We also discuss how aging might reduce the capacity of these systems and how tau mutations might further affect this balance. Finally, we discuss how small-molecule inhibitors are being used to probe and perturb the tau quality-control systems, playing a particularly prominent role in revealing the logic of tau homeostasis. As such, there is now interest in developing these chemical probes into therapeutics, with the goal of restoring normal tau homeostasis to treat disease.
Collapse
Affiliation(s)
- Zapporah T Young
- Institute for Neurodegenerative Disease, Department of Pharmaceutical Chemistry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Sue Ann Mok
- Institute for Neurodegenerative Disease, Department of Pharmaceutical Chemistry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Jason E Gestwicki
- Institute for Neurodegenerative Disease, Department of Pharmaceutical Chemistry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
18
|
Woo JA, Liu T, Zhao X, Trotter C, Yrigoin K, Cazzaro S, Narvaez ED, Khan H, Witas R, Bukhari A, Makati K, Wang X, Dickey C, Kang DE. Enhanced tau pathology via RanBP9 and Hsp90/Hsc70 chaperone complexes. Hum Mol Genet 2017; 26:3973-3988. [PMID: 29016855 PMCID: PMC6075219 DOI: 10.1093/hmg/ddx284] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/04/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Accumulation of amyloid β (Aβ) and tau represent the two major pathological hallmarks of Alzheimer's disease (AD). Despite the critical importance of Aβ accumulation as an early event in AD pathogenesis, multiple lines of evidence indicate that tau is required to mediate Aβ-induced neurotoxic signals in neurons. We have previously shown that the scaffolding protein Ran-binding protein 9 (RanBP9), which is highly elevated in brains of AD and AD mouse models, both enhances Aβ production and mediates Aβ-induced neurotoxicity. However, it is unknown whether and how RanBP9 transmits Aβ-induced neurotoxic signals to tau. Here we show for the first time that overexpression or knockdown of RanBP9 directly enhances and reduces tau levels, respectively, in vitro and in vivo. Such changes in tau levels are associated with the ability of RanBP9 to physically interact with tau and heat shock protein 90/heat shock cognate 70 (Hsp90/Hsc70) complexes. Meanwhile, both RanBP9 and tau levels are simultaneously reduced by Hsp90 or Hsc70 inhibitors, whereas overexpression or knockdown of RanBP9 significantly diminishes the anti-tau potency of Hsp90/Hsc70 inhibitors as well as Hsc70 variants (WT & E175S). Further, RanBP9 increases the capacity for Hsp90 and Hsc70 complexes to bind ATP and enhances their ATPase activities in vitro. These observations in vitro and cell lines are recapitulated in primary neurons and in vivo, as genetic reduction in RanBP9 not only ameliorates tauopathy in Tau-P301S mice but also rescues the deficits in synaptic integrity and plasticity.
Collapse
Affiliation(s)
- Jung A Woo
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Medicine
| | - Tian Liu
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Medicine
| | - Xingyu Zhao
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Medicine
| | - Courtney Trotter
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Medicine
| | | | | | | | | | | | | | | | - Xinming Wang
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL 33613, USA
| | - Chad Dickey
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Medicine
- James A. Haley Veteran’s Administration Hospital, Research Division, Tampa, FL 33612, USA
| | - David E Kang
- USF Health Byrd Alzheimer’s Institute
- Department of Molecular Medicine
- James A. Haley Veteran’s Administration Hospital, Research Division, Tampa, FL 33612, USA
| |
Collapse
|
19
|
Shorter J. Engineering therapeutic protein disaggregases. Mol Biol Cell 2017; 27:1556-60. [PMID: 27255695 PMCID: PMC4865313 DOI: 10.1091/mbc.e15-10-0693] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/23/2016] [Indexed: 11/11/2022] Open
Abstract
Therapeutic agents are urgently required to cure several common and fatal neurodegenerative disorders caused by protein misfolding and aggregation, including amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and Alzheimer's disease (AD). Protein disaggregases that reverse protein misfolding and restore proteins to native structure, function, and localization could mitigate neurodegeneration by simultaneously reversing 1) any toxic gain of function of the misfolded form and 2) any loss of function due to misfolding. Potentiated variants of Hsp104, a hexameric AAA+ ATPase and protein disaggregase from yeast, have been engineered to robustly disaggregate misfolded proteins connected with ALS (e.g., TDP-43 and FUS) and PD (e.g., α-synuclein). However, Hsp104 has no metazoan homologue. Metazoa possess protein disaggregase systems distinct from Hsp104, including Hsp110, Hsp70, and Hsp40, as well as HtrA1, which might be harnessed to reverse deleterious protein misfolding. Nevertheless, vicissitudes of aging, environment, or genetics conspire to negate these disaggregase systems in neurodegenerative disease. Thus, engineering potentiated human protein disaggregases or isolating small-molecule enhancers of their activity could yield transformative therapeutics for ALS, PD, and AD.
Collapse
Affiliation(s)
- James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
20
|
Jackrel ME, Shorter J. Protein-Remodeling Factors As Potential Therapeutics for Neurodegenerative Disease. Front Neurosci 2017; 11:99. [PMID: 28293166 PMCID: PMC5328956 DOI: 10.3389/fnins.2017.00099] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/15/2017] [Indexed: 12/13/2022] Open
Abstract
Protein misfolding is implicated in numerous neurodegenerative disorders including amyotrophic lateral sclerosis, Parkinson's disease, Alzheimer's disease, and Huntington's disease. A unifying feature of patients with these disorders is the accumulation of deposits comprised of misfolded protein. Aberrant protein folding can cause toxicity through a loss or gain of protein function, or both. An intriguing therapeutic approach to counter these disorders is the application of protein-remodeling factors to resolve these misfolded conformers and return the proteins to their native fold and function. Here, we describe the application of protein-remodeling factors to alleviate protein misfolding in neurodegenerative disease. We focus on Hsp104, Hsp110/Hsp70/Hsp40, NMNAT, and HtrA1, which can prevent and reverse protein aggregation. While many of these protein-remodeling systems are highly promising, their activity can be limited. Thus, engineering protein-remodeling factors to enhance their activity could be therapeutically valuable. Indeed, engineered Hsp104 variants suppress neurodegeneration in animal models, which opens the way to novel therapeutics and mechanistic probes to help understand neurodegenerative disease.
Collapse
Affiliation(s)
- Meredith E Jackrel
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania Philadelphia, PA, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
21
|
Khanna MR, Kovalevich J, Lee VMY, Trojanowski JQ, Brunden KR. Therapeutic strategies for the treatment of tauopathies: Hopes and challenges. Alzheimers Dement 2016; 12:1051-1065. [PMID: 27751442 PMCID: PMC5116305 DOI: 10.1016/j.jalz.2016.06.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/09/2016] [Indexed: 01/25/2023]
Abstract
A group of neurodegenerative diseases referred to as tauopathies are characterized by the presence of brain cells harboring inclusions of pathological species of the tau protein. These disorders include Alzheimer's disease and frontotemporal lobar degeneration due to tau pathology, including progressive supranuclear palsy, corticobasal degeneration, and Pick's disease. Tau is normally a microtubule (MT)-associated protein that appears to play an important role in ensuring proper axonal transport, but in tauopathies tau becomes hyperphosphorylated and disengages from MTs, with consequent misfolding and deposition into inclusions that mainly affect neurons but also glia. A body of experimental evidence suggests that the development of tau inclusions leads to the neurodegeneration observed in tauopathies, and there is a growing interest in developing tau-directed therapeutic agents. The following review provides a summary of strategies under investigation for the potential treatment of tauopathies, highlighting both the promises and challenges associated with these various therapeutic approaches.
Collapse
Affiliation(s)
- Mansi R Khanna
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Jane Kovalevich
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt R Brunden
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Fontaine SN, Zheng D, Sabbagh JJ, Martin MD, Chaput D, Darling A, Trotter JH, Stothert AR, Nordhues BA, Lussier A, Baker J, Shelton L, Kahn M, Blair LJ, Stevens SM, Dickey CA. DnaJ/Hsc70 chaperone complexes control the extracellular release of neurodegenerative-associated proteins. EMBO J 2016; 35:1537-49. [PMID: 27261198 PMCID: PMC4946142 DOI: 10.15252/embj.201593489] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 12/12/2022] Open
Abstract
It is now known that proteins associated with neurodegenerative disease can spread throughout the brain in a prionlike manner. However, the mechanisms regulating the trans-synaptic spread propagation, including the neuronal release of these proteins, remain unknown. The interaction of neurodegenerative disease-associated proteins with the molecular chaperone Hsc70 is well known, and we hypothesized that much like disaggregation, refolding, degradation, and even normal function, Hsc70 may dictate the extracellular fate of these proteins. Here, we show that several proteins, including TDP-43, α-synuclein, and the microtubule-associated protein tau, can be driven out of the cell by an Hsc70 co-chaperone, DnaJC5. In fact, DnaJC5 overexpression induced tau release in cells, neurons, and brain tissue, but only when activity of the chaperone Hsc70 was intact and when tau was able to associate with this chaperone. Moreover, release of tau from neurons was reduced in mice lacking the DnaJC5 gene and when the complement of DnaJs in the cell was altered. These results demonstrate that the dynamics of DnaJ/Hsc70 complexes are critically involved in the release of neurodegenerative disease proteins.
Collapse
Affiliation(s)
- Sarah N Fontaine
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA James A. Haley Veteran's Hospital, Tampa, FL, USA
| | - Dali Zheng
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Jonathan J Sabbagh
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA James A. Haley Veteran's Hospital, Tampa, FL, USA
| | - Mackenzie D Martin
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA James A. Haley Veteran's Hospital, Tampa, FL, USA
| | - Dale Chaput
- Department of Cell, Molecular and Life Sciences, University of South Florida, Tampa, FL, USA
| | - April Darling
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Justin H Trotter
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Andrew R Stothert
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Bryce A Nordhues
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - April Lussier
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Jeremy Baker
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Lindsey Shelton
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Mahnoor Kahn
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Laura J Blair
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Stanley M Stevens
- Department of Cell, Molecular and Life Sciences, University of South Florida, Tampa, FL, USA
| | - Chad A Dickey
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA James A. Haley Veteran's Hospital, Tampa, FL, USA
| |
Collapse
|
23
|
Martin MD, Baker JD, Suntharalingam A, Nordhues BA, Shelton LB, Zheng D, Sabbagh JJ, Haystead TA, Gestwicki JE, Dickey CA. Inhibition of Both Hsp70 Activity and Tau Aggregation in Vitro Best Predicts Tau Lowering Activity of Small Molecules. ACS Chem Biol 2016; 11:2041-8. [PMID: 27177119 DOI: 10.1021/acschembio.6b00223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Three scaffolds with inhibitory activity against the heat shock protein 70 (Hsp70) family of chaperones have been found to enhance the degradation of the microtubule associated protein tau in cells, neurons, and brain tissue. This is important because tau accumulation is linked to neurodegenerative diseases including Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Here, we expanded upon this study to investigate the anti-tau efficacy of additional scaffolds with Hsp70 inhibitory activity. Five of the nine scaffolds tested lowered tau levels, with the rhodacyanine and phenothiazine scaffolds exhibiting the highest potency as previously described. Because phenothiazines also inhibit tau aggregation in vitro, we suspected that this activity might be a more accurate predictor of tau lowering. Interestingly, the rhodacyanines did inhibit in vitro tau aggregation to a similar degree as phenothiazines, correlating well with tau-lowering efficacy in cells and ex vivo slices. Moreover, other Hsp70 inhibitor scaffolds with weaker tau-lowering activity in cells inhibited tau aggregation in vitro, albeit at lower potencies. When we tested six well-characterized tau aggregation inhibitors, we determined that this mechanism of action was not a better predictor of tau-lowering than Hsp70 inhibition. Instead, we found that compounds possessing both activities were the most effective at promoting tau clearance. Moreover, cytotoxicity and PAINS activity are critical factors that can lead to false-positive lead identification. Strategies designed around these principles will likely yield more efficacious tau-lowering compounds.
Collapse
Affiliation(s)
- Mackenzie D. Martin
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Jeremy D. Baker
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Amirthaa Suntharalingam
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Bryce A. Nordhues
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Lindsey B. Shelton
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Dali Zheng
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Jonathan J. Sabbagh
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Timothy A.J. Haystead
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Jason E. Gestwicki
- Department
of Pharmaceutical Chemistry, University of California, San Francisco, San
Francisco, California 94158, United States
| | - Chad A. Dickey
- Department of Molecular
Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
- James A. Haley Veteran’s Hospital, 13000 Bruce B. Downs Blvd. Tampa, Florida 33612, United States
| |
Collapse
|
24
|
Amaral MD, Balch WE. Hallmarks of therapeutic management of the cystic fibrosis functional landscape. J Cyst Fibros 2015; 14:687-99. [PMID: 26526359 PMCID: PMC4644672 DOI: 10.1016/j.jcf.2015.09.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 01/29/2023]
Abstract
The cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein does not operate in isolation, rather in a dynamic network of interacting components that impact its synthesis, folding, stability, intracellular location and function, referred to herein as the 'CFTR Functional Landscape (CFFL)'. For the prominent F508del mutation, many of these interactors are deeply connected to a protein fold management system, the proteostasis network (PN). However, CF encompasses an additional 2000 CFTR variants distributed along its entire coding sequence (referred to as CFTR2), and each variant contributes a differential liability to PN management of CFTR and to a protein 'social network' (SN) that directs the probability of the (patho)physiologic events that impact ion transport in each cell, tissue and patient in health and disease. Recognition of the importance of the PN and SN in driving the unique patient CFFL leading to disease highlights the importance of precision medicine in therapeutic management of disease progression. We take the view herein that it is not CFTR, rather the PN/SN, and their impact on the CFFL, that are the key physiologic forces driving onset and clinical progression of CF. We posit that a deep understanding of each patients PN/SN gained by merging genomic, proteomic (mass spectrometry (MS)), and high-content microscopy (HCM) technologies in the context of novel network learning algorithms will lead to a paradigm shift in CF clinical management. This should allow for generation of new classes of patient specific PN/SN directed therapeutics for personalized management of the CFFL in the clinic.
Collapse
Affiliation(s)
- Margarida D Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Lisboa, Portugal.
| | - William E Balch
- Department of Chemical Physiology, Department of Cell and Molecular Biology, The Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|