501
|
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
|
502
|
Ramachandran G, Udgaonkar JB. Mechanistic studies unravel the complexity inherent in tau aggregation leading to Alzheimer's disease and the tauopathies. Biochemistry 2013; 52:4107-26. [PMID: 23721410 DOI: 10.1021/bi400209z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aggregation of the protein tau into amyloid fibrils is known to be involved in the causation of the neurodegenerative tauopathies and the progression of cognitive decline in Alzheimer's disease. This review surveys the mechanism of tau aggregation with special emphasis on the information obtained from biochemical and biophysical studies. First, tau is described from a structure-function perspective. Subsequently, the connection of tau to neurodegeneration is explained, and a description of the tau amyloid fibril is provided. Lastly, studies of the mechanism of tau fibril formation are reviewed, and the physiological significance of these studies with reference to how they can clarify many aspects of disease progression is described. The aim of this review is to underscore how mechanistic studies reveal the complexity of the tau fibril formation pathway and the plethora of species populated on or off the pathway of aggregation, and how this information can be beneficial in the design of inhibitors or drugs that ameliorate neurodegeneration.
Collapse
Affiliation(s)
- Gayathri Ramachandran
- National Centre for Biological Sciences, Tata Institute of Fundamental Research , Bangalore 560065, India
| | | |
Collapse
|
503
|
López GE, Cruz A, Sepulveda-Chervony M, López-Garriga J, Torres-Lugo M. Using a reduced dimensionality model to compute the thermodynamic properties of finite polypeptide aggregates. J Biol Phys 2013; 38:383-95. [PMID: 23729904 DOI: 10.1007/s10867-011-9259-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 12/21/2011] [Indexed: 12/01/2022] Open
Abstract
By implementing a simple reduced dimensionality model to describe the interactions in finite systems composed of two seven-amino-acid peptides, the thermodynamic properties of ordered and disordered aggregates were computed. Within this model, the hydrophobicity of each amino acid was varied, and the stability of the systems computed. Accurate averages in the canonical ensemble were obtained using various replica exchange Monte Carlo algorithms. Low and high temperature regions were encountered where the ordered and disordered aggregates were stabilized. It was observed that as the degree of hydrophobicity increased, the stability of the aggregates increased, with a significant energetic stabilization obtained for the ordered aggregates. Upon decreasing the concentration of the solution, the stability of the amorphous aggregates increased when compared to the ordered systems.
Collapse
Affiliation(s)
- Gustavo E López
- Department of Chemistry, University of Puerto Rico, at Mayagüez, Mayagüez, P.R. 00681 USA ; Department of Chemistry, Lehman College-CUNY, Bronx, NY 10468 USA
| | | | | | | | | |
Collapse
|
504
|
Lin R, Cheetham AG, Zhang P, Lin YA, Cui H. Supramolecular filaments containing a fixed 41% paclitaxel loading. Chem Commun (Camb) 2013; 49:4968-70. [PMID: 23612448 PMCID: PMC3685178 DOI: 10.1039/c3cc41896k] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We report here the self-assembly of a rationally designed paclitaxel drug amphiphile into well-defined supramolecular filaments that possess a fixed 41% paclitaxel loading. These filaments can exert effective cytotoxicity against a number of cell lines comparable to that of free paclitaxel.
Collapse
Affiliation(s)
- Ran Lin
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Steet, Baltimore, MD 21218, United States
| | - Andrew G. Cheetham
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Steet, Baltimore, MD 21218, United States
| | - Pengcheng Zhang
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Steet, Baltimore, MD 21218, United States
| | - Yi-an Lin
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Steet, Baltimore, MD 21218, United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Steet, Baltimore, MD 21218, United States
| |
Collapse
|
505
|
Zheng J, Baghkhanian AM, Nowick JS. A Hydrophobic Surface Is Essential To Inhibit the Aggregation of a Tau-Protein-Derived Hexapeptide. J Am Chem Soc 2013; 135:6846-52. [DOI: 10.1021/ja310817d] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jing Zheng
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, United
States
| | - Arya M. Baghkhanian
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, United
States
| | - James S. Nowick
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, United
States
| |
Collapse
|
506
|
Lee MJ, Lee JH, Rubinsztein DC. Tau degradation: the ubiquitin-proteasome system versus the autophagy-lysosome system. Prog Neurobiol 2013; 105:49-59. [PMID: 23528736 DOI: 10.1016/j.pneurobio.2013.03.001] [Citation(s) in RCA: 259] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 03/03/2013] [Accepted: 03/13/2013] [Indexed: 01/01/2023]
Abstract
The ubiquitin-proteasome system (UPS) and the autophagy-lysosome system are two major protein quality control mechanisms in eukaryotic cells. While the UPS has been considered for decades as the critical regulator in the degradation of various aggregate-prone proteins, autophagy has more recently been shown to be an important pathway implicated in neuronal health and disease. The two hallmark lesions of Alzheimer's disease (AD) are extracellular β-amyloid plaques and intracellular tau tangles. It has been suggested that tau accumulation is pathologically more relevant to the development of neurodegeneration and cognitive decline in AD patients than β-amyloid plaques. Here, we review the UPS and autophagy-mediated tau clearance mechanisms and outline the biochemical connections between these two processes. In addition, we discuss pharmacological methods that target these degradation systems for the treatment and prevention of AD.
Collapse
Affiliation(s)
- Min Jae Lee
- Department of Applied Chemistry, College of Applied Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 446-701, Republic of Korea.
| | | | | |
Collapse
|
507
|
Yao J, Gao X, Sun W, Yao T, Shi S, Ji L. Molecular Hairpin: A Possible Model for Inhibition of Tau Aggregation by Tannic Acid. Biochemistry 2013; 52:1893-902. [DOI: 10.1021/bi400240c] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Junliang Yao
- Department of Chemistry, Tongji University, Shanghai 200092, China
| | - Xing Gao
- Department of Chemistry, Tongji University, Shanghai 200092, China
| | - Wenliang Sun
- Department of Chemistry, Tongji University, Shanghai 200092, China
| | - Tianming Yao
- Department of Chemistry, Tongji University, Shanghai 200092, China
| | - Shuo Shi
- Department of Chemistry, Tongji University, Shanghai 200092, China
| | - Liangnian Ji
- MOE Laboratory of Bioinorganic
and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
508
|
Anand U, Mukherjee M. Exploring the self-assembly of a short aromatic Aβ(16-24) peptide. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:2713-2721. [PMID: 23379750 DOI: 10.1021/la304585a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The use of self-assembling peptides as scaffolds for creating biomaterials has prompted the scientific community to carry out studies on short peptides as model systems. Short peptides help in dissecting contributions from different interactions, unlike large peptides, where multiple interactions make it difficult to dissect the contributions of individual interactions. This opens avenues for fine tuning peptides to carry out a wide range of physical or chemical properties. In this line of study Aβ(16-24) is a versatile building block not only as a scaffold for creating biomaterials but also because it forms the active core in the protein that forms amyloid plaques. In this study, we probe the self-assembly of peptide Aβ(16-24) using fluorescence spectroscopy, circular dichroism, isothermal titration calorimetry, transmission electron microscopy, and atomic force microscopy. The process of self-assembly is dictated by the burial of phenyl alanines in the hydrophobic core and guided by nonbonding interactions and H-bonding. The process of fibril formation is enthalpically driven, and the fibrils showed blue and green luminescence without the addition of any external agent or sensitizer. Because these short peptides are known to bind with fully formed amyloid fibrils, this opens a route to the study of amyloid systems in vitro or isolated from patients suffering from Alzheimer's disease.
Collapse
Affiliation(s)
- Uttam Anand
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Indore By-pass Road, Bhauri, Bhopal 462 030, Madhya Pradesh, India
| | | |
Collapse
|
509
|
Maltsev A, Dovidchenko N, Uteshev V, Sokolik V, Shtang O, Yakushin M, Sokolova N, Surin A, Galzitskaya O. Intensive protein synthesis in neurons and phosphorylation of beta-amyloid precursor protein and tau-protein are triggering factors of neuronal amyloidosis and Alzheimer's disease. ACTA ACUST UNITED AC 2013; 59:144-70. [DOI: 10.18097/pbmc20135902144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Recently the studies of Alzheimer’s disease have become particularly actual and have attracted scientists from all over the world to this problem as a result of dissemination of this dangerous disorder. The reason for such pathogenesis is not known, but the final image, for the first time obtained on microscopic brain sections from patients with this disease more than a hundred years ago, is well known to clinicists. This is the deposition of Ab amyloid in the brain tissue of senile plaques and fibrils. Many authors suppose that the deposition of beta-amyloid provokes secondary neuronal changes which are the reason of neuron death. Other authors associate the death of neurons with hyperphosphorylation of tau-proteins which form neurofibrillar coils inside nerve cells and lead to their death. For creation of methods of preclinical diagnostics and effective treatment of Alzheimer’s disease novel knowledge is required on the nature of triggering factors of sporadic isoforms of Alzheimer’s disease, on cause-effect relationships of phosphorylation of amyloid precursor protein with formation of pathogenic beta-amyloids, on the relationship with these factors of hyperphosphorylation of tau-protein and neuron death. In this review we analyze the papers describing the increasing of intensity of biosynthesis in neurons in normal conditions and under the stress, the possibility of development of energetic unbalanced neurons and activation of their protective systems. Phosphorylation and hyperphosphorylation of tau-proteins is also tightly connected with protective mechanisms of cells and with processes of evacuation of phosphates, adenosine mono-phosphates and pyrophosphates from the region of protein synthesis. Upon long and high intensity of protein synthesis the protective mechanisms are overloaded and the complementarity of metabolitic processes is disturbed. This results in dysfunction of neurons, transport collapse, and neuron death.
Collapse
Affiliation(s)
- A.V. Maltsev
- Russian Gerontological Research Clinical Center, Russian Ministry of Health Care; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences
| | | | - V.K. Uteshev
- Institute of Biophysics Cell, Russian Academy of Sciences
| | - V.V. Sokolik
- Institute of Neurology, Psychiatry and Addiction Medical Sciences of Ukraine
| | | | - M.A. Yakushin
- Russian Gerontological Research Clinical Center, Russian Ministry of Health Care
| | - N.M. Sokolova
- Russian Gerontological Research Clinical Center, Russian Ministry of Health Care
| | - A.K. Surin
- Insitute of Protein Research, Russian Academy of Sciences; State Research Center for Applied Microbiology & Biotechnology
| | | |
Collapse
|
510
|
Jinwal UK, Akoury E, Abisambra JF, O'Leary JC, Thompson AD, Blair LJ, Jin Y, Bacon J, Nordhues BA, Cockman M, Zhang J, Li P, Zhang B, Borysov S, Uversky VN, Biernat J, Mandelkow E, Gestwicki JE, Zweckstetter M, Dickey CA. Imbalance of Hsp70 family variants fosters tau accumulation. FASEB J 2012; 27:1450-9. [PMID: 23271055 DOI: 10.1096/fj.12-220889] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Dysfunctional tau accumulation is a major contributing factor in tauopathies, and the heat-shock protein 70 (Hsp70) seems to play an important role in this accumulation. Several reports suggest that Hsp70 proteins can cause tau degradation to be accelerated or slowed, but how these opposing activities are controlled is unclear. Here we demonstrate that highly homologous variants in the Hsp70 family can have opposing effects on tau clearance kinetics. When overexpressed in a tetracycline (Tet)-based protein chase model, constitutive heat shock cognate 70 (Hsc70) and inducible Hsp72 slowed or accelerated tau clearance, respectively. Tau synergized with Hsc70, but not Hsp72, to promote microtubule assembly at nearly twice the rate of either Hsp70 homologue in reconstituted, ATP-regenerating Xenopus extracts supplemented with rhodamine-labeled tubulin and human recombinant Hsp72 and Hsc70. Nuclear magnetic resonance spectroscopy with human recombinant protein revealed that Hsp72 had greater affinity for tau than Hsc70 (I/I0 ratio difference of 0.3), but Hsc70 was 30 times more abundant than Hsp72 in human and mouse brain tissue. This indicates that the predominant Hsp70 variant in the brain is Hsc70, suggesting that the brain environment primarily supports slower tau clearance. Despite its capacity to clear tau, Hsp72 was not induced in the Alzheimer's disease brain, suggesting a mechanism for age-associated onset of the disease. Through the use of chimeras that blended the domains of Hsp72 and Hsc70, we determined that the reason for these differences between Hsc70 and Hsp72 with regard to tau clearance kinetics lies within their C-terminal domains, which are essential for their interactions with substrates and cochaperones. Hsp72 but not Hsc70 in the presence of tau was able to recruit the cochaperone ubiquitin ligase CHIP, which is known to facilitate the ubiquitination of tau, describing a possible mechanism of how the C-termini of these homologous Hsp70 variants can differentially regulate tau triage. Thus, efforts to promote Hsp72 expression and inhibit Hsc70 could be therapeutically relevant for tauopathies.
Collapse
Affiliation(s)
- Umesh K Jinwal
- Department of Pharmaceutical Sciences, University of South Florida Health Byrd Alzheimer's Institute, University of South Florida, Tampa, Florida 33613, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
511
|
Liu C, Zhao M, Jiang L, Cheng PN, Park J, Sawaya MR, Pensalfini A, Gou D, Berk AJ, Glabe CG, Nowick J, Eisenberg D. Out-of-register β-sheets suggest a pathway to toxic amyloid aggregates. Proc Natl Acad Sci U S A 2012; 109:20913-8. [PMID: 23213214 PMCID: PMC3529048 DOI: 10.1073/pnas.1218792109] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although aberrant protein aggregation has been conclusively linked to dozens of devastating amyloid diseases, scientists remain puzzled about the molecular features that render amyloid fibrils or small oligomers toxic. Here, we report a previously unobserved type of amyloid fibril that tests as cytotoxic: one in which the strands of the contributing β-sheets are out of register. In all amyloid fibrils previously characterized at the molecular level, only in-register β-sheets have been observed, in which each strand makes its full complement of hydrogen bonds with the strands above and below it in the fibril. In out-of-register sheets, strands are sheared relative to one another, leaving dangling hydrogen bonds. Based on this finding, we designed out-of-register β-sheet amyloid mimics, which form both cylindrin-like oligomers and fibrils, and these mimics are cytotoxic. Structural and energetic considerations suggest that out-of-register fibrils can readily convert to toxic cylindrins. We propose that out-of-register β-sheets and their related cylindrins are part of a toxic amyloid pathway, which is distinct from the more energetically favored in-register amyloid pathway.
Collapse
Affiliation(s)
- Cong Liu
- aUCLA-DOE Institute for Genomics and Proteomics, The Howard Hughes Medical Institute, Molecular Biology Institute and
| | - Minglei Zhao
- aUCLA-DOE Institute for Genomics and Proteomics, The Howard Hughes Medical Institute, Molecular Biology Institute and
| | - Lin Jiang
- aUCLA-DOE Institute for Genomics and Proteomics, The Howard Hughes Medical Institute, Molecular Biology Institute and
| | | | - Jiyong Park
- aUCLA-DOE Institute for Genomics and Proteomics, The Howard Hughes Medical Institute, Molecular Biology Institute and
| | - Michael R. Sawaya
- aUCLA-DOE Institute for Genomics and Proteomics, The Howard Hughes Medical Institute, Molecular Biology Institute and
| | - Anna Pensalfini
- cMolecular Biology and Biochemistry, University of California, Irvine, CA 92697-2025
| | - Dawei Gou
- dMolecular Biology Institute and Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095; and
| | - Arnold J. Berk
- dMolecular Biology Institute and Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095; and
| | - Charles G. Glabe
- cMolecular Biology and Biochemistry, University of California, Irvine, CA 92697-2025
| | - James Nowick
- Departments of bChemistry and
- 3To whom correspondence may be addressed. E-mail: or
| | - David Eisenberg
- aUCLA-DOE Institute for Genomics and Proteomics, The Howard Hughes Medical Institute, Molecular Biology Institute and
- 3To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
512
|
Bioinformatics aggregation predictors in the study of protein conformational diseases of the human nervous system. Electrophoresis 2012; 33:3669-79. [DOI: 10.1002/elps.201200290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 07/04/2012] [Accepted: 07/19/2012] [Indexed: 11/07/2022]
|
513
|
Messing L, Decker JM, Joseph M, Mandelkow E, Mandelkow EM. Cascade of tau toxicity in inducible hippocampal brain slices and prevention by aggregation inhibitors. Neurobiol Aging 2012; 34:1343-1354. [PMID: 23158765 DOI: 10.1016/j.neurobiolaging.2012.10.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/10/2012] [Accepted: 10/23/2012] [Indexed: 11/18/2022]
Abstract
Mislocalization and aggregation of the axonal protein tau are hallmarks of Alzheimer's disease and other tauopathies. Here, we studied the relationship between tau aggregation, loss of spines and neurons, and reversibility by aggregation inhibitors. To this end we established an in vitro model of tauopathy based on regulatable transgenic hippocampal organotypic slice cultures prepared from mice expressing proaggregant Tau repeat domain with mutation ΔK280 (Tau(RD)ΔK). Transgene expression was monitored by a bioluminescence reporter assay. We observed abnormal tau phosphorylation and mislocalization of exogenous and endogenous tau into the somatodendritic compartment. This was paralleled by a reduction of dendritic spines, altered dendritic spine morphology, dysregulation of Ca(++) dynamics and elevated activation of microglia. Neurotoxicity was mediated by Caspase-3 activation and correlated with the expression level of proaggregant Tau(RD)ΔK. Finally, tau aggregates appeared in areas CA1 and CA3 after three weeks in vitro. Neurodegeneration was relieved by aggregation inhibitors or by switching off transgene expression. Thus the slice culture model is suitable for monitoring the development of tauopathy and the therapeutic benefit of antiaggregation drugs.
Collapse
Affiliation(s)
- Lars Messing
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Jochen Martin Decker
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Maria Joseph
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
- Max-Planck-Institute for Neurological Research (Cologne), Hamburg Outstation, c/o DESY, Notkestrasse 85, 22607 Hamburg
| | - Eva-Maria Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
- Max-Planck-Institute for Neurological Research (Cologne), Hamburg Outstation, c/o DESY, Notkestrasse 85, 22607 Hamburg
| |
Collapse
|
514
|
Flach K, Hilbrich I, Schiffmann A, Gärtner U, Krüger M, Leonhardt M, Waschipky H, Wick L, Arendt T, Holzer M. Tau oligomers impair artificial membrane integrity and cellular viability. J Biol Chem 2012; 287:43223-33. [PMID: 23129775 DOI: 10.1074/jbc.m112.396176] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The microtubule-associated protein Tau is mainly expressed in neurons, where it binds and stabilizes microtubules. In Alzheimer disease and other tauopathies, Tau protein has a reduced affinity toward microtubules. As a consequence, Tau protein detaches from microtubules and eventually aggregates into β-sheet-containing filaments. The fibrillization of monomeric Tau to filaments is a multistep process that involves the formation of various aggregates, including spherical and protofibrillar oligomers. Previous concepts, primarily developed for Aβ and α-synuclein, propose these oligomeric intermediates as the primary cytotoxic species mediating their deleterious effects through membrane permeabilization. In the present study, we thus analyzed whether this concept can also be applied to Tau protein. To this end, viability and membrane integrity were assessed on SH-SY5Y neuroblastoma cells and artificial phospholipid vesicles, treated with Tau monomers, Tau aggregation intermediates, or Tau fibrils. Our findings suggest that oligomeric Tau aggregation intermediates are the most toxic compounds of Tau fibrillogenesis, which effectively decrease cell viability and increase phospholipid vesicle leakage. Our data integrate Tau protein into the class of amyloidogenic proteins and enforce the hypothesis of a common toxicity-mediating mechanism for amyloidogenic proteins.
Collapse
Affiliation(s)
- Katharina Flach
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, Faculty of Medicine, Jahnallee 59, University of Leipzig, 04109 Leipzig, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
515
|
Nurbaiti S, Martoprawiro MA, Akhmaloka, Hertadi R. The role of electrostatic interactions on klentaq1 insight for domain separation. Bioinform Biol Insights 2012; 6:225-34. [PMID: 23136465 PMCID: PMC3491847 DOI: 10.4137/bbi.s9390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We investigated the relationship between the thermostability of Klentaq1 and factors stabilizing interdomain interactions. When thermal adaptation of Klentaq1 was analyzed at the atomic level, the protein was stable at 300 and 350 K. It gradually unfolded at 373 K and almost spontaneously unfolded at 400 K. Domain separation was induced by disrupting electrostatic interactions in two salt bridges formed by Lys354-Glu445 and Asp371-Arg435 on the interface domain. The role of these interactions in protein stability was evaluated by comparing free energy solvation (ΔΔGsolv) between wild type and mutants. Substitution of Asp371 by Glu or Asn, and also Glu445 by Asn resulted in a positive value of ΔΔGsolv, suggesting that mutations destabilized the protein structure. Nevertheless, substitution of Glu445 by Asp gave a negative value to ΔΔGsolv reflecting increasing protein stability. Our results demonstrate that interactions at the interface domains of Klentaq1 are essential factors correlated with the Klentaq1 thermostability.
Collapse
Affiliation(s)
- Santi Nurbaiti
- Biochemistry Research Group, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Indonesia
| | | | | | | |
Collapse
|
516
|
Combs B, Gamblin TC. FTDP-17 tau mutations induce distinct effects on aggregation and microtubule interactions. Biochemistry 2012; 51:8597-607. [PMID: 23043292 DOI: 10.1021/bi3010818] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
FTDP-17 mutations in the tau gene lead to early onset frontotemporal dementias characterized by the pathological aggregation of the microtubule-associated protein tau. Tau aggregation is closely correlated with the progression and severity of localized atrophy of certain regions in the brain. These mutations are primarily located in or near the microtubule-binding repeat regions of tau and can have vastly different effects on the protein. Some mutations have been linked to effects such as increased levels of aggregation, hyperphosphorylation, defects in mRNA splicing, and weakened interaction with microtubules. Given the differential effects of the mutations, it may not be surprising that the pathology associated with FTDP-17 can vary widely as well. Despite this variety, several of the mutations are commonly used interchangeably as aggregation inducers for in vitro and in vivo models of tauopathies. We generated recombinant forms of 12 FTDP-17 mutations chosen for their predicted effects on the charge, hydrophobicity, and secondary structure of the protein. We then examined the effects that the mutations had on the properties of in vitro aggregation of the protein and its ability to stabilize microtubule assembly. The group of mutations induced very different effects on the total amount of aggregation, the kinetics of aggregation, and filament morphology. Several of the mutations inhibited the microtubule stabilization ability of tau, while others had very little effect compared to wild-type tau. These results indicate that the mechanisms of disease progression may differ among FTDP-17 mutations and that the effects of the varying mutations may not be equal in all model systems.
Collapse
Affiliation(s)
- Benjamin Combs
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | | |
Collapse
|
517
|
Elbaum-Garfinkle S, Rhoades E. Identification of an aggregation-prone structure of tau. J Am Chem Soc 2012; 134:16607-13. [PMID: 22998648 DOI: 10.1021/ja305206m] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aggregation and deposition of normally soluble proteins is the hallmark of several devastating neurodegenerative disorders. For proteins such as tau in Alzheimer's disease and α-synuclein in Parkinson's disease, aggregation involves a transition from an intrinsically disordered monomer to a highly structured fiber. While understanding the role of these proteins in neurodegeneration requires elucidation of the structural basis of self-association, the conformational heterogeneity of disordered proteins makes their structural characterization inherently challenging. Here we use single molecule Förster resonance energy transfer to measure the conformational ensemble of tau in the absence and presence of heparin to identify critical conformational changes relevant to the initiation of aggregation. We find that different domains of tau display distinct conformational properties that are strongly correlated with their degree of disorder and that may relate to their roles in aggregation. Moreover, we observe that heparin binding induces a distinct two-state structural transition in tau characterized by a loss of long-range contacts and a concomitant compaction of the microtubule binding domain. Our results describe a conformational intermediate of tau that precedes the formation of aggregates and could serve as a target for tau-focused therapeutics.
Collapse
Affiliation(s)
- Shana Elbaum-Garfinkle
- Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, Connecticut 06520-8114, USA
| | | |
Collapse
|
518
|
Cheon M, Chang I, Hall CK. Influence of temperature on formation of perfect tau fragment fibrils using PRIME20/DMD simulations. Protein Sci 2012; 21:1514-27. [PMID: 22887126 PMCID: PMC3526993 DOI: 10.1002/pro.2141] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/06/2012] [Indexed: 02/06/2023]
Abstract
We investigate the fibrillization process for amyloid tau fragment peptides (VQIVYK) by applying the discontinuous molecular dynamics method to a system of 48 VQIVYK peptides modeled using a new protein model/force field, PRIME20. The aim of the article is to ascertain which factors are most important in determining whether or not a peptide system forms perfect coherent fibrillar structures. Two different directional criteria are used to determine when a hydrogen bond occurs: the original H-bond constraints and a parallel preference H-bond constraint that imparts a slight bias towards the formation of parallel versus antiparallel strands in a β-sheet. Under the original H-bond constraints, the resulting fibrillar structures contain a mixture of parallel and antiparallel pairs of strands within each β-sheet over the whole fibrillization temperature range. Under the parallel preference H-bond constraints, the β-sheets within the fibrillar structures are more likely to be parallel and indeed become perfectly parallel, consistent with X-ray crystallography, at a high temperature slightly below the fibrillization temperature. The high temperature environment encourages the formation of perfect fibril structures by providing enough time and space for peptides to rearrange during the aggregation process. There are two different kinetic mechanisms, template assembly with monomer addition at high temperature and merging/rearrangement without monomer addition at low temperature, which lead to significant differences in the final fibrillar structure. This suggests that the diverse fibril morphologies generally observed in vitro depend more on environmental conditions than has heretofore been appreciated.
Collapse
Affiliation(s)
- Mookyung Cheon
- Center for Proteome Biophysics, Department of Physics, Pusan National UniversityBusan 609-735, Korea
| | - Iksoo Chang
- Center for Proteome Biophysics, Department of Physics, Pusan National UniversityBusan 609-735, Korea
| | - Carol K Hall
- Department of Chemical and Biomolecular Engineering, North Carolina State UniversityRaleigh, North Carolina 27695-7905
| |
Collapse
|
519
|
Limited cleavage of tau with matrix-metalloproteinase MMP-9, but not MMP-3, enhances tau oligomer formation. Exp Neurol 2012; 237:470-6. [DOI: 10.1016/j.expneurol.2012.07.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 07/15/2012] [Accepted: 07/31/2012] [Indexed: 11/18/2022]
|
520
|
Ciasca G, Campi G, Battisti A, Rea G, Rodio M, Papi M, Pernot P, Tenenbaum A, Bianconi A. Continuous thermal collapse of the intrinsically disordered protein tau is driven by its entropic flexible domain. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:13405-13410. [PMID: 22891813 DOI: 10.1021/la302628y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The tau protein belongs to the category of Intrinsically Disordered Proteins (IDP), which in their native state lack a folded structure and fluctuate between many conformations. In its physiological state, tau helps nucleating and stabilizing the microtubules' (MTs) surfaces in the axons of the neurons. Tau is mainly composed by two domains: (i) the binding domain that tightly bounds the MT surfaces and (ii) the projection domain that exerts a long-range entropic repulsive force and thus provides the proper spacing between adjacent MTs. Tau is also involved in the genesis and in the development of the Alzheimer disease when it detaches from MT surfaces and aggregates in paired helical filaments. Unfortunately, the molecular mechanisms behind these phenomena are still unclear. Temperature variation, rarely considered in biological studies, is here used to provide structural information on tau correlated to its role as an entropic spacer between adjacent MTs surfaces. In this paper, by means of small-angle X-ray scattering and molecular dynamics simulation, we demonstrate that tau undergoes a counterintuitive collapse phenomenon with increasing temperature. A detailed analysis of our results, performed by the Ensemble Optimization Method, shows that the thermal collapse is coupled to the occurrence of a transient long-range contact between a region encompassing the end of the proline-rich domain P2 and the first part of the repeats domain, and the region of the N-terminal domain entailing residues 80-150. Interestingly these two regions involved in the tau temperature collapse belong to the flexible projection domain that acts as an entropic bristle and regulates the MTs' architecture. Our results show that temperature is an important parameter that influences the dynamics of the tau projection domain, and hence its entropic behavior.
Collapse
|
521
|
Cheng PN, Spencer R, Woods RJ, Glabe CG, Nowick JS. Heterodivalent linked macrocyclic β-sheets with enhanced activity against Aβ aggregation: two sites are better than one. J Am Chem Soc 2012; 134:14179-84. [PMID: 22827298 DOI: 10.1021/ja305416a] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This paper reports a series of heterodivalent linked macrocyclic β-sheets 6 that are not only far more active against amyloid-β (Aβ) aggregation than their monovalent components 1a and 1b but also are dramatically more active than their homodivalent counterparts 4 and 5. The macrocyclic β-sheet components 1a and 1b comprise pentapeptides derived from the N- and C-terminal regions of Aβ and molecular template and turn units that enforce a β-sheet structure and block aggregation. Thioflavin T fluorescence assays show that heterodivalent linked macrocyclic β-sheets 6 delay Aβ(1-40) aggregation 6-8-fold at equimolar concentrations and substantially delay aggregation at substoichiometric concentrations, while homodivalent linked macrocyclic β-sheets 4 and 5 and monovalent macrocyclic β-sheets 1a and 1b only exhibit more modest effects at equimolar or greater concentrations. A model to explain these observations is proposed, in which the inhibitors bind to and stabilize the early β-structured Aβ oligomers and thus delay aggregation. In this model, heterodivalent linked macrocyclic β-sheets 6 bind to the β-structured oligomers more strongly, because N-terminal-derived component 1a can bind to the N-terminal-based core of the β-structured oligomers, while the C-terminal-derived component 1b can achieve additional interactions with the C-terminal region of Aβ. The enhanced activity of the heterodivalent compounds suggests that polyvalent inhibitors that can target multiple regions of amyloidogenic peptides and proteins are better than those that only target a single region.
Collapse
Affiliation(s)
- Pin-Nan Cheng
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, USA
| | | | | | | | | |
Collapse
|
522
|
Delphin C, Bouvier D, Seggio M, Couriol E, Saoudi Y, Denarier E, Bosc C, Valiron O, Bisbal M, Arnal I, Andrieux A. MAP6-F is a temperature sensor that directly binds to and protects microtubules from cold-induced depolymerization. J Biol Chem 2012; 287:35127-35138. [PMID: 22904321 DOI: 10.1074/jbc.m112.398339] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microtubules are dynamic structures that present the peculiar characteristic to be ice-cold labile in vitro. In vivo, microtubules are protected from ice-cold induced depolymerization by the widely expressed MAP6/STOP family of proteins. However, the mechanism by which MAP6 stabilizes microtubules at 4 °C has not been identified. Moreover, the microtubule cold sensitivity and therefore the needs for microtubule stabilization in the wide range of temperatures between 4 and 37 °C are unknown. This is of importance as body temperatures of animals can drop during hibernation or torpor covering a large range of temperatures. Here, we show that in the absence of MAP6, microtubules in cells below 20 °C rapidly depolymerize in a temperature-dependent manner whereas they are stabilized in the presence of MAP6. We further show that in cells, MAP6-F binding to and stabilization of microtubules is temperature- dependent and very dynamic, suggesting a direct effect of the temperature on the formation of microtubule/MAP6 complex. We also demonstrate using purified proteins that MAP6-F binds directly to microtubules through its Mc domain. This binding is temperature-dependent and coincides with progressive conformational changes of the Mc domain as revealed by circular dichroism. Thus, MAP6 might serve as a temperature sensor adapting its conformation according to the temperature to maintain the cellular microtubule network in organisms exposed to temperature decrease.
Collapse
Affiliation(s)
- Christian Delphin
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France.
| | - Denis Bouvier
- the European Molecular Biology Laboratory, Grenoble Outstation, 6 rue Jules Horowitz, BP181, 38042 Grenoble Cedex 9, France
| | - Maxime Seggio
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Emilie Couriol
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Yasmina Saoudi
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Eric Denarier
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Christophe Bosc
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Odile Valiron
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Mariano Bisbal
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Isabelle Arnal
- Team 13 Dynamic and Structural Regulation of Cytoskeleton, Institut National de la Santé et de la Recherche Médicale, U836-GIN, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| | - Annie Andrieux
- Team 1 Physiopathology of Cytoskeleton; Commissariat à I'Energie Atomique, Institut National de la Santé et de la Recherche Médicale, U836-GIN iRTSV-GPC, Site Santé La Tronche, BP170, 38042 Grenoble, Cedex 9, France
| |
Collapse
|
523
|
Daebel V, Chinnathambi S, Biernat J, Schwalbe M, Habenstein B, Loquet A, Akoury E, Tepper K, Müller H, Baldus M, Griesinger C, Zweckstetter M, Mandelkow E, Vijayan V, Lange A. β-Sheet Core of Tau Paired Helical Filaments Revealed by Solid-State NMR. J Am Chem Soc 2012; 134:13982-9. [DOI: 10.1021/ja305470p] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Venita Daebel
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Subashchandrabose Chinnathambi
- DZNE, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee
2, 53175 Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175
Bonn, Germany
| | - Jacek Biernat
- DZNE, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee
2, 53175 Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175
Bonn, Germany
| | - Martin Schwalbe
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Birgit Habenstein
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Antoine Loquet
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Elias Akoury
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Katharina Tepper
- DZNE, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee
2, 53175 Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175
Bonn, Germany
| | - Henrik Müller
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Marc Baldus
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Christian Griesinger
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Markus Zweckstetter
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Grisebachstraße
5, 37077 Göttingen, Germany
| | - Eckhard Mandelkow
- DZNE, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee
2, 53175 Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175
Bonn, Germany
| | - Vinesh Vijayan
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| | - Adam Lange
- NMR-based
Structural Biology, Max Planck Institute for Biophysical Chemistry, Am
Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
524
|
Jeganathan S, Chinnathambi S, Mandelkow EM, Mandelkow E. Conformations of microtubule-associated protein Tau mapped by fluorescence resonance energy transfer. Methods Mol Biol 2012; 849:85-99. [PMID: 22528085 DOI: 10.1007/978-1-61779-551-0_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
The microtubule-associated protein Tau plays a physiological role of stabilizing neuronal microtubules by binding to their lateral surface. Tau belongs to the category of natively unfolded protein as it shows typical features of random coil, as analyzed by various biophysical techniques. In cells, it is subjected to several posttranslational modifications (e.g., phosphorylation, cleavage, ubiquitination, and glycosylation). In neurodegenerative diseases, Tau forms insoluble aggregates called paired helical filaments (PHFs). We have applied fluorescence resonance energy transfer (FRET) to examine the conformations of soluble Tau. We created a series of Tau mutants, each carrying one tryptophan and one cysteine (labeled by IEADANS). This made it possible to measure the distance between these FRET pairs placed in different domains of Tau. This approach enables one to analyze the global folding of soluble Tau and its alteration upon phosphorylation and denaturation.
Collapse
|
525
|
Abstract
Atomic force microscopy (AFM) has been used in numerous studies to visualize and analyze the structure and conformation of biological samples, from single molecules to biopolymers to cells. The possibility to analyze native samples without fixation, staining and in physiological buffer conditions, combined with the sub-nanometer resolution, makes AFM a versatile tool for the analysis of protein aggregation and amyloid structures. Here, we describe the application of AFM to study fibrillar Tau protein aggregates.
Collapse
|
526
|
Ishida T. [Overview of structural study on conformations and intermolecular interactions of biomolecules]. YAKUGAKU ZASSHI 2012; 132:785-816. [PMID: 22790026 DOI: 10.1248/yakushi.132.785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Information on the conformational feature and specific intermolecular interaction of biomolecules is important to understand the biological function and to develop device for treating disorder caused by the abnormal function. Thus the 3D structures of the biologically active molecules and the specific interactions with their target molecules at the atomic level have been investigated by various physicochemical approaches. Herein, the following five subjects are reviewed: (1) function-linked conformations of biomolecules including natural annular products, opioid peptides and neuropeptides; (2) π-π stacking interactions of tryptophan derivatives with coenzymes and nucleic acid bases; (3) mRNA cap recognition of eukaryotic initiation factor 4E and its regulation by 4E-binding protein; (4) conformational feature of histamine H2 receptor antagonists and design of cathepsin B inhibitors; (5) self-aggregation mechanism of tau protein and its inhibition.
Collapse
Affiliation(s)
- Toshimasa Ishida
- Laboratory of Physical Chemistry, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan.
| |
Collapse
|
527
|
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
|
528
|
Meng SR, Zhu YZ, Guo T, Liu XL, Chen J, Liang Y. Fibril-forming motifs are essential and sufficient for the fibrillization of human Tau. PLoS One 2012; 7:e38903. [PMID: 22701727 PMCID: PMC3372541 DOI: 10.1371/journal.pone.0038903] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 05/14/2012] [Indexed: 11/29/2022] Open
Abstract
Background The misfolding of amyloidogenic proteins including human Tau protein, human prion protein, and human α-synuclein is involved in neurodegenerative diseases such as Alzheimer disease, prion disease, and Parkinson disease. Although a lot of research on such amyloidogenic proteins has been done, we do not know the determinants that drive these proteins to form fibrils and thereby induce neurodegenerative diseases. In this study, we want to know the role of fibril-forming motifs from such amyloidogenic proteins in the fibrillization of human Tau protein. Methodology/Principal Findings As evidenced by thioflavin T binding and turbidity assays, transmission electron microscopy, and circular dichroism, fibril-forming motifs are essential and sufficient for the fibrillization of microtubule-associated protein Tau: only when both of its fibril-forming motifs, PHF6 and PHF6*, are deleted can recombinant human Tau fragment Tau244–372 lose its ability to form fibrils, and the insertion of unrelated fibril-forming motifs from other amyloidogenic proteins, such as human prion protein, yeast prion protein, human α-synuclein, and human amyloid β, into the disabled Tau protein can retrieve its ability to form fibrils. Furthermore, this retrieval is independent of the insertion location on Tau244–372. Conclusions/Significance We demonstrate for the first time that insertion of fibril-forming motifs can replace PHF6/PHF6* motifs, driving human Tau protein to form fibrils with different morphologies and different kinetic parameters. Our results suggest that fibril-forming motifs play a key role in the fibrillization of human Tau protein and could be the determinants of amyloidogenic proteins tending to misfold, thereby causing the initiation and development of neurodegenerative diseases. Our study also touches on the importance of amyloid “strains”: changes to the amyloidgenic driver region results in altered structural morphologies at the macromolecular level.
Collapse
Affiliation(s)
- Sheng-Rong Meng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying-Zhu Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Tong Guo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xiao-Ling Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jie Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yi Liang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
529
|
Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, Greco A, Seripa D, Pilotto A. Immunotherapy for Alzheimer's disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 2012; 4:213-38. [PMID: 22339463 DOI: 10.2217/imt.11.170] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The exact mechanisms leading to Alzheimer's disease (AD) are largely unknown, limiting the identification of effective disease-modifying therapies. The two principal neuropathological hallmarks of AD are extracellular β-amyloid (Aβ), peptide deposition (senile plaques) and intracellular neurofibrillary tangles containing hyperphosphorylated tau protein. During the last decade, most of the efforts of the pharmaceutical industry were directed against the production and accumulation of Aβ. The most innovative of the pharmacological approaches was the stimulation of Aβ clearance from the brain of AD patients via the administration of Aβ antigens (active vaccination) or anti-Aβ antibodies (passive vaccination). Several active and passive anti-Aβ vaccines are under clinical investigation. Unfortunately, the first active vaccine (AN1792, consisting of preaggregate Aβ and an immune adjuvant, QS-21) was abandoned because it caused meningoencephalitis in approximately 6% of treated patients. Anti-Aβ monoclonal antibodies (bapineuzumab and solanezumab) are now being developed. The clinical results of the initial studies with bapineuzumab were equivocal in terms of cognitive benefit. The occurrence of vasogenic edema after bapineuzumab, and more rarely brain microhemorrhages (especially in Apo E ε4 carriers), has raised concerns on the safety of these antibodies directed against the N-terminus of the Aβ peptide. Solanezumab, a humanized anti-Aβ monoclonal antibody directed against the midregion of the Aβ peptide, was shown to neutralize soluble Aβ species. Phase II studies showed a good safety profile of solanezumab, while studies on cerebrospinal and plasma biomarkers documented good signals of pharmacodynamic activity. Although some studies suggested that active immunization may be effective against tau in animal models of AD, very few studies regarding passive immunization against tau protein are currently available. The results of the large, ongoing Phase III trials with bapineuzumab and solanezumab will tell us if monoclonal anti-Aβ antibodies may slow down the rate of deterioration of AD. Based on the new diagnostic criteria of AD and on recent major failures of anti-Aβ drugs in mild-to-moderate AD patients, one could argue that clinical trials on potential disease-modifying drugs, including immunological approaches, should be performed in the early stages of AD.
Collapse
Affiliation(s)
- Francesco Panza
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, Foggia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
530
|
Sogawa K, Okuda R, In Y, Ishida T, Taniguchi T, Minoura K, Tomoo K. C-H ... π interplay between Ile308 and Tyr310 residues in the third repeat of microtubule binding domain is indispensable for self-assembly of three- and four-repeat tau. J Biochem 2012; 152:221-9. [PMID: 22659094 DOI: 10.1093/jb/mvs061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Information on the structural scaffold for tau aggregation is important in developing a method of preventing Alzheimer's disease (AD). Tau contains a microtubule binding domain (MBD) consisting of three or four repeats of 31 and 32 similar residues in its C-terminal half. Although the key event in tau aggregation has been considered to be the formation of β-sheet structures from a short hexapeptide (306)VQIVYK(311) in the third repeat of MBD, its aggregation pathway to filament formation differs between the three- and four-repeated MBDs, owing to the intermolecular and intramolecular disulphide bond formations, respectively. Therefore, the elucidation of a common structural element necessary for the self-assembly of three-/four-repeated full-length tau is an important research subject. Expanding the previous results on the aggregation mechanism of MBD, in this paper, we report that the C-H … π interaction between the Ile308 and Tyr310 side chains in the third repeat of MBD is indispensable for the self-assembly of three-/four-repeated full-length tau, where the interaction provides a conformational seed for triggering the molecular association. On the basis of the aggregation behaviours of a series of MBD and full-length tau mutants, a possible self-association model of tau is proposed and the relationship between the aggregation form (filament or granule) and the association pathway is discussed.
Collapse
Affiliation(s)
- Koushirou Sogawa
- Department of Physical Chemistry, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-11, Japan
| | | | | | | | | | | | | |
Collapse
|
531
|
Battisti A, Ciasca G, Grottesi A, Bianconi A, Tenenbaum A. Temporary secondary structures in tau, an intrinsically disordered protein. MOLECULAR SIMULATION 2012. [DOI: 10.1080/08927022.2011.633347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
532
|
Structure and pathology of tau protein in Alzheimer disease. Int J Alzheimers Dis 2012; 2012:731526. [PMID: 22690349 PMCID: PMC3368361 DOI: 10.1155/2012/731526] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia. In connection with the global trend of prolonging human life and the increasing number of elderly in the population, the AD becomes one of the most serious health and socioeconomic problems of the present. Tau protein promotes assembly and stabilizes microtubules, which contributes to the proper function of neuron. Alterations in the amount or the structure of tau protein can affect its role as a stabilizer of microtubules as well as some of the processes in which it is implicated. The molecular mechanisms governing tau aggregation are mainly represented by several posttranslational modifications that alter its structure and conformational state. Hence, abnormal phosphorylation and truncation of tau protein have gained attention as key mechanisms that become tau protein in a pathological entity. Evidences about the clinicopathological significance of phosphorylated and truncated tau have been documented during the progression of AD as well as their capacity to exert cytotoxicity when expressed in cell and animal models. This paper describes the normal structure and function of tau protein and its major alterations during its pathological aggregation in AD.
Collapse
|
533
|
Assigning backbone NMR resonances for full length tau isoforms: efficient compromise between manual assignments and reduced dimensionality. PLoS One 2012; 7:e34679. [PMID: 22529924 PMCID: PMC3329490 DOI: 10.1371/journal.pone.0034679] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 03/08/2012] [Indexed: 11/19/2022] Open
Abstract
Tau protein is the longest disordered protein for which nearly complete backbone NMR resonance assignments have been reported. Full-length tau protein was initially assigned using a laborious combination of bootstrapping assignments from shorter tau fragments and conventional triple resonance NMR experiments. Subsequently it was reported that assignments of comparable quality could be obtained in a fully automated fashion from data obtained using reduced dimensionality NMR (RDNMR) experiments employing a large number of indirect dimensions. Although the latter strategy offers many advantages, it presents some difficulties if manual intervention, confirmation, or correction of the assignments is desirable, as may often be the case for long disordered and degenerate polypeptide sequences. Here we demonstrate that nearly complete backbone resonance assignments for full-length tau isoforms can be obtained without resorting either to bootstrapping from smaller fragments or to very high dimensionality experiments and automation. Instead, a set of RDNMR triple resonance experiments of modest dimensionality lend themselves readily to efficient and unambiguous manual assignments. An analysis of the backbone chemical shifts obtained in this fashion indicates several regions in full length tau with a notable propensity for helical or strand-like structure that are in good agreement with previous observations.
Collapse
|
534
|
Künze G, Barré P, Scheidt HA, Thomas L, Eliezer D, Huster D. Binding of the three-repeat domain of tau to phospholipid membranes induces an aggregated-like state of the protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2302-13. [PMID: 22521809 DOI: 10.1016/j.bbamem.2012.03.019] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 12/21/2022]
Abstract
In patients with Alzheimer's disease, the microtubule-associated protein tau is found aggregated into paired helical filaments (PHFs) in neurofibrillary deposits. In solution, tau is intrinsically unstructured. However, the tubulin binding domain consisting of three or four 31-32 amino acid repeat regions exhibits both helical and β-structure propensity and makes up the proteolysis resistant core of PHFs. Here, we studied the structure and dynamics of the three-repeat domain of tau (i.e. K19) when bound to membranes consisting of a phosphatidylcholine and phosphatidylserine mixture or phosphatidylserine alone. Tau K19 binds to phospholipid vesicles with submicromolar affinity as measured by fluorescence spectroscopy. The interaction is driven by electrostatic forces between the positively charged protein and the phospholipid head groups. The structure of the membrane-bound state of K19 was studied using CD spectroscopy and solid-state magic-angle spinning NMR spectroscopy. To this end, the protein was selectively (13)C-labeled at all valine and leucine residues. Isotropic chemical shift values of tau K19 were consistent with a β-structure. In addition, motionally averaged (1)H-(13)C dipolar couplings indicated a high rigidity of the protein backbone. The structure formation of K19 was also shown to depend on the charge density of the membrane. Phosphatidylserine membranes induced a gain in the α-helix structure along with an immersion of K19 into the phospholipid bilayer as indicated by a reduction of the lipid chain (2)H NMR order parameter. Our results provide structural insights into the membrane-bound state of tau K19 and support a potential role of phospholipid membranes in mediating the physiological and pathological functions of tau.
Collapse
Affiliation(s)
- Georg Künze
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
535
|
Walker S, Ullman O, Stultz CM. Using intramolecular disulfide bonds in tau protein to deduce structural features of aggregation-resistant conformations. J Biol Chem 2012; 287:9591-600. [PMID: 22291015 PMCID: PMC3308815 DOI: 10.1074/jbc.m111.336107] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/17/2012] [Indexed: 01/11/2023] Open
Abstract
Because tau aggregation likely plays a role in a number of neurodegenerative diseases, understanding the processes that affect tau aggregation is of considerable importance. One factor that has been shown to influence the aggregation propensity is the oxidation state of the protein itself. Tau protein, which contains two naturally occurring cysteine residues, can form both intermolecular disulfide bonds and intramolecular disulfide bonds. Several studies suggest that intermolecular disulfide bonds can promote tau aggregation in vitro. By contrast, although there are data to suggest that intramolecular disulfide bond formation retards tau aggregation in vitro, the precise mechanism underlying this observation remains unclear. While it has been hypothesized that a single intramolecular disulfide bond in tau leads to compact conformations that cannot form extended structure consistent with tau fibrils, there are few data to support this conjecture. In the present study we generate oxidized forms of the truncation mutant, K18, which contains all four microtubule binding repeats, and isolate the monomeric fraction, which corresponds to K18 monomers that have a single intramolecular disulfide bond. We study the aggregation propensity of the oxidized monomeric fraction and relate these data to an atomistic model of the K18 unfolded ensemble. Our results argue that the main effect of intramolecular disulfide bond formation is to preferentially stabilize conformers within the unfolded ensemble that place the aggregation-prone tau subsequences, PHF6* and PHF6, in conformations that are inconsistent with the formation of cross-β-structure. These data further our understanding of the precise structural features that retard tau aggregation.
Collapse
Affiliation(s)
| | | | - Collin M. Stultz
- From the Research Laboratory of Electronics
- the Harvard-MIT Division of Health Sciences and Technology and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| |
Collapse
|
536
|
Pulawski W, Ghoshdastider U, Andrisano V, Filipek S. Ubiquitous amyloids. Appl Biochem Biotechnol 2012; 166:1626-43. [PMID: 22350870 PMCID: PMC3324686 DOI: 10.1007/s12010-012-9549-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 01/05/2012] [Indexed: 01/04/2023]
Abstract
The common view of amyloids and prion proteins is that they are associated with many currently incurable diseases and present a great danger to an organism. This danger comes from the fact that not only prion proteins, but also the infectious form(s) of amyloids, as it has been shown recently, are able to transmit the disease. On the other hand, organisms take advantage of the strength and durability of specific forms of amyloids. Such forms do not spread any disease. Also, in nanotechnology there is a constantly growing need to employ amyloid fibrils in many industrial applications. With increasing knowledge about amyloids and prion proteins we are aware that the amyloidal state is inherent to any protein, making the problem of amyloid formation a central one in aging-related diseases. However, the “good” amyloids can be beneficial and even necessary for our health. Furthermore, because of their mechanical properties, the amyloids are of great interest to engineers.
Collapse
Affiliation(s)
- Wojciech Pulawski
- Faculty of Chemistry, University of Warsaw, ul. Pasteura 1, 02-093 Warsaw, Poland
| | | | | | | |
Collapse
|
537
|
Matthes D, Gapsys V, de Groot BL. Driving forces and structural determinants of steric zipper peptide oligomer formation elucidated by atomistic simulations. J Mol Biol 2012; 421:390-416. [PMID: 22326493 DOI: 10.1016/j.jmb.2012.02.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 01/27/2012] [Accepted: 02/01/2012] [Indexed: 12/20/2022]
Abstract
Understanding the structural and energetic requirements of non-fibrillar oligomer formation harbors the potential to decipher an important yet still elusive part of amyloidogenic peptide and protein aggregation. Low-molecular-weight oligomers are described to be transient and polymorphic intermediates in the nucleated self-assembly process to highly ordered amyloid fibers and were additionally found to exhibit a profound cytotoxicity. However, detailed structural information on the oligomeric species involved in the nucleation cannot be readily inferred from experiments. Here, we study the spontaneous assembly of steric zipper peptides from the tau protein, insulin and α-synuclein with atomistic molecular dynamics simulations on the microsecond timescale. Detailed analysis of the forces driving the oligomerization reveals a common two-step process akin to a general condensation-ordering mechanism and thus provides a rational understanding of the molecular basis of peptide self-assembly. Our results suggest that the initial formation of partially ordered peptide oligomers is governed by the solvation free energy, whereas the dynamical ordering and emergence of β-sheets are mainly driven by optimized inter-peptide interactions in the collapsed state. A novel mapping technique based on collective coordinates is employed to highlight similarities and differences in the conformational ensemble of small oligomer structures. Elucidating the dynamical and polymorphic β-sheet oligomer conformations at atomistic detail furthermore suggests complementary sheet packing characteristics similar to steric zipper structures, but with a larger heterogeneity in the strand alignment pattern and sheet-to-sheet arrangements compared to the cross-β motif found in the fibrillar or crystalline states.
Collapse
Affiliation(s)
- Dirk Matthes
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max-Planck-Institute for Biophysical Chemistry Am Fassberg 11, 37077 Göttingen, Germany
| | | | | |
Collapse
|
538
|
Inoue M, Konno T, Tainaka K, Nakata E, Yoshida HO, Morii T. Positional effects of phosphorylation on the stability and morphology of tau-related amyloid fibrils. Biochemistry 2012; 51:1396-406. [PMID: 22304362 DOI: 10.1021/bi201451z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hyperphosphorylated forms of tau protein are the main component of paired helical filaments (PHFs) of neurofibrillary tangles in the brain of Alzheimer's disease patients. To understand the effect of phosphorylation on the fibrillation of tau, we utilized tau-derived phosphorylated peptides. The V(306)QIVYK(311) sequence (PHF6) in the microtubule-binding domain is known to play a key role in the fibrillation of tau, and the short peptide corresponding to the PHF6 sequence forms amyloid-type fibrils similar to those generated by full-length tau. We focused on the amino acid residue located at the N-terminus of the PHF6 sequence, serine or lysine in the native isoform of tau, and synthesized the PHF6 derivative peptides with serine or lysine at the N-terminus of PHF6. Peptides phosphorylated at serine and/or tyrosine were synthesized to mimic the possible phosphorylation at these positions. The critical concentrations of the fibrillation of peptides were determined to quantitatively assess fibril stability. The peptide with the net charge of near zero tended to form stable fibrils. Interestingly, the peptide phosphorylated at the N-terminal serine residue exhibited remarkably low fibrillation propensity as compared to the peptide possessing the same net charge. Transmission electron microscopy measurements of the fibrils visualized the paired helical or straight fibers and segregated masses of the fibers or heterogeneous rodlike fibers depending on the phosphorylation status. Further analyses of the fibrils by the X-ray fiber diffraction method and Fourier transform infrared spectroscopic measurements indicated that all the peptides shared a common cross-β structure. In addition, the phosphoserine-containing peptides showed the characteristics of β-sandwiches that could interact with both faces of the β-sheet. On the basis of these observations, possible protofilament models with four β-sheets were constructed to consider the positional effects of the serine and/or tyrosine phosphorylations. The electrostatic intersheet interaction between phosphate groups and the amino group of lysine enhanced the lateral association between β-sheets to compensate for the excess charge. In addition to the previously postulated net charge of the peptide, the position of the charged residue plays a critical role in the amyloid fibrillation of tau.
Collapse
Affiliation(s)
- Masafumi Inoue
- Institute of Advanced Energy, Kyoto University, Uji, Kyoto 611-0011, Japan
| | | | | | | | | | | |
Collapse
|
539
|
Voss K, Combs B, Patterson KR, Binder LI, Gamblin TC. Hsp70 alters tau function and aggregation in an isoform specific manner. Biochemistry 2012; 51:888-98. [PMID: 22236337 DOI: 10.1021/bi2018078] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tauopathies are characterized by abnormal aggregation of the microtubule associated protein tau. This aggregation is thought to occur when tau undergoes shifts from its native conformation to one that exposes hydrophobic areas on separate monomers, allowing contact and subsequent association into oligomers and filaments. Molecular chaperones normally function by binding to exposed hydrophobic stretches on proteins and assisting in their refolding. Chaperones of the heat shock protein 70 (Hsp70) family have been implicated in the prevention of abnormal tau aggregation in adult neurons. Tau exists as six alternatively spliced isoforms, and all six isoforms appear capable of forming the pathological aggregates seen in Alzheimer's disease. Because tau isoforms differ in primary sequence, we sought to determine whether Hsp70 would differentially affect the aggregation and microtubule assembly characteristics of the various tau isoforms. We found that Hsp70 inhibits tau aggregation directly and not through inducer-mediated effects. We also determined that Hsp70 inhibits the aggregation of each individual tau isoform and was more effective at inhibiting the three repeat isoforms. Finally, all tau isoforms robustly induced microtubule formation while in the presence of Hsp70. The results presented herein indicate that Hsp70 affects tau isoform dysfunction while having very little impact on the normal function of tau to mediate microtubule assembly. This indicates that targeting Hsp70 to tau may provide a therapeutic approach for the treatment of tauopathies that avoids disruption of normal tau function.
Collapse
Affiliation(s)
- Kellen Voss
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, United States
| | | | | | | | | |
Collapse
|
540
|
Clos AL, Kayed R, Lasagna-Reeves CA. Association of skin with the pathogenesis and treatment of neurodegenerative amyloidosis. Front Neurol 2012; 3:5. [PMID: 22319507 PMCID: PMC3262151 DOI: 10.3389/fneur.2012.00005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 01/04/2012] [Indexed: 12/12/2022] Open
Abstract
Amyloidosis are a large group of conformational diseases characterized by abnormal protein folding and assembly which results in the accumulation of insoluble protein aggregates that may accumulate systemically or locally in certain organs or tissue. In local amyloidosis, amyloid deposits are restricted to a particular organ or tissue. Alzheimer’s, Parkinson’s disease, and amyotrophic lateral sclerosis are some examples of neurodegenerative amyloidosis. Local manifestation of protein aggregation in the skin has also been reported. Brain and skin are highly connected at a physiological and pathological level. Recently several studies demonstrated a strong connection between brain and skin in different amyloid diseases. In the present review, we discuss the relevance of the “brain–skin connection” in different neurodegenerative amyloidosis, not only at the pathological level, but also as a strategy for the treatment of these diseases.
Collapse
Affiliation(s)
- Audra L Clos
- Department of Dermatology, MD Anderson Cancer Center, University of Texas Houston, TX, USA
| | | | | |
Collapse
|
541
|
Abstract
The protein tau is the most abundant microtubule associated protein in the central and peripheral nervous system. In the brain, tau plays a role in the assembly and stabilization of microtubules. The function of tau, however, appears to overlap with other microtubule binding proteins. The observation that tau is associated with neurodegenerative diseases has renewed interest in this protein. Various aspects of structure and biochemistry of tau, fibril formation and clinical perspectives, including therapeutic strategies are reviewed in this chapter.
Collapse
|
542
|
Thomas SN, Funk KE, Wan Y, Liao Z, Davies P, Kuret J, Yang AJ. Dual modification of Alzheimer's disease PHF-tau protein by lysine methylation and ubiquitylation: a mass spectrometry approach. Acta Neuropathol 2012; 123:105-17. [PMID: 22033876 PMCID: PMC3249157 DOI: 10.1007/s00401-011-0893-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 10/13/2011] [Accepted: 10/13/2011] [Indexed: 11/04/2022]
Abstract
In sporadic Alzheimer’s disease (AD), neurofibrillary lesion formation is preceded by extensive post-translational modification of the microtubule associated protein tau. To identify the modification signature associated with tau lesion formation at single amino acid resolution, immunopurified paired helical filaments were isolated from AD brain and subjected to nanoflow liquid chromatography–tandem mass spectrometry analysis. The resulting spectra identified monomethylation of lysine residues as a new tau modification. The methyl-lysine was distributed among seven residues located in the projection and microtubule binding repeat regions of tau protein, with one site, K254, being a substrate for a competing lysine modification, ubiquitylation. To characterize methyl lysine content in intact tissue, hippocampal sections prepared from post mortem late-stage AD cases were subjected to double-label confocal fluorescence microscopy using anti-tau and anti-methyl lysine antibodies. Anti-methyl lysine immunoreactivity colocalized with 78 ± 13% of neurofibrillary tangles in these specimens. Together these data provide the first evidence that tau in neurofibrillary lesions is post-translationally modified by lysine methylation.
Collapse
|
543
|
Abstract
Tauopathies are age-related neurodegenerative diseases that are characterized by the presence of aggregates of abnormally phosphorylated tau. As tau was originally discovered as a microtubule-associated protein, it has been hypothesized that neurodegeneration results from a loss of the ability of tau to associate with microtubules. However, tau has been found to have other functions aside from the promotion and stabilization of microtubule assembly. It is conceivable that such functions may be affected by the abnormal phosphorylation of tau and might have consequences for neuronal function or viability. This chapter provides an overview of tau structure, functions, and its involvement in neurodegenerative diseases.
Collapse
|
544
|
Dolai S, Shi W, Corbo C, Sun C, Averick S, Obeysekera D, Farid M, Alonso A, Banerjee P, Raja K. "Clicked" sugar-curcumin conjugate: modulator of amyloid-β and tau peptide aggregation at ultralow concentrations. ACS Chem Neurosci 2011; 2:694-9. [PMID: 22860163 DOI: 10.1021/cn200088r] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 10/13/2011] [Indexed: 01/24/2023] Open
Abstract
The synthesis of a water/plasma soluble, noncytotoxic, "clicked" sugar-derivative of curcumin with amplified bioefficacy in modulating amyloid-β and tau peptide aggregation is presented. Curcumin inhibits amyloid-β and tau peptide aggregation at micromolar concentrations; the sugar-curcumin conjugate inhibits Aβ and tau peptide aggregation at concentrations as low as 8 nM and 0.1 nM, respectively. In comparison to curcumin, this conveniently synthesized Alzheimer's drug candidate is a more powerful antioxidant.
Collapse
Affiliation(s)
| | - Wei Shi
- Department of Chemistry and Physical
Sciences, Felician College, Lodi, New Jersey
07644, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
545
|
Moore CL, Huang MH, Robbennolt SA, Voss KR, Combs B, Gamblin TC, Goux WJ. Secondary nucleating sequences affect kinetics and thermodynamics of tau aggregation. Biochemistry 2011; 50:10876-86. [PMID: 22085312 DOI: 10.1021/bi2014745] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tau protein was scanned for highly amyloidogenic sequences in amphiphilic motifs (X)(n)Z, Z(X)(n)Z (n ≥ 2), or (XZ)(n) (n ≥ 2), where X is a hydrophobic residue and Z is a charged or polar residue. N-Acetyl peptides homologous to these sequences were used to study aggregation. Transmission electron microscopy (TEM) showed seven peptides, in addition to well-known primary nucleating sequences Ac(275)VQIINK (AcPHF6*) and Ac(306)VQIVYK (AcPHF6), formed fibers, tubes, ribbons, or rolled sheets. Of the peptides shown by TEM to form amyloid, Ac(10)VME, AcPHF6*, Ac(375)KLTFR, and Ac(393)VYK were found to enhance the fraction of β-structure of AcPHF6 formed at equilibrium, and Ac(375)KLTFR was found to inhibit AcPHF6 and AcPHF6* aggregation kinetics in a dose-dependent manner, consistent with its participation in a hybrid steric zipper model. Single site mutants were generated which transformed predicted amyloidogenic sequences in tau into non-amyloidogenic ones. A M11K mutant had fewer filaments and showed a decrease in aggregation kinetics and an increased lag time compared to wild-type tau, while a F378K mutant showed significantly more filaments. Our results infer that sequences throughout tau, in addition to PHF6 and PHF6*, can seed amyloid formation or affect aggregation kinetics or thermodynamics.
Collapse
Affiliation(s)
- Christopher L Moore
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | | | | | | | | | | | | |
Collapse
|
546
|
Kapurniotu A. Terminierung des Amyloid-Reißverschlusses durch Design. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201105706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
547
|
Iqbal K, Liu F, Gong CX, Grundke-Iqbal I. Tau in Alzheimer disease and related tauopathies. Curr Alzheimer Res 2011; 7:656-64. [PMID: 20678074 DOI: 10.2174/156720510793611592] [Citation(s) in RCA: 699] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 01/20/2010] [Indexed: 01/17/2023]
Abstract
Tau is the major microtubule associated protein (MAP) of a mature neuron. The other two neuronal MAPs are MAP1 and MAP2. An established function of MAPs is their interaction with tubulin and promotion of its assembly into microtubules and stabilization of the microtubule network. The microtubule assembly promoting activity of tau, a phosphoprotein, is regulated by its degree of phosphorylation. Normal adult human brain tau contains 2-3 moles phosphate/mole of tau protein. Hyperphosphorylation of tau depresses this biological activity of tau. In Alzheimer disease (AD) brain tau is ~three to four-fold more hyperphosphorylated than the normal adult brain tau and in this hyperphosphorylated state it is polymerized into paired helical filaments ([PHF) admixed with straight filaments (SF) forming neurofibrillary tangles. Tau is transiently hyperphosphorylated during development and during anesthesia and hypothermia but not to the same state as in AD brain. The abnormally hyperphosphorylated tau in AD brain is distinguished from transiently hyperphosphorylated tau by its ability (1) to sequester normal tau, MAP1 and MAP2 and disrupt microtubules, and (2) to self-assemble into PHF/SF. The cytosolic abnormally hyperphosphorylated tau, because of oligomerization, unlike normal tau, is sedimentable and on self-assembly into PHF/SF, loses its ability to sequester normal MAPs. Some of the tau in AD brain is truncated which also promotes its self-assembly. Tau mutations found in frontotemporal dementia apparently promote its abnormal hyperphosphorylation. Thus, the AD abnormally hyperphosphorylated tau (1) is distinguishable from both normal and transiently hyperphosphorylated taus, and (2) is inhibitory when in a cytosolic/oligomeric state but not when it is self-assembled into PHF/SF. Inhibition of abnormal hyperphosphorylation of tau offers a promising therapeutic target for AD and related tauopathies.
Collapse
Affiliation(s)
- K Iqbal
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, New York 10314-6399, USA.
| | | | | | | |
Collapse
|
548
|
In vivo microdialysis reveals age-dependent decrease of brain interstitial fluid tau levels in P301S human tau transgenic mice. J Neurosci 2011; 31:13110-7. [PMID: 21917794 DOI: 10.1523/jneurosci.2569-11.2011] [Citation(s) in RCA: 275] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although tau is a cytoplasmic protein, it is also found in brain extracellular fluids, e.g., CSF. Recent findings suggest that aggregated tau can be transferred between cells and extracellular tau aggregates might mediate spread of tau pathology. Despite these data, details of whether tau is normally released into the brain interstitial fluid (ISF), its concentration in ISF in relation to CSF, and whether ISF tau is influenced by its aggregation are unknown. To address these issues, we developed a microdialysis technique to analyze monomeric ISF tau levels within the hippocampus of awake, freely moving mice. We detected tau in ISF of wild-type mice, suggesting that tau is released in the absence of neurodegeneration. ISF tau was significantly higher than CSF tau and their concentrations were not significantly correlated. Using P301S human tau transgenic mice (P301S tg mice), we found that ISF tau is fivefold higher than endogenous murine tau, consistent with its elevated levels of expression. However, following the onset of tau aggregation, monomeric ISF tau decreased markedly. Biochemical analysis demonstrated that soluble tau in brain homogenates decreased along with the deposition of insoluble tau. Tau fibrils injected into the hippocampus decreased ISF tau, suggesting that extracellular tau is in equilibrium with extracellular or intracellular tau aggregates. This technique should facilitate further studies of tau secretion, spread of tau pathology, the effects of different disease states on ISF tau, and the efficacy of experimental treatments.
Collapse
|
549
|
Sibille N, Huvent I, Fauquant C, Verdegem D, Amniai L, Leroy A, Wieruszeski JM, Lippens G, Landrieu I. Structural characterization by nuclear magnetic resonance of the impact of phosphorylation in the proline-rich region of the disordered Tau protein. Proteins 2011; 80:454-62. [PMID: 22072628 DOI: 10.1002/prot.23210] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 11/10/2022]
Abstract
Phosphorylation of the neuronal Tau protein is implicated in both the regulation of its physiological function of microtubule stabilization and its pathological propensity to aggregate into the fibers that characterize Alzheimer's diseased neurons. However, how specific phosphorylation events influence both aspects of Tau biology remains largely unknown. In this study, we address the structural impact of phosphorylation of the Tau protein by Nuclear Magnetic Resonance (NMR) spectroscopy on a functional fragment of Tau (Tau[Ser208-Ser324] = TauF4). TauF4 was phosphorylated by the proline-directed CDK2/CycA3 kinase on Thr231 (generating the AT180 epitope), Ser235, and equally on Thr212 and Thr217 in the Proline-rich region (Tau[Ser208-Gln244] or PRR). These modifications strongly decrease the capacity of TauF4 to polymerize tubulin into microtubules. While all the NMR parameters are consistent with a globally disordered Tau protein fragment, local clusters of structuration can be defined. The most salient result of our NMR analysis is that phosphorylation in the PRR stabilizes a short α-helix that runs from pSer235 till the very beginning of the microtubule-binding region (Tau[Thr245-Ser324] or MTBR of TauF4). Phosphorylation of Thr231/Ser235 creates a N-cap with helix stabilizing role while phosphorylation of Thr212/Thr217 does not induce modification of the local transient secondary structure, showing that the stabilizing effect is sequence specific. Using paramagnetic relaxation experiments, we additionally show a transient interaction between the PRR and the MTBR, observed in both TauF4 and phospho-TauF4.
Collapse
Affiliation(s)
- Nathalie Sibille
- CNRS-UMR 8576 UGSF-IFR 147, Université des Sciences et Technologies de Lille 1, 59655 Villeneuve d'Ascq Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
550
|
Patterson KR, Ward SM, Combs B, Voss K, Kanaan NM, Morfini G, Brady ST, Gamblin TC, Binder LI. Heat shock protein 70 prevents both tau aggregation and the inhibitory effects of preexisting tau aggregates on fast axonal transport. Biochemistry 2011; 50:10300-10. [PMID: 22039833 DOI: 10.1021/bi2009147] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aggregation and accumulation of the microtubule-associated protein tau are associated with cognitive decline and neuronal degeneration in Alzheimer's disease and other tauopathies. Thus, preventing the transition of tau from a soluble state to insoluble aggregates and/or reversing the toxicity of existing aggregates would represent a reasonable therapeutic strategy for treating these neurodegenerative diseases. Here we demonstrate that molecular chaperones of the heat shock protein 70 (Hsp70) family are potent inhibitors of tau aggregation in vitro, preventing the formation of both mature fibrils and oligomeric intermediates. Remarkably, addition of Hsp70 to a mixture of oligomeric and fibrillar tau aggregates prevents the toxic effect of these tau species on fast axonal transport, a critical process for neuronal function. When incubated with preformed tau aggregates, Hsp70 preferentially associated with oligomeric over fibrillar tau, suggesting that prefibrillar oligomeric tau aggregates play a prominent role in tau toxicity. Taken together, our data provide a novel molecular basis for the protective effect of Hsp70 in tauopathies.
Collapse
Affiliation(s)
- Kristina R Patterson
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|