1
|
Marvastizadeh N, Dabirmanesh B, Sajedi RH, Khajeh K. Anti-amyloidogenic effect of artemin on α-synuclein. Biol Chem 2021; 401:1143-1151. [PMID: 32673279 DOI: 10.1515/hsz-2019-0446] [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: 12/25/2019] [Accepted: 04/20/2020] [Indexed: 11/15/2022]
Abstract
α-Synuclein fibrillation is now regarded as a major pathogenic process in Parkinson's disease and its proteinaceous deposits are also detected in other neurological disorders including Alzheimer's disease. Therefore anti-amyloidegenic compounds may delay or prevent the progression of synucleinopathies disease. Molecular chaperones are group of proteins which mediate correct folding of proteins by preventing unsuitable interactions which may lead to aggregation. The objective of this study was to investigate the anti-amyloidogenic effect of molecular chaperone artemin on α-synuclein. As the concentration of artemin was increased up to 4 μg/ml, a decrease in fibril formation of α-synuclein was observed using thioflavin T (ThT) fluorescence and congo red (CR) assay. Transmission electron microscopy (TEM) images also demonstrated a reduction in fibrils in the presence of artemin. The secondary structure of α-synuclein was similar to its native form prior to fibrillation when incubated with artemin. A cell-based assay has shown that artemin inhibits α-synuclein aggregation and reduce cytotoxicity, apoptosis and reactive oxygen species (ROS) production. Our results revealed that artemin has efficient chaperon activity for preventing α-synuclein fibril formation and toxicity.
Collapse
Affiliation(s)
- Narges Marvastizadeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-175, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-175, Tehran, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-175, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-175, Tehran, Iran
| |
Collapse
|
2
|
Silva MC, Haggarty SJ. Tauopathies: Deciphering Disease Mechanisms to Develop Effective Therapies. Int J Mol Sci 2020; 21:ijms21238948. [PMID: 33255694 PMCID: PMC7728099 DOI: 10.3390/ijms21238948] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by the pathological accumulation of microtubule-associated protein tau (MAPT) in the form of neurofibrillary tangles and paired helical filaments in neurons and glia, leading to brain cell death. These diseases include frontotemporal dementia (FTD) and Alzheimer's disease (AD) and can be sporadic or inherited when caused by mutations in the MAPT gene. Despite an incredibly high socio-economic burden worldwide, there are still no effective disease-modifying therapies, and few tau-focused experimental drugs have reached clinical trials. One major hindrance for therapeutic development is the knowledge gap in molecular mechanisms of tau-mediated neuronal toxicity and death. For the promise of precision medicine for brain disorders to be fulfilled, it is necessary to integrate known genetic causes of disease, i.e., MAPT mutations, with an understanding of the dysregulated molecular pathways that constitute potential therapeutic targets. Here, the growing understanding of known and proposed mechanisms of disease etiology will be reviewed, together with promising experimental tau-directed therapeutics, such as recently developed tau degraders. Current challenges faced by the fields of tau research and drug discovery will also be addressed.
Collapse
|
3
|
Khosravi Z, Nasiri Khalili MA, Moradi S, Hassan Sajedi R, Zeinoddini M. The Molecular Chaperone Artemin Efficiently Blocks Fibrillization of TAU Protein In Vitro. CELL JOURNAL 2017; 19:569-577. [PMID: 29105391 PMCID: PMC5672095 DOI: 10.22074/cellj.2018.4510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022]
Abstract
Objective Aggregation of the TAU proteins in the form of neurofibrillary tangles (NFTs) in the brain is a common risk
factor in tauopathies including Alzheimer’s disease (AD). Several strategies have been implemented to target NFTs,
among which chaperones, which facilitate the proper folding of proteins, appear to hold great promise in effectively
inhibiting TAU polymerization. The aim of this study was to analyze the impact of the chaperone Artemin on TAU
aggregation in vitro.
Materials and Methods In this experimental study, recombinant TAU- or Artemin proteins were expressed in E.coli
bacteria, and purified using ion-exchange and affinity chromatography. Sodium dodecyl sulfate-poly acrylamide gel
electrophoresis (SDS-PAGE) was used to run the extracted proteins and check their purity. Heparin was used as an
aggregation inducer. The interaction kinetics of TAU aggregation and disassembly was performed using thioflavin T
(ThT) fluorescence analysis and circular dichroism (CD) spectroscopy.
Results Ion-exchange and affinity chromatography yielded highly pure TAU and Artemin proteins for subsequent
analyses. In addition, we found that heparin efficiently induced TAU fibrillization 48 hours post-incubation, as evidenced
by ThT assay. Importantly, Artemin was observed to effectively block the aggregation of both physiologic- and supra-
physiologic TAU concentrations in a dose-dependent manner, as judged by ThT and CD spectroscopy analyses.
Conclusion Our collective results show, for the first time, that the chaperone Artemin could significantly inhibit
aggregation of the TAU proteins in a dose-dependent manner, and support Artemin as a potential potent blocker of TAU
aggregation in people with AD.
Collapse
Affiliation(s)
- Zahra Khosravi
- Department of Biosciences and Biotechnology, Malek Ashtar University of Technology, Tehran, Iran
| | | | - Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Reza Hassan Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Zeinoddini
- Department of Biosciences and Biotechnology, Malek Ashtar University of Technology, Tehran, Iran
| |
Collapse
|
4
|
Rauch JN, Olson SH, Gestwicki JE. Interactions between Microtubule-Associated Protein Tau (MAPT) and Small Molecules. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a024034. [PMID: 27940599 DOI: 10.1101/cshperspect.a024034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tau aggregation is linked to multiple neurodegenerative disorders that are collectively termed tauopathies. Small molecules are powerful probes of the aggregation process, helping to reveal the key steps and serving as diagnostics and reporters. Moreover, some of these small molecules may have potential as therapeutics. This review details how small molecules and chemical biology have helped to elucidate the mechanisms of tau aggregation and how they are being used to detect and prevent tau aggregation. In addition, we comment on how new insights into tau prions are changing the approach to small molecule discovery.
Collapse
Affiliation(s)
- Jennifer N Rauch
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Steven H Olson
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Jason E Gestwicki
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
5
|
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: 78] [Impact Index Per Article: 9.8] [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
|
6
|
Pickhardt M, Neumann T, Schwizer D, Callaway K, Vendruscolo M, Schenk D, St George-Hyslop P, Mandelkow EM, Dobson CM, McConlogue L, Mandelkow E, Tóth G. Identification of Small Molecule Inhibitors of Tau Aggregation by Targeting Monomeric Tau As a Potential Therapeutic Approach for Tauopathies. Curr Alzheimer Res 2015; 12:814-28. [PMID: 26510979 PMCID: PMC4976804 DOI: 10.2174/156720501209151019104951] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 06/13/2015] [Accepted: 06/26/2015] [Indexed: 12/15/2022]
Abstract
A potential strategy to alleviate the aggregation of intrinsically disordered proteins (IDPs) is to maintain the native functional state of the protein by small molecule binding. However, the targeting of the native state of IDPs by small molecules has been challenging due to their heterogeneous conformational ensembles. To tackle this challenge, we applied a high-throughput chemical microarray surface plasmon resonance imaging screen to detect the binding between small molecules and monomeric full-length Tau, a protein linked with the onset of a range of Tauopathies. The screen identified a novel set of drug-like fragment and lead-like compounds that bound to Tau. We verified that the majority of these hit compounds reduced the aggregation of different Tau constructs in vitro and in N2a cells. These results demonstrate that Tau is a viable receptor of drug-like small molecules. The drug discovery approach that we present can be applied to other IDPs linked to other misfolding diseases such as Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Eckhard Mandelkow
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| | - Gergely Tóth
- Department of Clinical Neuroscienes, Wolfson Brain Imaging Center, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, United Kingdom.
| |
Collapse
|
7
|
Potential synergy between tau aggregation inhibitors and tau chaperone modulators. ALZHEIMERS RESEARCH & THERAPY 2013; 5:41. [PMID: 24041111 PMCID: PMC3979086 DOI: 10.1186/alzrt207] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tau is a soluble, microtubule-associated protein known to aberrantly form
amyloid-positive aggregates. This pathology is characteristic for more than 15
neuropathies, the most common of which is Alzheimer’s disease. Finding
therapeutics to reverse or remove this non-native tau state is of great
interest; however, at this time only one drug is entering phase III clinical
trials for treating tauopathies. Generally, tau manipulation by therapeutics can
either directly or indirectly alter tau aggregation and stability. Drugs that
bind and change the conformation of tau itself are largely classified as
aggregation inhibitors, while drugs that alter the activity of a tau-effector
protein fall into several categories, such as kinase inhibitors, microtubule
stabilizers, or chaperone modulators. Chaperone inhibitors that have proven
effective in tau models include heat shock protein 90 inhibitors, heat shock
protein 70 inhibitors and activators, as well as inducers of heat shock
proteins. While many of these compounds can alter tau levels and/or aggregation
states, it is possible that combining these approaches may produce the most
optimal outcome. However, because many of these compounds have multiple
off-target effects or poor blood–brain barrier permeability, the
development of this synergistic therapeutic strategy presents significant
challenges. This review will summarize many of the drugs that have been
identified to alter tau biology, with special focus on therapeutics that prevent
tau aggregation and regulate chaperone-mediated clearance of tau.
Collapse
|
8
|
Gerson JE, Kayed R. Formation and propagation of tau oligomeric seeds. Front Neurol 2013; 4:93. [PMID: 23882255 PMCID: PMC3713404 DOI: 10.3389/fneur.2013.00093] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/01/2013] [Indexed: 12/14/2022] Open
Abstract
Tau misfolding and aggregation leads to the formation of neurofibrillary tangles (NFTs), which have long been considered one of the main pathological hallmarks for numerous neurodegenerative diseases known as tauopathies, including Alzheimer’s Disease (AD) and Parkinson’s Disease (PD). However, recent studies completed both in vitro and in vivo suggest that intermediate forms of tau, known as tau oligomers, between the monomeric form and NFTs are the true toxic species in disease and the best targets for anti-tau therapies. However, the exact mechanism by which the spread of pathology occurs is unknown. Evidence suggests that tau oligomers may act as templates for the misfolding of native tau, thereby seeding the spread of the toxic forms of the protein. Recently, researchers have reported the ability of tau oligomers to enter and exit cells, propagating from disease-affected regions to unaffected areas. While the mechanism by which the spreading of misfolded tau occurs has yet to be elucidated, there are a few different models which have been proposed, including cell membrane stress and pore-formation, endocytosis and exocytosis, and non-traditional secretion of protein not enclosed by a membrane. Coming to an understanding of how toxic tau species seed and spread through the brain will be crucial to finding effective treatments for neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Julia E Gerson
- George P. and Cynthia Woods Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch , Galveston, TX , USA ; Department of Neurology, University of Texas Medical Branch , Galveston, TX , USA ; Department of Neuroscience and Cell Biology, University of Texas Medical Branch , Galveston, TX , USA
| | | |
Collapse
|
9
|
Bulic B, Pickhardt M, Mandelkow E. Progress and developments in tau aggregation inhibitors for Alzheimer disease. J Med Chem 2013; 56:4135-55. [PMID: 23484434 DOI: 10.1021/jm3017317] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pharmacological approaches directed toward Alzheimer disease are diversifying in parallel with a growing number of promising targets. Investigations on the microtubule-associated protein tau yielded innovative targets backed by recent findings about the central role of tau in numerous neurodegenerative diseases. In this review, we summarize the recent evolution in the development of nonpeptidic small molecules tau aggregation inhibitors (TAGIs) and their advancement toward clinical trials. The compounds are classified according to their chemical structures, providing correlative insights into their pharmacology. Overall, shared structure-activity traits are emerging, as well as specific binding modes related to their ability to engage in hydrogen bonding. Medicinal chemistry efforts on TAGIs together with encouraging in vivo data argue for successful translation to the clinic.
Collapse
Affiliation(s)
- Bruno Bulic
- Laboratory of Organic Synthesis of Functional Systems, Humboldt-Universität zu Berlin, Brook-Taylor-Strasse 2, 12489 Berlin, Germany.
| | | | | |
Collapse
|
10
|
Natural products as a rich source of tau-targeting drugs for Alzheimer's disease. Future Med Chem 2013; 4:1751-61. [PMID: 22924511 DOI: 10.4155/fmc.12.124] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia, affecting more than 5.4 million people in the USA. Although the cause of AD is not well understood, the cholinergic, amyloid and tau hypotheses were proposed to explain its development. Drug discovery for AD based on the cholinergic and amyloid theories have not been effective. In this article we summarize tau-based natural products as AD therapeutics from a variety of biological sources, including the anti-amyloid agent curcumin, isolated from turmeric, the microtubule stabilizer paclitaxel, from the Pacific Yew Taxus brevifolia, and the Streptomyces-derived Hsp90 inhibitor, geldanamycin. The overlooked approach of clearing tau aggregation will most likely be the next objective for AD drug discovery.
Collapse
|
11
|
Himmelstein DS, Ward SM, Lancia JK, Patterson KR, Binder LI. Tau as a therapeutic target in neurodegenerative disease. Pharmacol Ther 2012; 136:8-22. [PMID: 22790092 DOI: 10.1016/j.pharmthera.2012.07.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 06/22/2012] [Indexed: 01/16/2023]
Abstract
Tau is a microtubule-associated protein thought to help modulate the stability of neuronal microtubules. In tauopathies, including Alzheimer's disease and several frontotemporal dementias, tau is abnormally modified and misfolded resulting in its disassociation from microtubules and the generation of pathological lesions characteristic for each disease. A recent surge in the population of people with neurodegenerative tauopathies has highlighted the immense need for disease-modifying therapies for these conditions, and new attention has focused on tau as a potential target for intervention. In the current work we summarize evidence linking tau to disease pathogenesis and review recent therapeutic approaches aimed at ameliorating tau dysfunction. The primary therapeutic tactics considered include kinase inhibitors and phosphatase activators, immunotherapies, small molecule inhibitors of protein aggregation, and microtubule-stabilizing agents. Although the evidence for tau-based treatments is encouraging, additional work is undoubtedly needed to optimize each treatment strategy for the successful development of safe and effective therapeutics.
Collapse
Affiliation(s)
- Diana S Himmelstein
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Tarry 8-754, 300 E. Superior St., Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
12
|
Schafer KN, Murale DP, Kim K, Cisek K, Kuret J, Churchill DG. Structure-activity relationship of cyclic thiacarbocyanine tau aggregation inhibitors. Bioorg Med Chem Lett 2011; 21:3273-6. [PMID: 21549596 DOI: 10.1016/j.bmcl.2011.04.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Revised: 04/04/2011] [Accepted: 04/08/2011] [Indexed: 11/19/2022]
Abstract
Macrocyclic bis-thiacarbocyanines are efficacious inhibitors of tau protein aggregation. To extend the structure-activity relationship of this inhibitor class, N,N'-alkylene bis-thiacarbocyanines linked by chains of three to eight methylene carbons were synthesized and examined for inhibitory activity against recombinant human tau aggregation in vitro. At 10 micromolar concentration, inhibitory activity varied with linker length, with four methylene units being most efficacious. On the basis of absorbance spectroscopy measurements, linker length also affected compound folding and aggregation propensity, with a linker length of four methylene units being optimal for preserving open monomer conformation. These data suggest that inhibitory potency can be optimized through control of linker length, and that a contributory mechanism involves modulation of compound folding and aggregation.
Collapse
Affiliation(s)
- Kelsey N Schafer
- Center for Molecular Neurobiology, Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, 1060 Carmack Rd, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
13
|
Jones JR, Lebar MD, Jinwal UK, Abisambra JF, Koren J, Blair L, O'Leary JC, Davey Z, Trotter J, Johnson AG, Weeber E, Eckman CB, Baker BJ, Dickey CA. The diarylheptanoid (+)-aR,11S-myricanol and two flavones from bayberry (Myrica cerifera) destabilize the microtubule-associated protein tau. JOURNAL OF NATURAL PRODUCTS 2011; 74:38-44. [PMID: 21141876 PMCID: PMC3070757 DOI: 10.1021/np100572z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Target-based drug discovery for Alzheimer's disease (AD) centered on modulation of the amyloid β peptide has met with limited success. Therefore, recent efforts have focused on targeting the microtubule-associated protein tau. Tau pathologically accumulates in more than 15 neurodegenerative diseases and is most closely linked with postsymptomatic progression in AD. We endeavored to identify compounds that decrease tau stability rather than prevent its aggregation. An extract from Myrica cerifera (bayberry/southern wax myrtle) potently reduced both endogenous and overexpressed tau protein levels in cells and murine brain slices. The bayberry flavonoids myricetin and myricitrin were confirmed to contribute to this potency, but a diarylheptanoid, myricanol, was the most effective anti-tau component in the extract, with potency approaching the best targeted lead therapies. (+)-aR,11S-Myricanol, isolated from M. cerifera and reported here for the first time as the naturally occurring aglycone, was significantly more potent than commercially available (±)-myricanol. Myricanol may represent a novel scaffold for drug development efforts targeting tau turnover in AD.
Collapse
|
14
|
Ballatore C, Brunden KR, Trojanowski JQ, Lee VMY, Smith AB, Huryn DM. Modulation of protein-protein interactions as a therapeutic strategy for the treatment of neurodegenerative tauopathies. Curr Top Med Chem 2011; 11:317-30. [PMID: 21320060 PMCID: PMC3069499 DOI: 10.2174/156802611794072605] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Accepted: 04/16/2010] [Indexed: 12/26/2022]
Abstract
The recognition that malfunction of the microtubule (MT) associated protein tau is likely to play a defining role in the onset and/or progression of a number of neurodegenerative diseases, including Alzheimer's disease, has resulted in the initiation of drug discovery programs that target this protein. Tau is an endogenous MT-stabilizing agent that is highly expressed in the axons of neurons. The MT-stabilizing function of tau is essential for the axonal transport of proteins, neurotransmitters and other cellular constituents. Under pathological conditions, tau misfolding and aggregation results in axonal transport deficits that appear to have deleterious consequences for the affected neurons, leading to synapse dysfunction and, ultimately, neuronal loss. This review focuses on both progress and unresolved issues surrounding the development of novel therapeutics for the treatment of neurodegenerative tauopathies, which are based on (A) MT-stabilizing agents to compensate for the loss of normal tau function, and (B) small molecule inhibitors of tau aggregation.
Collapse
Affiliation(s)
- C Ballatore
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, 231 South 34th St., Philadelphia, PA 19104-6323, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Brunden KR, Ballatore C, Crowe A, Smith AB, Lee VMY, Trojanowski JQ. Tau-directed drug discovery for Alzheimer's disease and related tauopathies: a focus on tau assembly inhibitors. Exp Neurol 2010; 223:304-10. [PMID: 19744482 PMCID: PMC2864354 DOI: 10.1016/j.expneurol.2009.08.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 08/21/2009] [Accepted: 08/27/2009] [Indexed: 12/26/2022]
Abstract
The microtubule-associated protein tau forms insoluble filaments that deposit as neurofibrillary tangles (NFTs) in the brains of those with Alzheimer's disease (AD) and other related neurodegenerative disorders. The presence of both NFTs and amyloid beta (Abeta)-containing senile plaques within the brain is required to confirm the diagnosis of AD. However, the demonstration that familial AD can be caused by mutations that result in increased Abeta production has resulted in AD drug discovery strategies that are largely focused on reducing brain Abeta levels, with substantially less emphasis on tau-directed approaches. This trend may be changing, as there are an increasing number of research programs that are exploring ways to reduce NFTs in AD and related tauopathies. We briefly review recent advances in tau-based drug discovery, with an emphasis on the identification of compounds that inhibit the assembly of tau into multimers and fibrils.
Collapse
Affiliation(s)
- Kurt R Brunden
- Center for Neurodegenerative Disease Research, Institute on Aging, and Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E. Tau protein and tau aggregation inhibitors. Neuropharmacology 2010; 59:276-89. [PMID: 20149808 DOI: 10.1016/j.neuropharm.2010.01.016] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 01/20/2010] [Accepted: 01/26/2010] [Indexed: 12/13/2022]
Abstract
Alzheimer disease is characterized by pathological aggregation of two proteins, tau and Abeta-amyloid, both of which are considered to be toxic to neurons. In this review we summarize recent advances on small molecule inhibitors of protein aggregation with emphasis on tau, with activities mediated by the direct interference of self-assembly. The inhibitors can be clustered in several compound classes according to their chemical structure, with subsequent description of the structure-activity relationships, showing that hydrophobic interactions are prevailing. The description is extended to the pharmacological profile of the compounds in order to evaluate their drug-likeness, with special attention to toxicity and bioavailability. The collected data indicate that following the improvements of the in vitro inhibitory potencies, the consideration of the in vivo pharmacokinetics is an absolute prerequisite for the development of compounds suitable for a transfer from bench to bedside.
Collapse
Affiliation(s)
- Bruno Bulic
- Center for Advanced European Studies and Research, Bonn, Germany.
| | | | | | | |
Collapse
|
17
|
Meraz-Ríos MA, Lira-De León KI, Campos-Peña V, De Anda-Hernández MA, Mena-López R. Tau oligomers and aggregation in Alzheimer's disease. J Neurochem 2009; 112:1353-67. [PMID: 19943854 DOI: 10.1111/j.1471-4159.2009.06511.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We are analyzing the physiological function of Tau protein and its abnormal pathological behavior when this protein is self-assemble into pathological filaments. These aggregates of Tau protein are the main components in many diseases such as Alzheimer's disease (AD). Recent studies suggest that Tau acquires complex oligomeric conformations which may be toxic. In this review, we emphasized the possible phenomena implicated in the formation of these oligomers. Studies with chemical inductors indicates that the microtubule-binding domain is the most important region involved in Tau aggregation and showed the requirement of a pre-arrange Tau in abnormal conformation to promote self-assembly. Transgenic animal models and AD neuropathology studies showed that post-translational modifications are also implicated in Tau aggregation and neural cell death during AD development. Therefore, we analyzed some events that could be present during Tau aggregation. Finally, we included a brief discussion of the possible relation between glucose metabolism dysfunction in AD, and data of Tau aggregation by using aggregation inhibitors. In conclusion, the process Tau aggregation deserves further investigations to design possible therapeutic targets to inhibit the toxicity of these aggregates and it is possible that could be extended to other diseases with similar etiology.
Collapse
Affiliation(s)
- Marco A Meraz-Ríos
- Department of Molecular Biomedicine, Center of Research and Advanced Studies CINVESTAV-IPN, México DF, Mexico.
| | | | | | | | | |
Collapse
|
18
|
Advances in tau-focused drug discovery for Alzheimer's disease and related tauopathies. Nat Rev Drug Discov 2009; 8:783-93. [PMID: 19794442 DOI: 10.1038/nrd2959] [Citation(s) in RCA: 312] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Neuronal inclusions comprised of the microtubule-associated protein tau are found in numerous neurodegenerative diseases, commonly known as tauopathies. In Alzheimer's disease - the most prevalent tauopathy - misfolded tau is probably a key pathological agent. The recent failure of amyloid-beta-targeted therapeutics in Phase III clinical trials suggests that it is timely and prudent to consider alternative drug discovery strategies for Alzheimer's disease. Here, we focus on strategies directed at reducing misfolded tau and compensating for the loss of normal tau function.
Collapse
|
19
|
Congdon EE, Figueroa YH, Wang L, Toneva G, Chang E, Kuret J, Conrad C, Duff KE. Inhibition of tau polymerization with a cyanine dye in two distinct model systems. J Biol Chem 2009; 284:20830-9. [PMID: 19478088 PMCID: PMC2742848 DOI: 10.1074/jbc.m109.016089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 05/22/2009] [Indexed: 01/02/2023] Open
Abstract
In a host of neurodegenerative diseases Tau, a microtubule-associated protein, aggregates into insoluble lesions within neurons. Previous studies have utilized cyanine dyes as Tau aggregation inhibitors in vitro. Herein we utilize cyanine dye 3,3'-diethyl-9-methyl-thiacarbocyanine iodide (C11) to modulate Tau polymerization in two model systems, an organotypic slice culture model derived from Tau transgenic mice and a split green fluorescent protein complementation assay in Tau-expressing cells. In slice cultures, submicromolar concentrations (0.001 microm) of C11 produced a significant reduction of aggregated Tau and a corresponding increase in unpolymerized Tau. In contrast, treatment with a 1 microm dose promoted aggregation of Tau. These results were recapitulated in the complementation assay where administration of 1 microm C11 produced a significant increase in polymerized Tau relative to control, whereas treatment of cells with 0.01 microm C11 resulted in a marked reduction of aggregated Tau. In the organotypic slice cultures, modulation of Tau aggregation was independent of changes in phosphorylation at disease and microtubule binding relevant epitopes for both dosing regimes. Furthermore, treatment with 0.001 microm C11 resulted in a decrease in both total filament mass and number. There was no evidence of apoptosis or loss of synaptic integrity at either dose, however, whereas submicromolar concentrations of C11 did not interfere with microtubule binding, higher doses resulted in a decrease in the levels of microtubule-bound Tau. Overall, a cyanine dye can dissociate aggregated Tau in an ex vivo model of tauopathy with little toxicity and exploration of the use of these type of dyes as therapeutic agents is warranted.
Collapse
Affiliation(s)
- Erin E. Congdon
- From the Department of Pathology, Taub Institute, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 and
| | - Yvette H. Figueroa
- From the Department of Pathology, Taub Institute, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 and
| | - Lili Wang
- From the Department of Pathology, Taub Institute, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 and
| | - Galina Toneva
- From the Department of Pathology, Taub Institute, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 and
| | | | - Jeff Kuret
- Department of Cellular and Molecular Biology, Ohio State University, Columbus, Ohio 43210
| | - Christopher Conrad
- From the Department of Pathology, Taub Institute, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 and
| | - Karen E. Duff
- From the Department of Pathology, Taub Institute, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 and
| |
Collapse
|
20
|
Chang E, Congdon EE, Honson NS, Duff KE, Kuret J. Structure-activity relationship of cyanine tau aggregation inhibitors. J Med Chem 2009; 52:3539-47. [PMID: 19432420 DOI: 10.1021/jm900116d] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A structure-activity relationship for symmetrical cyanine inhibitors of human tau aggregation was elaborated using a filter trap assay. Antagonist activity depended on cyanine heterocycle, polymethine bridge length, and the nature of meso- and N-substituents. One potent member of the series, 3,3'-diethyl-9-methylthiacarbocyanine iodide (compound 11), retained submicromolar potency and had calculated physical properties consistent with blood-brain barrier and cell membrane penetration. Exposure of organotypic slices prepared from JNPL3 transgenic mice (which express human tau harboring the aggregation prone P301L tauopathy mutation) to compound 11 for one week revealed a biphasic dose response relationship. Low nanomolar concentrations decreased insoluble tau aggregates to half those observed in slices treated with vehicle alone. In contrast, high concentrations (> or =300 nM) augmented tau aggregation and produced abnormalities in tissue tubulin levels. These data suggest that certain symmetrical carbocyanine dyes can modulate tau aggregation in the slice biological model at concentrations well below those associated with toxicity.
Collapse
Affiliation(s)
- Edward Chang
- Center for Molecular Neurobiology, Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
21
|
Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H, Mandelkow E. Development of tau aggregation inhibitors for Alzheimer's disease. Angew Chem Int Ed Engl 2009; 48:1740-52. [PMID: 19189357 DOI: 10.1002/anie.200802621] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A variety of human diseases are suspected to be directly linked to protein misfolding. Highly organized protein aggregates, called amyloid fibrils, and aggregation intermediates are observed; these are considered to be mediators of cellular toxicity and thus attract a great deal of attention from investigators. Neurodegenerative pathologies such as Alzheimer's disease account for a major part of these protein misfolding diseases. The last decade has witnessed a renaissance of interest in inhibitors of tau aggregation as potential disease-modifying drugs for Alzheimer's disease and other "tauopathies". The recent report of a phase II clinical trial with the tau aggregation inhibitor MTC could hold promise for the validation of the concept. This Review summarizes the available data concerning small-molecule inhibitors of tau aggregation from a medicinal chemistry point of view.
Collapse
Affiliation(s)
- Bruno Bulic
- Max-Planck-Institute for Molecular Physiology, Dortmund, Germany.
| | | | | | | | | | | |
Collapse
|
22
|
Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H, Mandelkow E. Entwicklung von Inhibitoren der Tau-Aggregation bei Morbus Alzheimer. Angew Chem Int Ed Engl 2009. [DOI: 10.1002/ange.200802621] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
23
|
Honson NS, Jensen JR, Darby MV, Kuret J. Potent inhibition of tau fibrillization with a multivalent ligand. Biochem Biophys Res Commun 2007; 363:229-34. [PMID: 17854770 PMCID: PMC2048976 DOI: 10.1016/j.bbrc.2007.08.166] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 08/28/2007] [Indexed: 11/23/2022]
Abstract
Small-molecule inhibitors of tau fibrillization are under investigation as tools for interrogating the tau aggregation pathway and as potential therapeutic agents for Alzheimer's disease. Established inhibitors include thiacarbocyanine dyes, which can inhibit recombinant tau fibrillization in the presence of anionic surfactant aggregation inducers. In an effort to increase inhibitory potency, a cyclic bis-thiacarbocyanine molecule containing two thiacarbocyanine moieties was synthesized and characterized with respect to tau fibrillization inhibitory activity by electron microscopy and ligand aggregation state by absorbance spectroscopy. Results showed that the inhibitory activity of the bis-thiacarbocyanine was qualitatively similar to a monomeric cyanine dye, but was more potent with 50% inhibition achieved at approximately 80nM concentration. At all concentrations tested in aqueous solution, the bis-thiacarbocyanine collapsed to form a closed clamshell structure. However, the presence of tau protein selectively stabilized the open conformation. These results suggest that the inhibitory activity of bis-thiacarbocyanine results from multivalency, and reveal a route to more potent tau aggregation inhibitors.
Collapse
Affiliation(s)
- Nicolette S. Honson
- Center for Molecular Neurobiology, Department of Molecular & Cellular Biochemistry, College of Medicine
| | - Jordan R. Jensen
- Center for Molecular Neurobiology, Department of Molecular & Cellular Biochemistry, College of Medicine
| | - Michael V. Darby
- College of Pharmacy; The Ohio State University, Columbus, OH 43210
| | - Jeff Kuret
- Center for Molecular Neurobiology, Department of Molecular & Cellular Biochemistry, College of Medicine
- *Corresponding Author: Jeff Kuret, Ph.D., Center for Molecular Neurobiology, 1060 Carmack Rd., Columbus, OH 43210, Fax: 614-292-5379, Email address: (J. Kuret)
| |
Collapse
|