Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
J Mol Biol ; 436(12): 168607, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38734203

ABSTRACT

Polyglutamine (polyQ) sequences undergo repeat-length dependent formation of disease-associated, amyloid-like cross-ß core structures with kinetics and aggregate morphologies often influenced by the flanking sequences. In Huntington's disease (HD), the httNT segment on the polyQ's N-terminal flank enhances aggregation rates by changing amyloid nucleation from a classical homogeneous mechanism to a two-step process requiring an ɑ-helix-rich oligomeric intermediate. A folded, helix-rich httNT tetrameric structure suggested to be this critical intermediate was recently reported. Here we employ single alanine replacements along the httNT sequence to assess this proposed structure and refine the mechanistic model. We find that Ala replacement of hydrophobic residues within simple httNT peptides greatly suppresses helicity, supporting the tetramer model. These same helix-disruptive replacements in the httNT segment of an exon-1 analog greatly reduce aggregation kinetics, suggesting that an ɑ-helix rich multimer - either the tetramer or a larger multimer - plays an on-pathway role in nucleation. Surprisingly, several other Ala replacements actually enhance helicity and/or amyloid aggregation. The spatial localization of these residues on the tetramer surface suggests a self-association interface responsible for formation of the octomers and higher-order multimers most likely required for polyQ amyloid nucleation. Multimer docking of the tetramer, using the protein-protein docking algorithm ClusPro, predicts this symmetric surface to be a viable tetramer dimerization interface. Intriguingly, octomer formation brings the emerging polyQ chains into closer proximity at this tetramer-tetramer interface. Further supporting the potential importance of tetramer super-assembly, computational docking with a known exon-1 aggregation inhibitor predicts ligand contacts with residues at this interface.


Subject(s)
Amyloid , Exons , Huntingtin Protein , Peptides , Protein Multimerization , Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Huntingtin Protein/genetics , Humans , Amyloid/chemistry , Amyloid/metabolism , Peptides/chemistry , Peptides/metabolism , Huntington Disease/metabolism , Huntington Disease/genetics , Kinetics , Hydrophobic and Hydrophilic Interactions , Protein Aggregates , Models, Molecular
2.
Acta Neuropathol Commun ; 9(1): 70, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33853668

ABSTRACT

Huntington's disease (HD) is a late onset, inherited neurodegenerative disorder for which early pathogenic events remain poorly understood. Here we show that mutant exon 1 HTT proteins are recruited to a subset of cytoplasmic aggregates in the cell bodies of neurons in brain sections from presymptomatic HD, but not wild-type, mice. This occurred in a disease stage and polyglutamine-length dependent manner. We successfully adapted a high-resolution correlative light and electron microscopy methodology, originally developed for mammalian and yeast cells, to allow us to correlate light microscopy and electron microscopy images on the same brain section within an accuracy of 100 nm. Using this approach, we identified these recruitment sites as single membrane bound, vesicle-rich endolysosomal organelles, specifically as (1) multivesicular bodies (MVBs), or amphisomes and (2) autolysosomes or residual bodies. The organelles were often found in close-proximity to phagophore-like structures. Immunogold labeling localized mutant HTT to non-fibrillar, electron lucent structures within the lumen of these organelles. In presymptomatic HD, the recruitment organelles were predominantly MVBs/amphisomes, whereas in late-stage HD, there were more autolysosomes or residual bodies. Electron tomograms indicated the fusion of small vesicles with the vacuole within the lumen, suggesting that MVBs develop into residual bodies. We found that markers of MVB-related exocytosis were depleted in presymptomatic mice and throughout the disease course. This suggests that endolysosomal homeostasis has moved away from exocytosis toward lysosome fusion and degradation, in response to the need to clear the chronically aggregating mutant HTT protein, and that this occurs at an early stage in HD pathogenesis.


Subject(s)
Endosomes/pathology , Huntington Disease/pathology , Inclusion Bodies/ultrastructure , Lysosomes/pathology , Neurons/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Endosomes/metabolism , Endosomes/ultrastructure , Gene Knock-In Techniques , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Lysosomes/metabolism , Lysosomes/ultrastructure , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mutation , Neurons/metabolism , Neurons/ultrastructure
3.
Acc Chem Res ; 53(10): 2347-2357, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32975927

ABSTRACT

Huntington's disease (HD) is a progressive, familial neurodegenerative disease triggered by the expansion of a polyglutamine (polyQ) track in the protein huntingtin (htt). PolyQ sequences up to Q36 in htt are not known to be toxic, while polyQ lengths above Q36 almost invariably lead to increased disease risk and decreased ages of onset. The large number of physical states (monomers, dimers, tetramers, non-ß oligomers, nanofibrils, and clustered amyloid fibrils) on the self-association landscape, with their overlapping kinetics of formation, have greatly complicated identification of the molecular species responsible for HD toxicity, drawing attention to the need for innovative approaches.After reports of HD-associated intraneuronal htt inclusions in 1997, we elucidated aggregation mechanisms of both simple polyQ sequences and the more complex polyQ-containing "exon1" fragment of htt (htt-ex1). Grounded in this work, the more recent results described here were made possible by breakthroughs in the molecular design of diagnostic polyQ derivatives and in fluorescence applications for characterizing amyloid assembly intermediates. Thus, insertion of ß-turn-promoting mutations into relatively short, disordered polyQ sequences created "pro-ß-hairpin" polyQs (ßHPs) that exhibit amyloid formation rates comparable to the enhanced rates seen with expanded polyQ peptides. Introduction of "ß-breaker" mutations into these ßHP polyQ sequences created molecules that are blocked from aggregating into amyloid and also can inhibit amyloid formation by other polyQ proteins. These mutational effects were then successfully transferred into more complex htt-ex1 sequence backgrounds. Insights into the aggregation properties of htt-ex1 derivatives-as well as into the nucleation process itself-were obtained using fluorescence correlation spectroscopy (FCS) and a novel thioflavin-T (ThT) protocol that allows quantitation of htt-ex1 assembly intermediates.Using these tools, we quantified physical states of htt-ex1 at different growth times in mammalian PC12 cells engineered for inducible expression of both normal and expanded polyQ repeat length versions of htt-ex1. For expanded polyQ versions, we found tetramers, oligomers, and fibrils (but no monomers) all populated in these cells at a time when the first indication of toxicity (nuclear DNA damage) was observed. These experiments provided a strong hint that monomeric forms of htt-ex1 are not involved in toxicity, but we were otherwise unable to implicate a specific toxic self-assembled state because of the overlapping kinetics of formation. To gain a more intimate focus and control over the timelines of htt-ex1 self-assembly and the resulting toxic response, we engineered various htt-ex1-ßHP molecules-with and without added ß-breaker mutations-that could be expressed in rat neuronal and Drosophila models of HD. In both models, novel htt-ex1-ßHP analogues exhibiting strong aggregation in spite of their very short polyQ repeat lengths proved to be toxic, dramatically breaking the "repeat length paradigm" and strongly suggesting that the toxic species must be some kind of aggregate. In both models, ß-breaker analogues of htt-ex1-ßHP that are slow to make amyloid-instead favoring accumulation of non-ß oligomers-were nontoxic. In contrast, htt-ex1-ßHP analogues that rapidly progress to amyloid states were toxic, suggesting that an aggregate possessing the fundamental amyloid folding motif is very likely the major toxic species in HD.


Subject(s)
Huntington Disease/pathology , Peptides/metabolism , Amino Acid Sequence , Amyloid/metabolism , Animals , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Kinetics , Mutagenesis , PC12 Cells , Peptides/chemistry , Protein Aggregates , Rats
4.
Neurobiol Dis ; 120: 126-138, 2018 12.
Article in English | MEDLINE | ID: mdl-30171891

ABSTRACT

In Huntington disease (HD), an expanded polyglutamine (polyQ > 37) sequence within huntingtin (htt) exon1 leads to enhanced disease risk. It has proved difficult, however, to determine whether the toxic form generated by polyQ expansion is a misfolded or avid-binding monomer, an α-helix-rich oligomer, or a ß-sheet-rich amyloid fibril. Here we describe an engineered htt exon1 analog featuring a short polyQ sequence that nonetheless quickly forms amyloid fibrils and causes HD-like toxicity in rat neurons and Drosophila. Additional modifications within the polyQ segment produce htt exon1 analogs that populate only spherical oligomers and are non-toxic in cells and flies. Furthermore, in mixture with expanded-polyQ htt exon1, the latter analogs in vitro suppress amyloid formation and promote oligomer formation, and in vivo rescue neurons and flies expressing mhtt exon1 from dysfunction and death. Thus, in our experiments, while htt exon1 toxicity tracks with aggregation propensity, it does so in spite of the toxic construct's possessing polyQ tracts well below those normally considered to be disease-associated. That is, aggregation propensity proves to be a more accurate surrogate for toxicity than is polyQ repeat length itself, strongly supporting a major toxic role for htt exon1 aggregation in HD. In addition, the results suggest that the aggregates that are most toxic in these model systems are amyloid-related. These engineered analogs are novel tools for mapping properties of polyQ self-assembly intermediates and products that should similarly be useful in the analysis of other expanded polyQ diseases. Small molecules with similar amyloid inhibitory properties might be developed into effective therapeutic agents.


Subject(s)
Amyloid/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Mutation/genetics , Peptides/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Animals, Newborn , Drosophila , Humans , Rats
5.
Methods Mol Biol ; 1777: 121-144, 2018.
Article in English | MEDLINE | ID: mdl-29744831

ABSTRACT

The red shift in the fluorescence excitation spectra of thioflavin dyes upon binding to fibrils has been a boon to the amyloid field, offering simple and effective methods for the qualitative detection of amyloid in tissue samples and for quantitation of particular fibril preparations with gravimetric linearity. The quantitative aspect of the thioflavin T (ThT) response, however, comes with an important caveat that bestows both significant limitations and great untapped power. It is now well established that amyloid fibrils of different proteins, as well as polymorphic fibrils of the same protein, can exhibit vastly different ThT fluorescence intensities for the same weight concentration of aggregates. Furthermore, the aggregated intermediates commonly observed in amyloid assembly reactions can exhibit aggregate weight-normalized (AWN) ThT fluorescence intensities that vary from essentially zero through a wide range of intermediate values before reaching the intensity of homogeneous, mature amyloid. These features make it very difficult to quantitatively interpret, without additional data, the time-dependent development of ThT fluorescence intensity in an assembly reaction. In this chapter, we describe a method for coupling ex situ ThT fluorescence determinations with an analytical HPLC supported sedimentation assay (also described in detail) that can provide significant new insights into amyloid assembly reactions. The time dependent aggregation data provided by the sedimentation assay reveals a time course of aggregation that is largely independent of aggregate properties. In addition, the combination of these data with ThT measurements of the same reaction time points reveals important aspects of average aggregate structure at each time point. Examples of the use and potential value of AWN-ThT measurements during amyloid assembly Aß and polyglutamine peptides are provided.


Subject(s)
Amyloid/chemistry , Benzothiazoles/chemistry , Protein Multimerization , Chromatography, High Pressure Liquid , Data Interpretation, Statistical , Protein Aggregates , Protein Aggregation, Pathological
6.
J Biol Chem ; 293(4): 1353-1362, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29217771

ABSTRACT

It has been long assumed that post-mitotic neurons only utilize the error-prone non-homologous end-joining pathway to repair double-strand breaks (DSBs) associated with oxidative damage to DNA, given the inability of non-replicating neuronal DNA to utilize a sister chromatid template in the less error-prone homologous recombination (HR) repair pathway. However, we and others have found recently that active transcription triggers a replication-independent recombinational repair mechanism in G0/G1 phase of the cell cycle. Here we observed that the HR repair protein RAD52 is recruited to sites of DNA DSBs in terminally differentiated, post-mitotic neurons. This recruitment is dependent on the presence of a nascent mRNA generated during active transcription, providing evidence that an RNA-templated HR repair mechanism exists in non-dividing, terminally differentiated neurons. This recruitment of RAD52 in neurons is decreased by transcription inhibition. Importantly, we found that high concentrations of amyloid ß, a toxic protein associated with Alzheimer's disease, inhibits the expression and DNA damage response of RAD52, potentially leading to a defect in the error-free, RNA-templated HR repair mechanism. This study shows a novel RNA-dependent repair mechanism of DSBs in post-mitotic neurons and demonstrates that defects in this pathway may contribute to neuronal genomic instability and consequent neurodegenerative phenotypes such as those seen in Alzheimer's disease.


Subject(s)
DNA Breaks, Double-Stranded , Mitosis/physiology , Neurons/metabolism , RNA/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Recombination, Genetic/physiology , Animals , G1 Phase/physiology , Neurons/cytology , RNA/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Rats , Resting Phase, Cell Cycle/physiology
7.
Nat Commun ; 8: 15462, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28537272

ABSTRACT

Polyglutamine expansion in the huntingtin protein is the primary genetic cause of Huntington's disease (HD). Fragments coinciding with mutant huntingtin exon1 aggregate in vivo and induce HD-like pathology in mouse models. The resulting aggregates can have different structures that affect their biochemical behaviour and cytotoxic activity. Here we report our studies of the structure and functional characteristics of multiple mutant htt exon1 fibrils by complementary techniques, including infrared and solid-state NMR spectroscopies. Magic-angle-spinning NMR reveals that fibrillar exon1 has a partly mobile α-helix in its aggregation-accelerating N terminus, and semi-rigid polyproline II helices in the proline-rich flanking domain (PRD). The polyglutamine-proximal portions of these domains are immobilized and clustered, limiting access to aggregation-modulating antibodies. The polymorphic fibrils differ in their flanking domains rather than the polyglutamine amyloid structure. They are effective at seeding polyglutamine aggregation and exhibit cytotoxic effects when applied to neuronal cells.


Subject(s)
Amyloid/chemistry , Huntingtin Protein/genetics , Huntington Disease/genetics , Peptides/chemistry , Protein Aggregation, Pathological/genetics , Amyloid/genetics , Amyloid/metabolism , Amyloid/toxicity , Animals , Cell Line , Exons/genetics , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Huntingtin Protein/toxicity , Huntington Disease/pathology , Magnetic Resonance Spectroscopy , Mice , Microscopy, Electron, Transmission , Mutation , Neurons , Peptides/genetics , Peptides/metabolism , Peptides/toxicity , Protein Aggregation, Pathological/pathology , Protein Structure, Secondary/genetics
8.
J Mol Biol ; 429(2): 308-323, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27986569

ABSTRACT

Candidates for the toxic molecular species in the expanded polyglutamine (polyQ) repeat diseases range from various types of aggregates to "misfolded" monomers. One way to vet these candidates is to develop mutants that restrict conformational landscapes. Previously, we inserted two self-complementary ß-hairpin enhancing motifs into a short polyQ sequence to generate a mutant, here called "ßHP," that exhibits greatly improved amyloid nucleation without measurably enhancing ß-structure in the monomer ensemble. We extend these studies here by introducing single-backbone H-bond impairing modifications αN-methyl Gln or l-Pro at key positions within ßHP. Modifications predicted to allow formation of a fully H-bonded ß-hairpin at the fibril edge while interfering with H-bonding to the next incoming monomer exhibit poor amyloid formation and act as potent inhibitors in trans of simple polyQ peptide aggregation. In contrast, a modification that disrupts intra-ß-hairpin H-bonding within ßHP, while also aggregating poorly, is ineffective at inhibiting amyloid formation in trans. The inhibitors constitute a dynamic version of the edge-protection negative design strategy used in protein evolution to limit unwanted protein aggregation. Our data support a model in which polyQ peptides containing strong ß-hairpin encouraging motifs only rarely form ß-hairpin conformations in the monomer ensemble, but nonetheless take on such conformations at key steps during amyloid formation. The results provide insights into polyQ solution structure and fibril formation while also suggesting an approach to the design of inhibitors of polyQ amyloid growth that focuses on conformational requirements for fibril and nucleus elongation.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptides/chemistry , Protein Engineering , Amino Acid Sequence , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Peptides/antagonists & inhibitors , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Reproducibility of Results
9.
Nat Commun ; 7: 12419, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27546208

ABSTRACT

Since early oligomeric intermediates in amyloid assembly are often transient and difficult to distinguish, characterize and quantify, the mechanistic basis of the initiation of spontaneous amyloid growth is often opaque. We describe here an approach to the analysis of the Aß aggregation mechanism that uses Aß-polyglutamine hybrid peptides designed to retard amyloid maturation and an adjusted thioflavin intensity scale that reveals structural features of aggregation intermediates. The results support an aggregation initiation mechanism for Aß-polyQ hybrids, and by extension for full-length Aß peptides, in which a modular Aß C-terminal segment mediates rapid, non-nucleated formation of α-helical oligomers. The resulting high local concentration of tethered amyloidogenic segments within these α-oligomers facilitates transition to a ß-oligomer population that, via further remodelling and/or elongation steps, ultimately generates mature amyloid. Consistent with this mechanism, an engineered Aß C-terminal fragment delays aggregation onset by Aß-polyglutamine peptides and redirects assembly of Aß42 fibrils.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Protein Aggregation, Pathological/pathology , Protein Multimerization , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Benzothiazoles , Circular Dichroism , Fluorescent Dyes/chemistry , Humans , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptides/chemistry , Peptides/genetics , Protein Engineering , Protein Structure, Secondary , Spectrum Analysis/methods , Thiazoles/chemistry , Time Factors
10.
PLoS One ; 11(6): e0155747, 2016.
Article in English | MEDLINE | ID: mdl-27271685

ABSTRACT

Expansion of the polyglutamine (polyQ) track of the Huntingtin (HTT) protein above 36 is associated with a sharply enhanced risk of Huntington's disease (HD). Although there is general agreement that HTT toxicity resides primarily in N-terminal fragments such as the HTT exon1 protein, there is no consensus on the nature of the physical states of HTT exon1 that are induced by polyQ expansion, nor on which of these states might be responsible for toxicity. One hypothesis is that polyQ expansion induces an alternative, toxic conformation in the HTT exon1 monomer. Alternative hypotheses posit that the toxic species is one of several possible aggregated states. Defining the nature of the toxic species is particularly challenging because of facile interconversion between physical states as well as challenges to identifying these states, especially in vivo. Here we describe the use of fluorescence correlation spectroscopy (FCS) to characterize the detailed time and repeat length dependent self-association of HTT exon1-like fragments both with chemically synthesized peptides in vitro and with cell-produced proteins in extracts and in living cells. We find that, in vitro, mutant HTT exon1 peptides engage in polyQ repeat length dependent dimer and tetramer formation, followed by time dependent formation of diffusible spherical and fibrillar oligomers and finally by larger, sedimentable amyloid fibrils. For expanded polyQ HTT exon1 expressed in PC12 cells, monomers are absent, with tetramers being the smallest molecular form detected, followed in the incubation time course by small, diffusible aggregates at 6-9 hours and larger, sedimentable aggregates that begin to build up at 12 hrs. In these cell cultures, significant nuclear DNA damage appears by 6 hours, followed at later times by caspase 3 induction, mitochondrial dysfunction, and cell death. Our data thus defines limits on the sizes and concentrations of different physical states of HTT exon1 along the reaction profile in the context of emerging cellular distress. The data provide some new candidates for the toxic species and some new reservations about more well-established candidates. Compared to other known markers of HTT toxicity, nuclear DNA damage appears to be a relatively early pathological event.


Subject(s)
Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Multimerization , Amyloid/chemistry , Amyloid/genetics , Amyloid/metabolism , Animals , Cell Survival/genetics , DNA Damage/genetics , Huntingtin Protein/metabolism , Mutant Proteins/metabolism , Mutation/physiology , PC12 Cells , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Folding , Rats
11.
Proc Natl Acad Sci U S A ; 113(6): 1546-51, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26831073

ABSTRACT

Polyglutamine expansion within the exon1 of huntingtin leads to protein misfolding, aggregation, and cytotoxicity in Huntington's disease. This incurable neurodegenerative disease is the most prevalent member of a family of CAG repeat expansion disorders. Although mature exon1 fibrils are viable candidates for the toxic species, their molecular structure and how they form have remained poorly understood. Using advanced magic angle spinning solid-state NMR, we directly probe the structure of the rigid core that is at the heart of huntingtin exon1 fibrils and other polyglutamine aggregates, via measurements of long-range intramolecular and intermolecular contacts, backbone and side-chain torsion angles, relaxation measurements, and calculations of chemical shifts. These experiments reveal the presence of ß-hairpin-containing ß-sheets that are connected through interdigitating extended side chains. Despite dramatic differences in aggregation behavior, huntingtin exon1 fibrils and other polyglutamine-based aggregates contain identical ß-strand-based cores. Prior structural models, derived from X-ray fiber diffraction and computational analyses, are shown to be inconsistent with the solid-state NMR results. Internally, the polyglutamine amyloid fibrils are coassembled from differently structured monomers, which we describe as a type of "intrinsic" polymorphism. A stochastic polyglutamine-specific aggregation mechanism is introduced to explain this phenomenon. We show that the aggregation of mutant huntingtin exon1 proceeds via an intramolecular collapse of the expanded polyglutamine domain and discuss the implications of this observation for our understanding of its misfolding and aggregation mechanisms.


Subject(s)
Exons/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptides/chemistry , Amino Acid Sequence , Amyloid/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/ultrastructure , Peptides/genetics , Protein Structure, Secondary , Stochastic Processes
12.
Methods Mol Biol ; 1345: 67-87, 2016.
Article in English | MEDLINE | ID: mdl-26453206

ABSTRACT

Fluorescence correlation spectroscopy (FCS) is a highly sensitive analytical technique used to measure dynamic molecular parameters, such as diffusion time (from which particle size can be calculated), conformation, and concentration of fluorescent molecules. It has been particularly powerful in characterizing size distributions in molecular associations (e.g., dimer/multimer formation) both in well-behaved thermodynamically equilibrated systems in vitro as well as in more complex environments in vivo. Protein aggregation reactions like amyloid formation, in contrast, are complex, often involving a series of uniquely structured aggregation intermediates appearing at different time scales. Nonetheless, FCS can be used in appropriate cases to characterize the early stages of some aggregation reactions. Here are described step-by-step protocols and experimental procedures for the study of molecular complex formation in aggregation systems as observed in simple buffer systems, cell extracts, and living cells. The methods described are illustrated with examples from studies of the self-assembly of huntingtin fragments, but in principle can be adapted for any aggregating system.


Subject(s)
Amyloid/chemistry , Macromolecular Substances/chemistry , Protein Aggregation, Pathological/genetics , Spectrometry, Fluorescence/methods , Humans , Huntingtin Protein , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics
13.
J Mol Biol ; 428(2 Pt A): 274-291, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26122432

ABSTRACT

Aß43, a product of the proteolysis of the amyloid precursor protein APP, is related to Aß42 by an additional Thr residue at the C-terminus. Aß43 is typically generated at low levels compared with the predominant Aß42 and Aß40 forms, but it has been suggested that this longer peptide might have an impact on amyloid-ß aggregation and Alzheimer's disease that is out of proportion to its brain content. Here, we report that both Aß42 and Aß43 spontaneously aggregate into mature amyloid fibrils via sequential appearance of the same series of oligomeric and protofibrillar intermediates, the earliest of which appears to lack ß-structure. In spite of the additional ß-branched amino acid at the C-terminus, Aß43 fibrils have fewer strong backbone H-bonds than Aß42 fibrils, some of which are lost at the C-terminus. In contrast to previous reports, we found that Aß43 spontaneously aggregates more slowly than Aß42. In addition, Aß43 fibrils are very inefficient at seeding Aß42 amyloid formation, even though Aß42 fibrils efficiently seed amyloid formation by Aß43 monomers. Finally, mixtures of Aß42 and Aß43 aggregate more slowly than Aß42 alone. Both in this Aß42/Aß43 co-aggregation reaction and in cross-seeding by Aß42 fibrils, the structure of the Aß43 in the product fibrils is influenced by the presence of Aß42. The results provide new details of amyloid structure and assembly pathways, an example of structural plasticity in prion-like replication, and data showing that low levels of Aß43 in the brain are unlikely to favorably impact the aggregation of Aß42.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Protein Aggregation, Pathological , Protein Denaturation , Protein Multimerization , Threonine/metabolism
14.
Nat Rev Dis Primers ; 1: 15005, 2015 04 23.
Article in English | MEDLINE | ID: mdl-27188817

ABSTRACT

Huntington disease is devastating to patients and their families - with autosomal dominant inheritance, onset typically in the prime of adult life, progressive course, and a combination of motor, cognitive and behavioural features. The disease is caused by an expanded CAG trinucleotide repeat (of variable length) in HTT, the gene that encodes the protein huntingtin. In mutation carriers, huntingtin is produced with abnormally long polyglutamine sequences that confer toxic gains of function and predispose the protein to fragmentation, resulting in neuronal dysfunction and death. In this Primer, we review the epidemiology of Huntington disease, noting that prevalence is higher than previously thought, geographically variable and increasing. We describe the relationship between CAG repeat length and clinical phenotype, as well as the concept of genetic modifiers of the disease. We discuss normal huntingtin protein function, evidence for differential toxicity of mutant huntingtin variants, theories of huntingtin aggregation and the many different mechanisms of Huntington disease pathogenesis. We describe the genetic and clinical diagnosis of the condition, its clinical assessment and the multidisciplinary management of symptoms, given the absence of effective disease-modifying therapies. We review past and present clinical trials and therapeutic strategies under investigation, including impending trials of targeted huntingtin-lowering drugs and the progress in development of biomarkers that will support the next generation of trials. For an illustrated summary of this Primer, visit: http://go.nature.com/hPMENh.


Subject(s)
Huntington Disease/genetics , Adult , Humans , Huntingtin Protein/genetics , Huntington Disease/epidemiology , Huntington Disease/therapy , Nerve Tissue Proteins/genetics , Peptides/genetics , Phenotype , Prevalence , Trinucleotide Repeat Expansion
15.
Biochim Biophys Acta ; 1852(1): 61-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25463631

ABSTRACT

Triosephosphate isomerase (TPI) is a glycolytic enzyme which homodimerizes for full catalytic activity. Mutations of the TPI gene elicit a disease known as TPI Deficiency, a glycolytic enzymopathy noted for its unique severity of neurological symptoms. Evidence suggests that TPI Deficiency pathogenesis may be due to conformational changes of the protein, likely affecting dimerization and protein stability. In this report, we genetically and physically characterize a human disease-associated TPI mutation caused by an I170V substitution. Human TPI(I170V) elicits behavioral abnormalities in Drosophila. An examination of hTPI(I170V) enzyme kinetics revealed this substitution reduced catalytic turnover, while assessments of thermal stability demonstrated an increase in enzyme stability. The crystal structure of the homodimeric I170V mutant reveals changes in the geometry of critical residues within the catalytic pocket. Collectively these data reveal new observations of the structural and kinetic determinants of TPI Deficiency pathology, providing new insights into disease pathogenesis.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Carbohydrate Metabolism, Inborn Errors/pathology , Catalytic Domain , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Animals , Behavior, Animal , Carbohydrate Metabolism, Inborn Errors/enzymology , Disease Models, Animal , Drosophila , Enzyme Stability , Humans , Mutation , Triose-Phosphate Isomerase/chemistry , Triose-Phosphate Isomerase/genetics
16.
Biochemistry ; 53(42): 6653-66, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25280367

ABSTRACT

In Huntington's disease, expansion of a polyglutamine (polyQ) domain in the huntingtin (htt) protein leads to misfolding and aggregation. There is much interest in the molecular features that distinguish monomeric, oligomeric, and fibrillar species that populate the aggregation pathway and likely differ in cytotoxicity. The mechanism and rate of aggregation are greatly affected by the domains flanking the polyQ segment within exon 1 of htt. A "protective" C-terminal proline-rich flanking domain inhibits aggregation by inducing polyproline II structure (PPII) within an extended portion of polyQ. The N-terminal flanking segment (htt(NT)) adopts an α-helical structure as it drives aggregation, helps stabilize oligomers and fibrils, and is seemingly integral to their supramolecular assembly. Via solid-state nuclear magnetic resonance (ssNMR), we probe how, in the mature fibrils, the htt flanking domains impact the polyQ domain and in particular the localization of the ß-structured amyloid core. Using residue-specific and uniformly labeled samples, we find that the amyloid core occupies most of the polyQ domain but ends just prior to the prolines. We probe the structural and dynamical features of the remarkably abrupt ß-sheet to PPII transition and discuss the potential connections to certain htt-binding proteins. We also examine the htt(NT) α-helix outside the polyQ amyloid core. Despite its presumed structural and demonstrated stabilizing roles in the fibrils, quantitative ssNMR measurements of residue-specific dynamics show that it undergoes distinct solvent-coupled motion. This dynamical feature seems reminiscent of molten-globule-like α-helix-rich features attributed to the nonfibrillar oligomeric species of various amyloidogenic proteins.


Subject(s)
Amyloid/chemistry , Nerve Tissue Proteins/chemistry , Peptides/chemistry , Exons , Humans , Huntingtin Protein , Nerve Tissue Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Protein Structure, Secondary
17.
Biochemistry ; 53(24): 3897-907, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24921664

ABSTRACT

Repeat length disease thresholds vary among the 10 expanded polyglutamine (polyQ) repeat diseases, from about 20 to about 50 glutamine residues. The unique amino acid sequences flanking the polyQ segment are thought to contribute to these repeat length thresholds. The specific portions of the flanking sequences that modulate polyQ properties are not always clear, however. This ambiguity may be important in Huntington's disease (HD), for example, where in vitro studies of aggregation mechanisms have led to distinctly different mechanistic models. Most in vitro studies of the aggregation of the huntingtin (HTT) exon1 fragment implicated in the HD mechanism have been conducted on inexact molecules that are imprecise either on the N-terminus (recombinantly produced peptides) or on the C-terminus (chemically synthesized peptides). In this paper, we investigate the aggregation properties of chemically synthesized HTT exon1 peptides that are full-length and complete, containing both normal and expanded polyQ repeat lengths, and compare the results directly to previously investigated molecules containing truncated C-termini. The results on the full-length peptides are consistent with a two-step aggregation mechanism originally developed based on studies of the C-terminally truncated analogues. Thus, we observe relatively rapid formation of spherical oligomers containing from 100 to 600 HTT exon1 molecules and intermediate formation of short protofibril-like structures containing from 500 to 2600 molecules. In contrast to this relatively rapid assembly, mature HTT exon1 amyloid requires about one month to dissociate in vitro, which is similar to the time required for neuronal HTT exon1 aggregates to disappear in vivo after HTT production is discontinued.


Subject(s)
Nerve Tissue Proteins/chemistry , Protein Structure, Quaternary , Exons , Humans , Huntingtin Protein , Huntington Disease/genetics , Kinetics , Nerve Tissue Proteins/chemical synthesis , Peptides/chemistry
18.
Nat Commun ; 5: 3508, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24691351

ABSTRACT

Naturally occurring fragments of the abundant semen proteins prostatic acid phosphatase (PAP) and semenogelins form amyloid fibrils in vitro. These fibrils boost HIV infection and may play a key role in the spread of the AIDS pandemic. However, the presence of amyloid fibrils in semen remained to be demonstrated. Here, we use state of the art confocal and electron microscopy techniques for direct imaging of amyloid fibrils in human ejaculates. We detect amyloid aggregates in all semen samples and find that they partially consist of PAP fragments, interact with HIV particles and increase viral infectivity. Our results establish semen as a body fluid that naturally contains amyloid fibrils that are exploited by HIV to promote its sexual transmission.


Subject(s)
Amyloid/metabolism , HIV Infections/metabolism , HIV-1/physiology , Semen/metabolism , Acid Phosphatase , Amyloid/ultrastructure , HIV Infections/virology , Humans , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Protein Tyrosine Phosphatases/metabolism , Semen/virology , Seminal Vesicle Secretory Proteins/metabolism
19.
J Biol Chem ; 289(15): 10254-10260, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24596088

ABSTRACT

There are now 10 expanded CAG repeat diseases in which both disease risk and age of onset are strongly dependent on the repeat length of the polyglutamine (polyQ) sequence in the disease protein. Large, polyQ-rich inclusions in patient brains and in cell and animal models are consistent with the involvement of polyQ aggregation in the disease mechanism. This possibility is reinforced by studies showing strong repeat length dependence to the aggregation process, qualitatively mirroring the repeat length dependence of disease risk. Our understanding of the underlying biophysical principles that mediate the repeat length dependence of aggregation, however, is far from complete. A previous study of simple polyQ peptides showed that N*, the size of the critical nucleus that controls onset of aggregation, decreases from unfavorable tetramer to favorable monomer over the range Q23 to Q26. These data, however, do not explain why, for all peptides exhibiting N* ∼ 1, spontaneous aggregation rates continue to increase with increasing repeat length. Here we describe a novel kinetics analyses that maps out the nonlinear dependence with repeat length of a nucleation efficiency term that is likely related to aspects of nucleus structure. This trend accounts for why nucleus size increases to tetrameric at repeat lengths of Q23 or below. Intriguingly, both aggregation and age of onset trend with repeat length in similar ways, exhibiting large changes per added Gln at low repeat lengths and small changes per added Gln at relatively long repeat lengths. Fibril stability also increases with repeat length in a nonlinear fashion.


Subject(s)
Amyloid/chemistry , Trinucleotide Repeats , Age of Onset , Biophysical Phenomena , Humans , Huntington Disease/metabolism , Kinetics , Peptides/chemistry , Protein Conformation , Temperature
20.
Biopolymers ; 102(2): 206-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24488729

ABSTRACT

Many amyloidogenic peptides are highly hydrophobic, introducing significant challenges to obtaining high quality peptides by chemical synthesis. For example, while good yield and purity can be obtained in the solid-phase synthesis of the Alzheimer's plaque peptide Aß40, addition of a C-terminal Ile-Ala sequence to generate the more toxic Aß42 molecule creates a much more difficult synthesis resulting in low yields and purities. We describe here a new method that significantly improves the Fmoc solid-phase synthesis of Aß peptides. In our method, Lys residues are linked to the desired peptide's C-terminus through standard peptide bonds during the synthesis. These Lys residues are then removed post-purification using immobilized carboxypeptidase B (CPB). With this method we obtained both Aß42 and Aß46 of superior quality that, for Aß42, rivals that obtained by recombinant expression. Intriguingly, the method appears to provide independent beneficial effects on both the total synthetic yield and on purification yield and final purity. Reversible Lys addition with CPB removal should be a generally useful method for making hydrophobic peptides that is applicable to any sequence not ending in Arg or Lys. As expected from the additional hydrophobicity of Aß46, which is extended from the sequence Aß42 by a C-terminal Thr-Val-Ile-Val sequence, this peptide makes typical amyloid at rates significantly faster than for Aß42 or Aß40. The enhanced amyloidogenicity of Aß46 suggests that, even though it is present in relatively low amounts in the human brain, it could play a significant role in helping to initiate Aß amyloid formation.


Subject(s)
Amyloid beta-Peptides/chemical synthesis , Carboxypeptidase B/metabolism , Hydrophobic and Hydrophilic Interactions , Lysine/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/isolation & purification , Amyloid beta-Peptides/ultrastructure , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Enzymes, Immobilized/metabolism , Kinetics , Molecular Sequence Data , Protein Aggregates , Spectrometry, Mass, Electrospray Ionization , Spectroscopy, Fourier Transform Infrared , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...