Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 41
1.
Mol Neurodegener ; 17(1): 54, 2022 08 20.
Article En | MEDLINE | ID: mdl-35987691

BACKGROUND: Amyloid plaque deposition and axonal degeneration are early events in AD pathogenesis. Aß disrupts microtubules in presynaptic dystrophic neurites, resulting in the accumulation of impaired endolysosomal and autophagic organelles transporting ß-site amyloid precursor protein cleaving enzyme (BACE1). Consequently, dystrophic neurites generate Aß42 and significantly contribute to plaque deposition. Farnesyltransferase inhibitors (FTIs) have recently been investigated for repositioning toward the treatment of neurodegenerative disorders and block the action of farnesyltransferase (FTase) to catalyze farnesylation, a post-translational modification that regulates proteins involved in lysosome function and microtubule stability. In postmortem AD brains, FTase and its downstream signaling are upregulated. However, the impact of FTIs on amyloid pathology and dystrophic neurites is unknown. METHODS: We tested the effects of the FTIs LNK-754 and lonafarnib in the 5XFAD mouse model of amyloid pathology. RESULTS: In 2-month-old 5XFAD mice treated chronically for 3 months, LNK-754 reduced amyloid plaque burden, tau hyperphosphorylation, and attenuated the accumulation of BACE1 and LAMP1 in dystrophic neurites. In 5-month-old 5XFAD mice treated acutely for 3 weeks, LNK-754 reduced dystrophic neurite size and LysoTracker-Green accumulation in the absence of effects on Aß deposits. Acute treatment with LNK-754 improved memory and learning deficits in hAPP/PS1 amyloid mice. In contrast to LNK-754, lonafarnib treatment was less effective at reducing plaques, tau hyperphosphorylation and dystrophic neurites, which could have resulted from reduced potency against FTase compared to LNK-754. We investigated the effects of FTIs on axonal trafficking of endolysosomal organelles and found that lonafarnib and LNK-754 enhanced retrograde axonal transport in primary neurons, indicating FTIs could support the maturation of axonal late endosomes into lysosomes. Furthermore, FTI treatment increased levels of LAMP1 in mouse primary neurons and in the brains of 5XFAD mice, demonstrating that FTIs stimulated the biogenesis of endolysosomal organelles. CONCLUSIONS: We show new data to suggest that LNK-754 promoted the axonal trafficking and function of endolysosomal compartments, which we hypothesize decreased axonal dystrophy, reduced BACE1 accumulation and inhibited amyloid deposition in 5XFAD mice. Our results agree with previous work identifying FTase as a therapeutic target for treating proteinopathies and could have important therapeutic implications in treating AD.


Amyloid , Amyloidosis , Farnesyltranstransferase , Alzheimer Disease/metabolism , Amyloid/drug effects , Amyloid/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Aspartic Acid Endopeptidases/metabolism , Axons/drug effects , Axons/pathology , Disease Models, Animal , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Mice , Mice, Transgenic , Plaque, Amyloid/pathology
2.
Neuron ; 104(5): 869-884.e11, 2019 12 04.
Article En | MEDLINE | ID: mdl-31648898

Age-related neurodegenerative disorders are characterized by a slow, persistent accumulation of aggregated proteins. Although cells can elicit physiological responses to enhance cellular clearance and counteract accumulation, it is unclear how pathogenic proteins evade this process in disease. We find that Parkinson's disease α-synuclein perturbs the physiological response to lysosomal stress by impeding the SNARE protein ykt6. Cytosolic ykt6 is normally autoinhibited by a unique farnesyl-mediated regulatory mechanism; however, during lysosomal stress, it activates and redistributes into membranes to preferentially promote hydrolase trafficking and enhance cellular clearance. α-Synuclein aberrantly binds and deactivates ykt6 in patient-derived neurons, thereby disabling the lysosomal stress response and facilitating protein accumulation. Activating ykt6 by small-molecule farnesyltransferase inhibitors restores lysosomal activity and reduces α-synuclein in patient-derived neurons and mice. Our findings indicate that α-synuclein creates a permissive environment for aggregate persistence by inhibiting regulated cellular clearance and provide a therapeutic strategy to restore protein homeostasis by harnessing SNARE activity.


Lysosomes/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , R-SNARE Proteins/metabolism , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Transport/physiology , Stress, Physiological/physiology
3.
J Neurochem ; 119(2): 389-97, 2011 Oct.
Article En | MEDLINE | ID: mdl-21848810

α-Synuclein causes Parkinson's disease if mutated or aberrantly produced in neurons. α-Synuclein-lipid interactions are important for the normal function of the protein, but can also contribute to pathogenesis. We previously reported that deletion of the first 10 N-terminal amino acids dramatically reduced lipid binding in vitro, as well as membrane binding and toxicity in yeast. Here we extend this study to human neuroblastoma SHSY-5Y cells, and find that in these cells the first 10 N-terminal residues do not affect α-synuclein membrane binding, self-association and cell viability, contrary to yeast. Differences in lipid composition, membrane fluidity and cytosolic factors between yeast and neuronal cells may account for the distinct binding behavior of the truncated variant in these two systems. Retinoic acid promotes differentiation and α-synuclein oligomer formation in neuroblastoma cells, while addition of a proteasomal inhibitor induces neurite outgrowth and toxicity to certain wild-type and truncated α-synuclein clones. Yeast recapitulate several features of α-synuclein (patho)biology, but its simplicity sets limitations; verification of yeast results in more relevant model systems is, therefore, essential.


Neuroblastoma/metabolism , Saccharomyces cerevisiae/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Blotting, Western , Cell Cycle/physiology , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival/physiology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , Microscopy, Fluorescence , Saccharomyces cerevisiae/drug effects , Transfection , Tretinoin/pharmacology , alpha-Synuclein/toxicity
4.
J Med Chem ; 53(7): 2709-18, 2010 Apr 08.
Article En | MEDLINE | ID: mdl-20232802

We recently described a set of drug-like molecules obtained from an in silico screen that stabilize mutant superoxide dismutase-1 (SOD-1) linked to familial amyotrophic lateral sclerosis (ALS) against unfolding and aggregation but exhibited poor binding specificity toward SOD-1 in presence of blood plasma. A reasonable but not a conclusive model for the binding of these molecules was proposed on the basis of restricted docking calculations and site-directed mutagenesis of key residues at the dimer interface. A set of hydrogen bonding constraints obtained from these experiments were used to guide docking calculations with compound library around the dimer interface. A series of chemically unrelated hits were predicted, which were experimentally tested for their ability to block aggregation. At least six of the new molecules exhibited high specificity of binding toward SOD-1 in the presence of blood plasma. These molecules represent a new class of molecules for further development into clinical candidates.


Amyotrophic Lateral Sclerosis/enzymology , Computational Biology , Mutant Proteins/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Superoxide Dismutase/metabolism , Absorption , Binding Sites , Buffers , DNA Mutational Analysis , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Ligands , Models, Molecular , Molecular Conformation , Mutagenesis, Site-Directed , Mutant Proteins/blood , Mutant Proteins/chemistry , Mutant Proteins/genetics , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Substrate Specificity , Superoxide Dismutase/blood , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Uracil/analogs & derivatives , Uracil/metabolism
5.
Proc Natl Acad Sci U S A ; 106(12): 4635-40, 2009 Mar 24.
Article En | MEDLINE | ID: mdl-19261853

Ubiquitin C-terminal hydrolase-L1 (UCH-L1) is linked to Parkinson's disease (PD) and memory and is selectively expressed in neurons at high levels. Its expression pattern suggests a function distinct from that of its widely expressed homolog UCH-L3. We report here that, in contrast to UCH-L3, UCH-L1 exists in a membrane-associated form (UCH-L1(M)) in addition to the commonly studied soluble form. C-terminal farnesylation promotes the association of UCH-L1 with cellular membranes, including the endoplasmic reticulum. The amount of UCH-L1(M) in transfected cells is shown to correlate with the intracellular level of alpha-synuclein, a protein whose accumulation is associated with neurotoxicity and the development of PD. Reduction of UCH-L1(M) in cell culture models of alpha-synuclein toxicity by treatment with a farnesyltransferase inhibitor (FTI-277) reduces alpha-synuclein levels and increases cell viability. Proteasome function is not affected by UCH-L1(M), suggesting that it may negatively regulate the lysosomal degradation of alpha-synuclein. Therefore, inhibition of UCH-L1 farnesylation may be a therapeutic strategy for slowing the progression of PD and related synucleinopathies.


Cell Membrane/enzymology , Neurons/drug effects , Parkinson Disease/enzymology , Parkinson Disease/therapy , Prenylation/drug effects , Ubiquitin Thiolesterase/metabolism , alpha-Synuclein/toxicity , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , COS Cells , Cell Line, Tumor , Cell Membrane/drug effects , Chlorocebus aethiops , Cysteine/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/enzymology , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Neurons/metabolism , Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects
6.
J Mol Biol ; 389(2): 413-24, 2009 Jun 05.
Article En | MEDLINE | ID: mdl-19285989

Alpha-synuclein (alpha-syn), a protein implicated in Parkinson's disease, is structurally diverse. In addition to its random-coil state, alpha-syn can adopt an alpha-helical structure upon lipid membrane binding or a beta-sheet structure upon aggregation. We used yeast biology and in vitro biochemistry to detect how sequence changes alter the structural propensity of alpha-syn. The N-terminus of the protein, which adopts an alpha-helical conformation upon lipid binding, is essential for membrane binding in yeast, and variants that are more prone to forming an alpha-helical structure in vitro are generally more toxic to yeast. beta-Sheet structure and inclusion formation, on the other hand, appear to be protective, possibly by sequestering the protein from the membrane. Surprisingly, sequential deletion of residues 2 through 11 caused a dramatic drop in alpha-helical propensity, vesicle binding in vitro, and membrane binding and toxicity in yeast, part of which could be mimicked by mutating aspartic acid at position 2 to alanine. Variants with distinct structural preferences, identified here by a reductionist approach, provide valuable tools for elucidating the nature of toxic forms of alpha-syn in neurons.


Cell Membrane/metabolism , Yeasts/chemistry , alpha-Synuclein/chemistry , Amino Acid Sequence , Amino Acid Substitution , Fungal Proteins , Membrane Proteins , Protein Binding , Protein Structure, Secondary , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
7.
J Biol Chem ; 283(24): 16895-905, 2008 Jun 13.
Article En | MEDLINE | ID: mdl-18343814

alpha-Synuclein (alpha-syn) phosphorylation at serine 129 is characteristic of Parkinson disease (PD) and related alpha-synulceinopathies. However, whether phosphorylation promotes or inhibits alpha-syn aggregation and neurotoxicity in vivo remains unknown. This understanding is critical for elucidating the role of alpha-syn in the pathogenesis of PD and for development of therapeutic strategies for PD. To better understand the structural and molecular consequences of Ser-129 phosphorylation, we compared the biochemical, structural, and membrane binding properties of wild type alpha-syn to those of the phosphorylation mimics (S129E, S129D) as well as of in vitro phosphorylated alpha-syn using a battery of biophysical techniques. Our results demonstrate that phosphorylation at Ser-129 increases the conformational flexibility of alpha-syn and inhibits its fibrillogenesis in vitro but does not perturb its membrane-bound conformation. In addition, we show that the phosphorylation mimics (S129E/D) do not reproduce the effect of phosphorylation on the structural and aggregation properties of alpha-syn in vitro. Our findings have significant implications for current strategies to elucidate the role of phosphorylation in modulating protein structure and function in health and disease and provide novel insight into the underlying mechanisms that govern alpha-syn aggregation and toxicity in PD and related alpha-synulceinopathies.


Serine/chemistry , alpha-Synuclein/metabolism , Chromatography, Gel , Circular Dichroism , Humans , Liposomes/metabolism , Magnetic Resonance Spectroscopy , Micelles , Models, Biological , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Conformation , Recombinant Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
J Clin Invest ; 118(2): 777-88, 2008 Feb.
Article En | MEDLINE | ID: mdl-18172548

Altered degradation of alpha-synuclein (alpha-syn) has been implicated in the pathogenesis of Parkinson disease (PD). We have shown that alpha-syn can be degraded via chaperone-mediated autophagy (CMA), a selective lysosomal mechanism for degradation of cytosolic proteins. Pathogenic mutants of alpha-syn block lysosomal translocation, impairing their own degradation along with that of other CMA substrates. While pathogenic alpha-syn mutations are rare, alpha-syn undergoes posttranslational modifications, which may underlie its accumulation in cytosolic aggregates in most forms of PD. Using mouse ventral medial neuron cultures, SH-SY5Y cells in culture, and isolated mouse lysosomes, we have found that most of these posttranslational modifications of alpha-syn impair degradation of this protein by CMA but do not affect degradation of other substrates. Dopamine-modified alpha-syn, however, is not only poorly degraded by CMA but also blocks degradation of other substrates by this pathway. As blockage of CMA increases cellular vulnerability to stressors, we propose that dopamine-induced autophagic inhibition could explain the selective degeneration of PD dopaminergic neurons.


Autophagy/genetics , Dopamine/metabolism , Molecular Chaperones/metabolism , Parkinson Disease/etiology , alpha-Synuclein/metabolism , Animals , Lysosomes/metabolism , Male , Mice , Mice, Mutant Strains , Parkinson Disease/pathology , Phosphorylation , Protein Processing, Post-Translational , Rats , Rats, Wistar , alpha-Synuclein/genetics
9.
Biochemistry ; 46(24): 7107-18, 2007 Jun 19.
Article En | MEDLINE | ID: mdl-17530780

The third and most recently identified Parkinson's disease-linked variant of the neuronal protein alpha-synuclein to be identified (E46K) results in widespread brain pathology and early onset Parkinson symptoms (Zarranz et al. (2004) Ann. Neurol. 55, 164-173). Herein, we present biochemical and biophysical characterization of E46K alpha-synuclein in various states of aggregation. Circular dichroism and nuclear magnetic resonance spectroscopy illustrate that the E46K mutation results in subtle changes in the conformation of the monomeric protein both free in solution and in the presence of SDS micelles. However, it does not alter the overall helical propensity of the protein in the presence of phospholipids. E46K alpha-synuclein formed insoluble fibrils in vitro more rapidly than the wild type protein, and electron microscopy revealed that E46K alpha-synuclein fibrils possess a typical amyloid ultrastructure. E46K alpha-synuclein protofibrils, soluble aggregates that form during the transition from the monomeric form to the fibrillar form of alpha-synuclein, were characterized by electron microscopy and gel filtration and were found to include annular species. The unique ability of a subfraction of E46K and wild type alpha-synuclein protofibrils containing porelike species to permeabilize lipid vesicles was demonstrated in vitro using a real-time chromatographic method. In contrast to simplistic expectations, the total amount of protofibrils and the amount of permeabilizing activity per mole protein in the protofibril fraction were reduced by the E46K mutation. These results suggest that if the porelike activity of alpha-synuclein is important for neurotoxicity, there must be factors in the neuronal cytoplasm that reverse the trends in the intrinsic properties of E46K versus WT alpha-synuclein that are observed in vitro.


Point Mutation , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , Amino Acid Substitution , Circular Dichroism , Humans , In Vitro Techniques , Lipids/chemistry , Micelles , Microscopy, Electron , Models, Neurological , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Parkinson Disease/etiology , Parkinson Disease/genetics , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , alpha-Synuclein/ultrastructure
10.
J Biol Chem ; 282(14): 10567-75, 2007 Apr 06.
Article En | MEDLINE | ID: mdl-17259170

Deubiquitinating enzymes (DUBs) are negative regulators of protein ubiquitination and play an important role in ubiquitin-dependent processes. Recent studies have found that diverse cellular mechanisms are employed to control the activity of DUBs. Ubiquitin C-terminal hydrolase-L1 (UCH-L1) is a highly expressed neuronal DUB linked to Parkinson disease; however, little is known about its specific functions or modes of regulation. Here, we demonstrate that UCH-L1 is post-translationally modified by monoubiquitin in cells, at lysine residues near the active site. This modification restricts enzyme activity by preventing binding to ubiquitinated targets, and permanent monoubiquitination, as mimicked by a ubiquitin-UCH-L1 fusion, inhibits UCH-L1 in its capacity to increase free ubiquitin levels in cells. Interestingly, UCH-L1 catalyzes its own deubiquitination in an intramolecular manner, thereby regulating the lifetime of this modification. Our results illustrate monoubiquitination as a reversible regulatory mechanism for DUB activity involving auto-deubiquitination.


Parkinson Disease/enzymology , Protein Processing, Post-Translational/physiology , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cysteine Endopeptidases/metabolism , Half-Life , Humans
11.
J Mol Biol ; 366(5): 1510-22, 2007 Mar 09.
Article En | MEDLINE | ID: mdl-17222866

To investigate the alpha-synuclein protein and its role in Parkinson's disease, we screened a library of random point mutants both in vitro and in yeast to find variants in an unbiased way that could help us understand the sequence-phenotype relationship. We developed a rapid purification method that allowed us to screen 59 synuclein mutants in vitro and discovered two double-point mutants that fibrillized slowly relative to wild-type, A30P, and A53T alpha-synucleins. The yeast toxicity of all of these proteins was measured, and we found no correlation with fibrillization rate, suggesting that fibrillization is not necessary for synuclein-induced yeast toxicity. We found that beta-synuclein was of intermediate toxicity to yeast, and gamma-synuclein was non-toxic. Co-expression of Parkinson's disease-related genes DJ-1, parkin, Pink1, UCH-L1, or synphilin, with synuclein, did not affect synuclein toxicity. A second screen, of several thousand library clones in yeast, identified 25 non-toxic alpha-synuclein sequence variants. Most of these contained a mutation to either proline or glutamic acid that caused a defect in membrane binding. We hypothesize that yeast toxicity is caused by synuclein binding directly to membranes at levels sufficient to non-specifically disrupt homeostasis.


Cell Membrane/metabolism , Nerve Tissue Proteins/metabolism , Saccharomyces cerevisiae/physiology , alpha-Synuclein/chemistry , alpha-Synuclein/toxicity , Amino Acid Sequence , Electroporation , Escherichia coli/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Molecular Sequence Data , Mutagenesis , Nerve Tissue Proteins/genetics , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Point Mutation , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/growth & development , Sequence Homology, Amino Acid , Transformation, Genetic , Two-Hybrid System Techniques , alpha-Synuclein/genetics , alpha-Synuclein/isolation & purification , alpha-Synuclein/metabolism
12.
Biochemistry ; 45(49): 14717-25, 2006 Dec 12.
Article En | MEDLINE | ID: mdl-17144664

Deubiquitinating enzymes regulate essential cellular processes, and their dysregulation is implicated in multiple disease states. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) has garnered attention for its links with Parkinson's disease and cancer; however, the mechanism of action of this enzyme in cells remains poorly understood. In order to advance our understanding of UCH-L1 function, we have been developing small molecule modulators of the enzyme for use as tools to probe its role in cells. In support of these efforts, an investigation of the mechanism of UCH-L1 catalysis was previously reported. Here, we extend this mechanistic evaluation and examine substrate recognition by UCH-L1. We developed a panel of ubiquitin fusions to test the contribution of specific residues of ubiquitin to binding and catalysis by the enzyme, and determined the activation parameters of selected variants to gain additional mechanistic insight. Ubiquitin side chains critical for establishing the Michaelis complex and enabling catalysis were identified, and features of this complex that differ between UCH-L1 and a homologue, UCH-L3, were revealed. These data provide dramatic examples of differences in substrate specificity between these enzymes. The implications of our experiments with UCH-L1 for selective inhibitor design and the relationship to disease are discussed.


Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Amino Acid Substitution , Catalysis , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Humans , Kinetics , Models, Molecular , Parkinson Disease/enzymology , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Thermodynamics , Ubiquitin/metabolism
13.
Nature ; 443(7113): 774-9, 2006 Oct 19.
Article En | MEDLINE | ID: mdl-17051203

The correlation between neurodegenerative disease and protein aggregation in the brain has long been recognized, but a causal relationship has not been unequivocally established, in part because a discrete pathogenic aggregate has not been identified. The complexity of these diseases and the dynamic nature of protein aggregation mean that, despite progress towards understanding aggregation, its relationship to disease is difficult to determine in the laboratory. Nevertheless, drug candidates that inhibit aggregation are now being tested in the clinic. These have the potential to slow the progression of Alzheimer's disease, Parkinson's disease and related disorders and could, if administered presymptomatically, drastically reduce the incidence of these diseases. The clinical trials could also settle the century-old debate about causality.


Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid/drug effects , Amyloid/metabolism , Animals , Humans , Models, Biological , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/physiopathology , Protein Denaturation/drug effects
14.
Q Rev Biophys ; 39(2): 167-201, 2006 May.
Article En | MEDLINE | ID: mdl-16978447

Protein fibrillization is implicated in the pathogenesis of most, if not all, age-associated neurodegenerative diseases, but the mechanism(s) by which it triggers neuronal death is unknown. Reductionist in vitro studies suggest that the amyloid protofibril may be the toxic species and that it may amplify itself by inhibiting proteasome-dependent protein degradation. Although its pathogenic target has not been identified, the properties of the protofibril suggest that neurons could be killed by unregulated membrane permeabilization, possibly by a type of protofibril referred to here as the 'amyloid pore'. The purpose of this review is to summarize the existing supportive circumstantial evidence and to stimulate further studies designed to test the validity of this hypothesis.


Aging , Amyloid/physiology , Neurodegenerative Diseases/etiology , Neurons/pathology , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/chemistry , Cell Death , Humans , Lewy Body Disease/etiology , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Proteasome Endopeptidase Complex/physiology
15.
Cell ; 126(4): 655-7, 2006 Aug 25.
Article En | MEDLINE | ID: mdl-16923386

Memory loss is an early symptom of Alzheimer's Disease (AD). The findings of Gong et al. (2006) now indicate that enhancing the activity of UCH-L1, a ubiquitin hydrolase, alleviates the synaptic dysfunction and memory loss associated with a mouse model of AD. This work also raises the question of what role UCH-L1 might play in other diseases involving protein aggregation, such as Parkinson's Disease.


Alzheimer Disease/physiopathology , Disease Models, Animal , Memory Disorders/physiopathology , Synapses/physiology , Ubiquitin Thiolesterase/metabolism , Alzheimer Disease/pathology , Animals , Humans , Mice
16.
Proc Natl Acad Sci U S A ; 103(12): 4675-80, 2006 Mar 21.
Article En | MEDLINE | ID: mdl-16537382

The ubiquitin C-terminal hydrolase UCH-L1 (PGP9.5) comprises >1% of total brain protein but is almost absent from other tissues [Wilkinson, K. D., et al. (1989) Science 246, 670-673]. Mutations in the UCH-L1 gene have been reported to be linked to susceptibility to and protection from Parkinson's disease [Leroy, E., et al. (1998) Nature 395, 451-452; Maraganore, D. M., et al. (1999) Neurology 53, 1858-1860]. Abnormal overexpression of UCH-L1 has been shown to correlate with several forms of cancer [Hibi, K., et al. (1998) Cancer Res. 58, 5690-5694]. Because the amino acid sequence of UCH-L1 is similar to that of other ubiquitin C-terminal hydrolases, including the ubiquitously expressed UCH-L3, which appear to be unconnected to neurodegenerative disease, the structure of UCH-L1 and the effects of disease associated mutations on the structure and function are of considerable importance. We have determined the three-dimensional structure of human UCH-L1 at 2.4-A resolution by x-ray crystallography. The overall fold resembles that of other ubiquitin hydrolases, including UCH-L3, but there are a number of significant differences. In particular, the geometry of the catalytic residues in the active site of UCH-L1 is distorted in such a way that the hydrolytic activity would appear to be impossible without substrate induced conformational rearrangements.


Parkinson Disease/enzymology , Ubiquitin Thiolesterase/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dimerization , Humans , Models, Biological , Molecular Sequence Data , Protein Conformation
18.
Nucleic Acids Res ; 33(11): 3667-77, 2005.
Article En | MEDLINE | ID: mdl-15990391

A computer program for the generation and analysis of in silico random point mutagenesis libraries is described. The program operates by mutagenizing an input nucleic acid sequence according to mutation parameters specified by the user for each sequence position and type of point mutation. The program can mimic almost any type of random mutagenesis library, including those produced via error-prone PCR (ep-PCR), mutator Escherichia coli strains, chemical mutagenesis, and doped or random oligonucleotide synthesis. The program analyzes the generated nucleic acid sequences and/or the associated protein library to produce several estimates of library diversity (number of unique sequences, point mutations, and single point mutants) and the rate of saturation of these diversities during experimental screening or selection of clones. This information allows one to select the optimal screen size for a given mutagenesis library, necessary to efficiently obtain a certain coverage of the sequence-space. The program also reports the abundance of each specific protein mutation at each sequence position, which is useful as a measure of the level and type of mutation bias in the library. Alternatively, one can use the program to evaluate the relative merits of preexisting libraries, or to examine various hypothetical mutation schemes to determine the optimal method for creating a library that serves the screen/selection of interest. Simulated libraries of at least 10(9) sequences are accessible by the numerical algorithm with currently available personal computers; an analytical algorithm is also available which can rapidly calculate a subset of the numerical statistics in libraries of arbitrarily large size. A multi-type double-strand stochastic model of ep-PCR is developed in an appendix to demonstrate the applicability of the algorithm to amplifying mutagenesis procedures. Estimators of DNA polymerase mutation-type-specific error rates are derived using the model. Analyses of an alpha-synuclein ep-PCR library and NNS synthetic oligonucleotide libraries are given as examples.


Computational Biology/methods , Gene Library , Mutagenesis , Sequence Analysis, DNA/methods , Sequence Analysis, Protein/methods , Software , Algorithms , Genetic Variation , Nerve Tissue Proteins/genetics , Oligonucleotides/chemistry , Point Mutation , Polymerase Chain Reaction , Synucleins , alpha-Synuclein
19.
Proc Natl Acad Sci U S A ; 102(10): 3639-44, 2005 Mar 08.
Article En | MEDLINE | ID: mdl-15738401

Familial amyotrophic lateral sclerosis (FALS) is a fatal motor neuron disease that is caused by mutations in the gene encoding superoxide dismutase-type 1 (SOD1). The affected regions of the FALS brain are characterized by aggregated SOD1, and the mutations that destabilize SOD1 appear to promote its aggregation in vitro. Because dissociation of the native SOD1 dimer is required for its in vitro aggregation, we initiated an in silico screening program to find drug-like molecules that would stabilize the SOD1 dimer. A potential binding site for such molecules at the SOD1 dimer interface was identified, and its importance was validated by mutagenesis. About 1.5 million molecules from commercial databases were docked at the dimer interface. Of the 100 molecules with the highest predicted binding affinity, 15 significantly inhibited in vitro aggregation and denaturation of A4V, a FALS-linked variant of SOD1. In the presence of several of these molecules, A4V and other FALS-linked SOD1 mutants such as G93A and G85R behaved similarly to wild-type SOD1, suggesting that these compounds could be leads toward effective therapeutics against FALS.


Amyotrophic Lateral Sclerosis/enzymology , Mutation , Protein Folding , Superoxide Dismutase/chemistry , Superoxide Dismutase/drug effects , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Binding Sites , Dimerization , Humans , Protein Denaturation , Superoxide Dismutase/genetics , Superoxide Dismutase-1
20.
Science ; 305(5688): 1292-5, 2004 Aug 27.
Article En | MEDLINE | ID: mdl-15333840

Aberrant alpha-synuclein degradation is implicated in Parkinson's disease pathogenesis because the protein accumulates in the Lewy inclusion bodies associated with the disease. Little is known, however, about the pathways by which wild-type alpha-synuclein is normally degraded. We found that wild-type alpha-synuclein was selectively translocated into lysosomes for degradation by the chaperone-mediated autophagy pathway. The pathogenic A53T and A30P alpha-synuclein mutants bound to the receptor for this pathway on the lysosomal membrane, but appeared to act as uptake blockers, inhibiting both their own degradation and that of other substrates. These findings may underlie the toxic gain-of-function by the mutants.


Autophagy , Lysosomes/metabolism , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Amino Acid Motifs , Animals , Antigens, CD/metabolism , Cells, Cultured , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/pharmacology , Half-Life , Intracellular Membranes/metabolism , Lysosomal Membrane Proteins , Male , Mice , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/metabolism , PC12 Cells , Protein Binding , Protein Transport , Rats , Rats, Wistar , Synucleins , alpha-Synuclein
...