Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Sci Adv ; 6(30): eaba3916, 2020 07.
Article in English | MEDLINE | ID: mdl-32832664

ABSTRACT

Intrinsically disordered proteins (IDPs) can be degraded in a ubiquitin-independent process by the 20S proteasome. Decline in 20S activity characterizes neurodegenerative diseases. Here, we examine 20S degradation of IDP tau, a protein that aggregates into insoluble deposits in Alzheimer's disease. We show that cleavage of tau by the 20S proteasome is most efficient within the aggregation-prone repeat region of tau and generates both short, aggregation-deficient peptides and two long fragments containing residues 1 to 251 and 1 to 218. Phosphorylation of tau by the non-proline-directed Ca2+/calmodulin-dependent protein kinase II inhibits degradation by the 20S proteasome. Phosphorylation of tau by GSK3ß, a major proline-directed tau kinase, modulates tau degradation kinetics in a residue-specific manner. The study provides detailed insights into the degradation products of tau generated by the 20S proteasome, the residue specificity of degradation, single-residue degradation kinetics, and their regulation by posttranslational modification.

2.
J Control Release ; 285: 96-105, 2018 09 10.
Article in English | MEDLINE | ID: mdl-29964137

ABSTRACT

A 2-dimensional high-throughput screening method is presented to select peptide sequences from large peptide libraries for precision formulation additives, having a high capacity to specifically host a drug of interest and provide tailored drug release properties. The identified sequences are conjugated with poly(ethylene glycol) (PEG) to obtain peptide-PEG conjugates that proved to be valuable as solubilizers for small organic molecule drugs to overcome limitations of poor water-solubility and low bio-availability. The 2D-screening method selects peptide sequences on both (i) high loading capacities and (ii) preferred drug-release capabilities as demonstrated on an experimental Tau-protein aggregation inhibitor/Tau- deaggregator with potentials for an anti-Alzheimer disease drug (BB17). To enable 2D-screening, a one-bead one-compound (OBOC) peptide library was immobilized on a glass slide, allocating individual beads to permanent positions. While the first screening step involved incubation of the supported OBOC library with BB17 to identify beads with high drug binding capacities by fluorescence scanner readouts, the second step reveals release properties of the high capacity binders by incubation with blood plasma protein model solutions. Efficiently peptides with high BB17 capacities and either keeper or medium or fast releaser properties can be identified by direct sequence readouts from the glass slide supported resin beads via matrix-assisted laser desorption/ionization time-of-flight tandem mass spectrometry. Four peptides are synthesized as peptide-PEG solubilizers representing strong, medium, weak releasers and non-binders. Loading capacities reached up to 1:3.4 (mol drug per mol carrier) and release kinetics (fast/medium/slow) are in agreement with the selection process as investigated by fluorescence anisotropy and fluorescence correlation spectroscopy. The ability of BB17/conjugate complexes to inhibit the aggregation of Tau4RDΔK (four repeat Tau ((M)Q244-E372 with deletion of K280), 129 residues) in N2a cells is studied by a Tau-pelleting assay showing the modulation of cellular Tau aggregation. Promising effects such as the reduction of 55% of total Tau load are observed for the strong releaser additive. Studies of in vitro Thioflavin S Tau-aggregation assays show half-maximal inhibitory activities (IC50 values) of BB17/conjugates in the low micro-molar range.


Subject(s)
Drug Carriers/chemistry , Peptides/chemistry , Drug Compounding , Drug Liberation , Humans , Models, Molecular , Peptide Library , Pharmaceutical Preparations/administration & dosage , Polyethylene Glycols/chemistry , Solubility , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
Neurobiol Aging ; 32(12): 2314-5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20097445

ABSTRACT

We have established a novel transgenic rat line carrying human microtubule-associated protein Tau-40 with mutation P301L. hTau-40/P301L transgenic male and female rats were followed up to 2 years of age. The hTau-40/P301L rats expressed human tau mRNA and protein in the limbic cortex and associated white matter, hippocampus and spinal cord. With increasing age, the staining density for phosphorylated tau increased in all these areas. Neither silver stains nor Fluoro-Jade staining indicated the presence of dying neurons, or axonal degeneration, and there was no evidence of increased gliosis or inflammation. However, some neurons did display dendritic abnormalities, and immunoblots revealed the presence of sarcosyl insoluble tau. A large test battery revealed no behavioral abnormalities in these rats, except a mild hyperactivity in the elevated plus maze. In conclusion, this transgenic tau rat may be a useful model for 'pretangle' pathology, although in this study conditions were not sufficient to induce significant neuronal loss or behavioral deficits.


Subject(s)
Brain Chemistry/genetics , Models, Animal , Mutation/genetics , tau Proteins/chemistry , tau Proteins/genetics , Animals , Female , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Limbic System/chemistry , Limbic System/metabolism , Male , Rats , Rats, Transgenic , Spinal Cord/chemistry , Spinal Cord/metabolism
4.
Neurodegener Dis ; 7(1-3): 103-7, 2010.
Article in English | MEDLINE | ID: mdl-20173337

ABSTRACT

We have studied the mechanism of aggregation in an inducible cell model of Tau pathology. When the repeat domain of human Tau (Tau(RD)) carrying the FTDP-17 mutation DeltaK280 is expressed, the cells develop aggregates, as seen by thioflavin S fluorescence, electron microscopy, and sarkosyl extraction methods. By contrast, mutants of Tau(RD) that are unable to generate beta-structure do not aggregate. Enhanced aggregation leads to enhanced toxicity, visible by live cell microscopy and LDH release assay. The aggregation process is initiated by the sequential cleavage of Tau(RD) which yields highly amyloidogenic fragments. This cleavage occurs only with proaggregant Tau(RD), and not with the nonaggregating mutants, indicating that beta-structure makes Tau(RD) vulnerable to both proteolytic degradation and aggregation. Aggregation is reversed by switching off the expression of Tau(RD), by inhibitor compounds, and by certain protease inhibitors. In all cases, the enhanced toxicity is rescued. The clearance of the aggregates involves autophagy, whereas proteasomal degradation plays only a minor role.


Subject(s)
Autophagy/physiology , Tauopathies/physiopathology , tau Proteins/genetics , tau Proteins/metabolism , Animals , Humans , Mutation/genetics , Tauopathies/metabolism , Tauopathies/pathology
5.
FASEB J ; 24(6): 1637-48, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20071654

ABSTRACT

Kinases of the MARK/Par-1 family of S/T protein kinases are regulators of diverse cellular processes in Caenorhabditis elegans, Drosophila, yeast, and mammalian cells. They are involved in nematode embryogenesis, epithelial cell polarization, cell signaling, and neuronal differentiation. MARK phosphorylates microtubule-associated proteins such as tau and is a key regulator of microtubule-based intracellular transport. Hyperphosphorylation of tau causes defects in neuronal transport and may induce abnormal aggregation of tau in Alzheimer disease and other tauopathies. Recent high-resolution structure analysis of MARK fragments covering the kinase domain and accessory regulatory domains has revealed important details regarding the autoregulation of MARK, but their interpretation has remained controversial. Here we focus on the structural aspects of MARK activity and autoregulation. Comparison of the available MARK structures with related kinases of the AMPK family and with new structures of MARK isoforms (MARK2 and 3) reveals unexpected structural similarities between these kinases that may help to resolve the existing controversies.


Subject(s)
Protein Kinases/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Receptor, PAR-1/chemistry , Receptor, PAR-1/physiology , AMP-Activated Protein Kinase Kinases , Animals , Humans
6.
Eur J Neurol ; 16(3): 297-309, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19364361

ABSTRACT

Tauopathies with parkinsonism represent a spectrum of disease entities unified by the pathologic accumulation of hyperphosphorylated tau protein fragments within the central nervous system. These pathologic characteristics suggest shared pathogenetic pathways and possible molecular targets for disease-modifying therapeutic interventions. Natural history studies, for instance, in progressive supranuclear palsy, frontotemporal dementia with parkinsonism linked to chromosome 17, corticobasal degeneration, and Niemann-Pick disease type C as well as in amyotrophic lateral sclerosis/Parkinson-dementia complex permit clinical characterization of the disease phenotypes and are crucial to the development and validation of biological markers for differential diagnostics and disease monitoring, for example, by use of neuroimaging or proteomic approaches. The wide pathologic and clinical spectrum of the tauopathies with parkinsonism is reviewed in this article, and perspectives on future advances in the understanding of the pathogenesis are given, together with potential therapeutic strategies.


Subject(s)
Parkinsonian Disorders/complications , Tauopathies/complications , Animals , Biomarkers , Dementia/complications , Dementia/genetics , Dementia/physiopathology , Drug Design , Geography , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Models, Biological , Mutation , Niemann-Pick Disease, Type C/complications , Niemann-Pick Disease, Type C/diagnosis , Niemann-Pick Disease, Type C/physiopathology , Parkinson Disease, Postencephalitic/complications , Parkinson Disease, Postencephalitic/physiopathology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/therapy , Pick Disease of the Brain/complications , Pick Disease of the Brain/pathology , Protein Serine-Threonine Kinases/genetics , Supranuclear Palsy, Progressive/complications , Supranuclear Palsy, Progressive/diagnosis , Supranuclear Palsy, Progressive/physiopathology , Tauopathies/pathology , Tauopathies/physiopathology , Tauopathies/therapy , tau Proteins/genetics
7.
Curr Alzheimer Res ; 4(4): 397-402, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17908042

ABSTRACT

Cell models of tauopathy were generated in order to study mechanisms of neurodegeneration involving abnormal changes of tau. They are based on neuroblastoma cell lines (N2a) that inducibly express different forms of the repeat domain of tau (tau(RD)), e.g. the 4-repeat domain of tau with the wild-type sequence, the repeat domain with the DeltaK280 mutation ("pro-aggregation mutant"), or the repeat domain with DeltaK280 and two proline point mutations ("anti-aggregation mutant"). The data indicate that the aggregation of tau(RD) is toxic, and that aggregation and toxicity can be prevented by low molecular weight compounds, notably compounds based on the N-phenylamine core. Thus the cell models are suitable for developing aggregation inhibitor drugs.


Subject(s)
Aniline Compounds/metabolism , Tauopathies/drug therapy , Tauopathies/genetics , Animals , Cell Line, Tumor/pathology , Drug Evaluation, Preclinical , Humans , Models, Biological , Mutation , Neuroblastoma/pathology , Protein Structure, Tertiary
8.
Proc Natl Acad Sci U S A ; 104(24): 10252-7, 2007 Jun 12.
Article in English | MEDLINE | ID: mdl-17535890

ABSTRACT

Tau is a highly soluble protein, yet it aggregates abnormally in Alzheimer's disease. Here, we address the question of proteolytic processing of tau and the nucleation of aggregates by tau fragments. We show in neuronal cell models that fragments of the repeat domain of tau containing mutations of FTDP17 (frontotemporal dementia with parkinsonism linked to chromosome 17), produced by endogenous proteases, can induce the aggregation of full-length tau. Fragments are generated by successive cleavages, first N-terminally between K257 and S258, then C-terminally around residues 353-364; conversely, when the N-terminal cleavage is inhibited, no fragmentation and aggregation takes place. The C-terminal truncation and the coaggregation of fragments with full-length tau depends on the propensity for beta-structure. The aggregation is modulated by phosphorylation but does not depend on it. Aggregation but not fragmentation as such is toxic to cells; conversely, toxicity can be prevented by inhibiting either aggregation or proteolysis. The results reveal a novel pathway of abnormal tau aggregation in neuronal cells.


Subject(s)
Protein Processing, Post-Translational , tau Proteins/metabolism , Alzheimer Disease/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line, Tumor , Fluorescent Antibody Technique, Indirect , Humans , Hydrolysis , Mice , Microscopy, Fluorescence , Models, Biological , Models, Chemical , Neuroblastoma/pathology , Neurons/metabolism , Peptide Fragments/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Protein Isoforms , Protein Structure, Secondary , Protein Structure, Tertiary , Solubility , Thrombin/chemistry , tau Proteins/chemistry , tau Proteins/genetics
9.
Neurodegener Dis ; 3(4-5): 197-206, 2006.
Article in English | MEDLINE | ID: mdl-17047358

ABSTRACT

The abnormal aggregation of the microtubule-associated protein tau into paired helical filaments is one the hallmarks of Alzheimer's disease. This aggregation is based in the partial formation of beta-structure. In contrast, the soluble protein shows a mostly random coil structure, as judged by circular dichroism, Fourier transform infrared, X-ray scattering and biochemical assays. Here, we review the basis of the natively unstructured character of tau, as well as recent studies of residual structure and long-range interactions between different domains of the protein. Analysis of the primary structure reveals a very low content of hydrophobic amino acids and a high content of charged residues, both of which tend to counteract a well-folded globular state of proteins. In the case of tau, the low overall hydrophobicity is sufficient to explain the lack of folding. This is in contrast to other proteins which also carry an excess charge at physiological pH. By tryptophan scanning mutagenesis and fluorimetry we found that most of the sequence is solvent exposed. Analysis of the hydrodynamic radii confirms a mostly random coil structure of various tau isoforms and tau domains. The proteins can be further expanded by denaturation with GdHCl which indicates some global folding. This was substantiated by a FRET-based approach where the distances between different domains of tau were determined. The combined data show that tau is mostly disordered and flexible but tends to assume a hairpin-like overall fold which may be important in the transition to a pathological aggregate.


Subject(s)
tau Proteins/chemistry , Animals , Humans , Protein Isoforms , Protein Structure, Secondary , Spectrum Analysis
10.
Neurodegener Dis ; 3(4-5): 207-17, 2006.
Article in English | MEDLINE | ID: mdl-17047359

ABSTRACT

The hyperphosphorylation of tau is an early step in the degeneration of neurons in Alzheimer's disease and other tauopathies. Of particular importance is the phosphorylation of tau in the repeat domain which detaches tau from microtubules. This makes microtubules dynamic for their role in differentiation and neurite outgrowth, and it controls the level of tau on the microtubule surface which keeps the tracks clear for axonal transport. However, the detachment of tau from microtubules can also initiate the reactions that lead to pathological aggregation into neurofibrillary tangles. Phosphorylation of tau in the repeat domain is achieved by the kinase MARK/Par-1, a member of the calcium/calmodulin-dependent protein kinase group of kinases. In this report, we focus on the modes of MARK regulation. MARK contains several domains which offer multiple ways of regulation by posttranslational modification (e.g. phosphorylation), interactions with scaffolding proteins and subcellular targeting (e.g. 14-3-3), and interactions with other proteins. We consider in particular the interactions between MARK and other kinases, notably MARKK/TAO-1 and PAK5. MARKK (a member of the Ste20 family of kinases) activates MARK by phosphorylating it at a critical threonine residue within the activation loop. Activated MARK in turn phosphorylates tau, causes its detachment from microtubules and renders them labile. PAK5 inactivates MARK, not by phosphorylation, but by binding to the catalytic domain. PAK5 contributes to microtubule stability by preventing the MARK-induced phosphorylation of tau; conversely, PAK5 contributes to actin dynamics, presumably through the activation of cofilin, an F-actin severing protein. Thus, MARK and its regulators MARKK and PAK5 appear to mediate the crosstalk between the actin and microtubule cytoskeleton in an antagonistic fashion.


Subject(s)
Cytoskeleton/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor Cross-Talk/physiology , Signal Transduction/physiology , tau Proteins/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Enzyme Activation/physiology , Humans , Microtubules/physiology , Neurofibrillary Tangles/enzymology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Transfection
11.
J Muscle Res Cell Motil ; 27(2): 125-37, 2006.
Article in English | MEDLINE | ID: mdl-16362723

ABSTRACT

Kinesins are a family of microtubule-dependent motor proteins that carry cargoes such as vesicles, organelles, or protein complexes along microtubules. Here we summarize structural studies of the "conventional" motor protein kinesin-1 and its interactions with microtubules, as determined by X-ray crystallography and cryo-electron microscopy. In particular, we consider the docking between the kinesin motor domain and tubulin subunits and summarize the evidence that kinesin binds mainly to beta tubulin with the switch-2 helix close to the intradimer interface between alpha and beta tubulin.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Subunits/metabolism , Tubulin/metabolism , Animals , Crystallography, X-Ray/methods , Humans , Kinesins , Microtubule-Associated Proteins/chemistry , Microtubules/ultrastructure , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , Tubulin/chemistry
12.
Adv Protein Chem ; 71: 299-344, 2005.
Article in English | MEDLINE | ID: mdl-16230115

ABSTRACT

Microtubules are the intracellular tracks for two classes of motor proteins: kinesins and dyneins. During the past few years, the motor domain structures of several kinesins from different organisms have been determined by X-ray crystallography. Compared with kinesins, dyneins are much larger proteins and attempts to crystallize them have failed so far. Structural information about these proteins comes mostly from electron microscopy. In this chapter, we mainly focus on the crystal structures of kinesin motor domains.


Subject(s)
Kinesins/chemistry , Microtubule Proteins/chemistry , Molecular Motor Proteins/chemistry , Animals , Dyneins/chemistry , Humans , Kinesins/classification , Myosins/chemistry , Protein Conformation , Protein Structure, Tertiary
13.
J Mol Biol ; 339(3): 539-53, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15147841

ABSTRACT

Tau is a neuronal, microtubule-associated protein that stabilizes microtubules and promotes neurite outgrowth. Tau is largely unfolded in solution and presumably forms mostly random coil. Because of its hydrophilic nature and flexible structure, tau complexed to microtubules is largely invisible by standard electron microscopy methods. We applied a combination of high-resolution metal-shadowing and cryo-electron microscopy to study the interactions between tau and microtubules. We used recombinant tau variants with different domain compositions, (1) full length tau, (2) the repeat domain that mediates microtubule binding (K19), and (3) two GFP-tau fusion proteins that contain a globular marker (GFP) attached to full-length tau at either end. All of these constructs bind exclusively to the outside of microtubules. Most of the tau-related mass appears randomly distributed, creating a "halo" of low-density mass spread across the microtubule surface. Only a small fraction of tau creates a periodic signal at an 8 nm interval, centered on alpha-tubulin subunits. Our data suggest that tau retains most of its disordered structure even when bound to the microtubule surface. Hence, it binds along, as well as across protofilaments. Nevertheless, even minute concentrations of tau have a strong stabilizing effect and effectively scavenge unpolymerized tubulin.


Subject(s)
Microtubules/chemistry , tau Proteins/chemistry , Cryoelectron Microscopy , Humans , Kinesins/chemistry , Microtubules/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/ultrastructure , Tubulin/chemistry
14.
Neurobiol Aging ; 24(8): 1079-85, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14643379

ABSTRACT

Loss of synapses and dying back of axons are considered early events in brain degeneration during Alzheimer's disease. This is accompanied by an aberrant behavior of the microtubule-associated protein tau (hyperphosphorylation, aggregation). Since microtubules are the tracks for axonal transport, we are testing the hypothesis that tau plays a role in the malfunctioning of transport. Experiments with various neuronal and non-neuronal cells show that tau is capable of reducing net anterograde transport of vesicles and cell organelles by blocking the microtubule tracks. Thus, a misregulation of tau could cause the starvation of synapses and enhanced oxidative stress, long before tau detaches from microtubules and aggregates into Alzheimer neurofibrillary tangles. In particular, the transport of amyloid precursor protein is retarded when tau is elevated, suggesting a possible link between the two key proteins that show abnormal behavior in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Axonal Transport/physiology , Brain/metabolism , Synapses/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/pathology , Brain/physiopathology , Cells, Cultured , Mice , Microtubules/metabolism , Microtubules/pathology , Models, Neurological , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Organelles/metabolism , Oxidative Stress/physiology , Synapses/pathology , Transport Vesicles/metabolism
15.
J Neural Transm (Vienna) ; 110(10): 1149-60, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14523627

ABSTRACT

This study evaluated the sensitivity and specificity of the cerebrospinal fluid (CSF) levels of tau-protein, amyloid-beta-peptide 1-42 (Abeta1-42), ApoE-genotype and the degree of cognitive decline as diagnostic markers for Alzheimer's disease (AD). Data was obtained from 105 AD patients and 68 controls. Median CSF-tau levels were increased (512 pg/ml vs. 145 pg/ml, p<0.001) and Abeta1-42-levels were decreased (238.5 pg/ml vs. 310 pg/ml, p<0.001) in AD patients compared to controls. A weak correlation was found between CSF-Abeta1-42 and MMSE score (r=.245). Within all subjects, a correlation of CSF-Abeta1-42 (r=-.337) and CSF-tau (r=.384) with age was found. The combination of CSF-tau levels and MMSE revealed the highest sensitivity (92%) and specificity (87%). In summary, CSF-tau was a useful biological marker to discriminate AD from normal aging, neurological and psychiatric disorders. CSF-Abeta1-42 showed no additional benefit in discriminating patients from controls but might be useful for tracking the severity of the disease.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoproteins E/genetics , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Adolescent , Adult , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Biomarkers/cerebrospinal fluid , Case-Control Studies , Cognition Disorders/diagnosis , Diagnosis, Differential , Female , Genotype , Humans , Male , Middle Aged , Psychiatric Status Rating Scales
16.
Biochemistry ; 41(50): 14885-96, 2002 Dec 17.
Article in English | MEDLINE | ID: mdl-12475237

ABSTRACT

Alzheimer's disease is characterized by aggregates of tau protein. Attempts to study the conditions for aggregation in vitro have led to different experimental systems, some of which appear mutually exclusive (e.g., oxidative vs reductive conditions, induction by polyanions vs fatty acids). We show here that different approaches and pathways can be viewed in a common framework, and that apparent differences can be explained by variations in the kinetics of subreactions. A unified view of PHF aggregation should help to analyze the causes of PHF aggregation and devise methods to prevent it.


Subject(s)
Alzheimer Disease/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Alzheimer Disease/pathology , Anions/chemistry , Arachidonic Acid/chemistry , Cross-Linking Reagents/chemistry , Cysteine/chemistry , Cysteine/genetics , Disulfides/chemistry , Heparin/chemistry , Humans , Mutagenesis, Site-Directed , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/ultrastructure , Polyelectrolytes , Polymers/chemistry , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/ultrastructure , Protein Processing, Post-Translational/genetics , Spectrometry, Fluorescence , tau Proteins/genetics , tau Proteins/ultrastructure
17.
J Cell Biol ; 156(6): 1051-63, 2002 Mar 18.
Article in English | MEDLINE | ID: mdl-11901170

ABSTRACT

We studied the effect of microtubule-associated tau protein on trafficking of vesicles and organelles in primary cortical neurons, retinal ganglion cells, and neuroblastoma cells. Tau inhibits kinesin-dependent transport of peroxisomes, neurofilaments, and Golgi-derived vesicles into neurites. Loss of peroxisomes makes cells vulnerable to oxidative stress and leads to degeneration. In particular, tau inhibits transport of amyloid precursor protein (APP) into axons and dendrites, causing its accumulation in the cell body. APP tagged with yellow fluorescent protein and transfected by adenovirus associates with vesicles moving rapidly forward in the axon (approximately 80%) and slowly back (approximately 20%). Both movements are strongly inhibited by cotransfection with fluorescently tagged tau (cyan fluorescent protein-tau) as seen by two-color confocal microscopy. The data suggests a linkage between tau and APP trafficking, which may be significant in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Neurites/metabolism , Neurofilament Proteins/metabolism , Organelles/metabolism , Oxidative Stress/genetics , Protein Transport/genetics , Transport Vesicles/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cell Survival/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Chick Embryo , Fluorescent Antibody Technique , Gene Expression Regulation/physiology , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Neurites/pathology , Neurofilament Proteins/genetics , Organelles/genetics , Organelles/pathology , Peroxisomes/genetics , Peroxisomes/metabolism , Rats , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Time Factors , Transport Vesicles/genetics , Tumor Cells, Cultured , tau Proteins/genetics
18.
EMBO J ; 20(22): 6213-25, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11707393

ABSTRACT

We determined the crystal structure of the motor domain of the fast fungal kinesin from Neurospora crassa (NcKin). The structure has several unique features. (i) Loop 11 in the switch 2 region is ordered and enables one to describe the complete nucleotide-binding pocket, including three inter-switch salt bridges between switch 1 and 2. (ii) Loop 9 in the switch 1 region bends outwards, making the nucleotide-binding pocket very wide. The displacement in switch 1 resembles that of the G-protein ras complexed with its guanosine nucleotide exchange factor. (iii) Loop 5 in the entrance to the nucleotide-binding pocket is remarkably long and interacts with the ribose of ATP. (iv) The linker and neck region is not well defined, indicating that it is mobile. (v) Image reconstructions of ice-embedded microtubules decorated with NcKin show that it interacts with several tubulin subunits, including a central beta-tubulin monomer and the two flanking alpha-tubulin monomers within the microtubule protofilament. Comparison of NcKin with other kinesins, myosin and G-proteins suggests that the rate-limiting step of ADP release is accelerated in the fungal kinesin and accounts for the unusually high velocity and ATPase activity.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Dimaprit/analogs & derivatives , Kinesins/chemistry , Microtubules/metabolism , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Dimaprit/metabolism , GTP-Binding Proteins/chemistry , Image Processing, Computer-Assisted , Kinetics , Models, Molecular , Myosins/chemistry , Neurospora crassa/chemistry , Nucleotides/metabolism , Protein Binding , Protein Structure, Tertiary , Ribose/chemistry , Salts/chemistry , Tubulin/chemistry
19.
J Mol Biol ; 312(5): 1011-26, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11580246

ABSTRACT

We describe a theoretical and experimental analysis of the interaction between microtubules and dimeric motor proteins (kinesin, NCD), with special emphasis on the stoichiometry of the interaction, cooperative effects, and their consequences for the interpretation of biochemical and image reconstruction results. Monomeric motors can bind equivalently to microtubules without interference, at a stoichiometry of one motor head per tubulin subunit (alphabeta-heterodimer). By contrast, dimeric motors can interact with stoichiometries ranging between one and two heads per tubulin subunit, depending on binding constants of the first head and the subsequent binding of the second head, and the concentration of dimers in solution. Further, we show that an attractive interaction between the bound motor molecules can explain the higher periodicities observed in decorated microtubules (e.g. 16 nm periodicity), and the non-uniform decoration of a population of microtubules and give an estimate of the strength of this interaction.


Subject(s)
Kinesins/chemistry , Kinesins/metabolism , Microtubules/chemistry , Microtubules/metabolism , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/metabolism , Allosteric Site , Animals , Computer Simulation , Decapodiformes , Dimerization , Kinesins/ultrastructure , Kinetics , Microscopy, Electron , Microtubules/ultrastructure , Molecular Motor Proteins/ultrastructure , Protein Binding , Protein Structure, Quaternary , Protein Subunits , Thermodynamics , Tubulin/chemistry , Tubulin/metabolism
20.
J Biol Chem ; 276(51): 48165-74, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11606569

ABSTRACT

The microtubule-associated protein tau is a natively unfolded protein in solution, yet it is able to polymerize into the ordered paired helical filaments (PHF) of Alzheimer's disease. In the splice isoforms lacking exon 10, this process is facilitated by the formation of beta-structure around the hexapeptide motif PHF6 ((306)VQIVYK(311)) encoded by exon 11. We have investigated the structural requirements for PHF polymerization in the context of adult tau isoforms containing four repeats (including exon 10). In addition to the PHF6 motif there exists a related PHF6* motif ((275)VQIINK(280)) in the repeat encoded by the alternatively spliced exon 10. We show that this PHF6* motif also promotes aggregation by the formation of beta-structure and that there is a cross-talk between the two hexapeptide motifs during PHF aggregation. We also show that two of the tau mutations found in hereditary frontotemporal dementias, DeltaK280 and P301L, have a much stronger tendency for PHF aggregation which correlates with their high propensity for beta-structure around the hexapeptide motifs.


Subject(s)
Dementia/metabolism , Mutation , tau Proteins/genetics , Amino Acid Motifs , Amino Acid Sequence , Biopolymers , Dementia/pathology , Microscopy, Electron , Molecular Sequence Data , Oligopeptides/chemistry , Protein Conformation , Spectroscopy, Fourier Transform Infrared , tau Proteins/chemistry , tau Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...