Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(26): 5766-5783.e25, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38134874

ABSTRACT

The enhanced cognitive abilities characterizing the human species result from specialized features of neurons and circuits. Here, we report that the hominid-specific gene LRRC37B encodes a receptor expressed in human cortical pyramidal neurons (CPNs) and selectively localized to the axon initial segment (AIS), the subcellular compartment triggering action potentials. Ectopic expression of LRRC37B in mouse CPNs in vivo leads to reduced intrinsic excitability, a distinctive feature of some classes of human CPNs. Molecularly, LRRC37B binds to the secreted ligand FGF13A and to the voltage-gated sodium channel (Nav) ß-subunit SCN1B. LRRC37B concentrates inhibitory effects of FGF13A on Nav channel function, thereby reducing excitability, specifically at the AIS level. Electrophysiological recordings in adult human cortical slices reveal lower neuronal excitability in human CPNs expressing LRRC37B. LRRC37B thus acts as a species-specific modifier of human neuron excitability, linking human genome and cell evolution, with important implications for human brain function and diseases.


Subject(s)
Neurons , Pyramidal Cells , Voltage-Gated Sodium Channels , Animals , Humans , Mice , Action Potentials/physiology , Axons/metabolism , Neurons/metabolism , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
2.
Nat Rev Mol Cell Biol ; 24(12): 912-933, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37684425

ABSTRACT

Despite advances in machine learning-based protein structure prediction, we are still far from fully understanding how proteins fold into their native conformation. The conventional notion that polypeptides fold spontaneously to their biologically active states has gradually been replaced by our understanding that cellular protein folding often requires context-dependent guidance from molecular chaperones in order to avoid misfolding. Misfolded proteins can aggregate into larger structures, such as amyloid fibrils, which perpetuate the misfolding process, creating a self-reinforcing cascade. A surge in amyloid fibril structures has deepened our comprehension of how a single polypeptide sequence can exhibit multiple amyloid conformations, known as polymorphism. The assembly of these polymorphs is not a random process but is influenced by the specific conditions and tissues in which they originate. This observation suggests that, similar to the folding of native proteins, the kinetics of pathological amyloid assembly are modulated by interactions specific to cells and tissues. Here, we review the current understanding of how intrinsic protein conformational propensities are modulated by physiological and pathological interactions in the cell to shape protein misfolding and aggregation pathology.


Subject(s)
Amyloid , Protein Folding , Protein Conformation , Amyloid/metabolism , Peptides , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
3.
Cell ; 170(3): 443-456.e14, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753424

ABSTRACT

Alzheimer's disease (AD)-linked mutations in Presenilins (PSEN) and the amyloid precursor protein (APP) lead to production of longer amyloidogenic Aß peptides. The shift in Aß length is fundamental to the disease; however, the underlying mechanism remains elusive. Here, we show that substrate shortening progressively destabilizes the consecutive enzyme-substrate (E-S) complexes that characterize the sequential γ-secretase processing of APP. Remarkably, pathogenic PSEN or APP mutations further destabilize labile E-S complexes and thereby promote generation of longer Aß peptides. Similarly, destabilization of wild-type E-S complexes by temperature, compounds, or detergent promotes release of amyloidogenic Aß. In contrast, E-Aßn stabilizers increase γ-secretase processivity. Our work presents a unifying model for how PSEN or APP mutations enhance amyloidogenic Aß production, suggests that environmental factors may increase AD risk, and provides the theoretical basis for the development of γ-secretase/substrate stabilizing compounds for the prevention of AD.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Presenilin-1/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Brain/metabolism , Brain/pathology , Cell Line , Endopeptidases , Enzyme Stability , Female , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Models, Molecular , Mutation , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Presenilin-1/chemistry , Presenilin-1/genetics
5.
Cell ; 166(1): 193-208, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27293189

ABSTRACT

γ-Secretases are a family of intramembrane-cleaving proteases involved in various signaling pathways and diseases, including Alzheimer's disease (AD). Cells co-express differing γ-secretase complexes, including two homologous presenilins (PSENs). We examined the significance of this heterogeneity and identified a unique motif in PSEN2 that directs this γ-secretase to late endosomes/lysosomes via a phosphorylation-dependent interaction with the AP-1 adaptor complex. Accordingly, PSEN2 selectively cleaves late endosomal/lysosomal localized substrates and generates the prominent pool of intracellular Aß that contains longer Aß; familial AD (FAD)-associated mutations in PSEN2 increased the levels of longer Aß further. Moreover, a subset of FAD mutants in PSEN1, normally more broadly distributed in the cell, phenocopies PSEN2 and shifts its localization to late endosomes/lysosomes. Thus, localization of γ-secretases determines substrate specificity, while FAD-causing mutations strongly enhance accumulation of aggregation-prone Aß42 in intracellular acidic compartments. The findings reveal potentially important roles for specific intracellular, localized reactions contributing to AD pathogenesis.


Subject(s)
Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/analysis , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Presenilin-2/analysis , Adaptor Protein Complex 1/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Motifs , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Endosomes/chemistry , Humans , Lysosomes/chemistry , Mice , Presenilin-1/analysis , Presenilin-1/chemistry , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/chemistry , Presenilin-2/genetics , Presenilin-2/metabolism , Rats , Substrate Specificity
6.
Nature ; 612(7938): 123-131, 2022 12.
Article in English | MEDLINE | ID: mdl-36385530

ABSTRACT

Aggregates of medin amyloid (a fragment of the protein MFG-E8, also known as lactadherin) are found in the vasculature of almost all humans over 50 years of age1,2, making it the most common amyloid currently known. We recently reported that medin also aggregates in blood vessels of ageing wild-type mice, causing cerebrovascular dysfunction3. Here we demonstrate in amyloid-ß precursor protein (APP) transgenic mice and in patients with Alzheimer's disease that medin co-localizes with vascular amyloid-ß deposits, and that in mice, medin deficiency reduces vascular amyloid-ß deposition by half. Moreover, in both the mouse and human brain, MFG-E8 is highly enriched in the vasculature and both MFG-E8 and medin levels increase with the severity of vascular amyloid-ß burden. Additionally, analysing data from 566 individuals in the ROSMAP cohort, we find that patients with Alzheimer's disease have higher MFGE8 expression levels, which are attributable to vascular cells and are associated with increased measures of cognitive decline, independent of plaque and tau pathology. Mechanistically, we demonstrate that medin interacts directly with amyloid-ß to promote its aggregation, as medin forms heterologous fibrils with amyloid-ß, affects amyloid-ß fibril structure, and cross-seeds amyloid-ß aggregation both in vitro and in vivo. Thus, medin could be a therapeutic target for prevention of vascular damage and cognitive decline resulting from amyloid-ß deposition in the blood vessels of the brain.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Animals , Humans , Mice , Middle Aged , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cognitive Dysfunction , Mice, Transgenic , Plaque, Amyloid/metabolism , tau Proteins/metabolism
7.
Trends Biochem Sci ; 47(3): 194-205, 2022 03.
Article in English | MEDLINE | ID: mdl-34561149

ABSTRACT

Protein aggregation propensity is a pervasive and seemingly inescapable property of proteomes. Strikingly, a significant fraction of the proteome is supersaturated, meaning that, for these proteins, their native conformation is less stable than the aggregated state. Maintaining the integrity of a proteome under such conditions is precarious and requires energy-consuming proteostatic regulation. Why then is aggregation propensity maintained at such high levels over long evolutionary timescales? Here, we argue that the conformational stability of the native and aggregated states are correlated thermodynamically and that codon usage strengthens this correlation. As a result, the folding of stable proteins requires kinetic control to avoid aggregation, provided by aggregation gatekeepers. These unique residues are evolutionarily selected to kinetically favor native folding, either on their own or by coopting chaperones.


Subject(s)
Molecular Chaperones , Protein Folding , Kinetics , Molecular Chaperones/metabolism , Protein Aggregates , Protein Conformation , Proteome
8.
EMBO J ; 41(2): e108591, 2022 12 17.
Article in English | MEDLINE | ID: mdl-34842295

ABSTRACT

It is still unclear why pathological amyloid deposition initiates in specific brain regions or why some cells or tissues are more susceptible than others. Amyloid deposition is determined by the self-assembly of short protein segments called aggregation-prone regions (APRs) that favour cross-ß structure. Here, we investigated whether Aß amyloid assembly can be modified by heterotypic interactions between Aß APRs and short homologous segments in otherwise unrelated human proteins. Mining existing proteomics data of Aß plaques from AD patients revealed an enrichment in proteins that harbour such homologous sequences to the Aß APRs, suggesting heterotypic amyloid interactions may occur in patients. We identified homologous APRs from such proteins and show that they can modify Aß assembly kinetics, fibril morphology and deposition pattern in vitro. Moreover, we found three of these proteins upon transient expression in an Aß reporter cell line promote Aß amyloid aggregation. Strikingly, we did not find a bias towards heterotypic interactions in plaques from AD mouse models where Aß self-aggregation is observed. Based on these data, we propose that heterotypic APR interactions may play a hitherto unrealized role in amyloid-deposition diseases.


Subject(s)
Amyloid beta-Peptides/metabolism , Protein Interaction Maps , Proteome/metabolism , Amyloid beta-Peptides/chemistry , HEK293 Cells , Humans , Protein Binding , Protein Multimerization , Proteome/chemistry
9.
Mol Cell ; 71(5): 689-702.e9, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193096

ABSTRACT

Hsp90 is an essential chaperone that guards proteome integrity and amounts to 2% of cellular protein. We now find that Hsp90 also has the ability to directly interact with and deform membranes via an evolutionarily conserved amphipathic helix. Using a new cell-free system and in vivo measurements, we show this amphipathic helix allows exosome release by promoting the fusion of multivesicular bodies (MVBs) with the plasma membrane. We dissect the relationship between Hsp90 conformation and membrane-deforming function and show that mutations and drugs that stabilize the open Hsp90 dimer expose the helix and allow MVB fusion, while these effects are blocked by the closed state. Hence, we structurally separated the Hsp90 membrane-deforming function from its well-characterized chaperone activity, and we show that this previously unrecognized function is required for exosome release.


Subject(s)
Cell Membrane/metabolism , Exosomes/metabolism , HSP90 Heat-Shock Proteins/metabolism , Animals , Cell-Free System/metabolism , Drosophila/metabolism , Female , Male , Molecular Chaperones/metabolism , Multivesicular Bodies/metabolism , Protein Binding/physiology , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 120(9): e2214921120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36812200

ABSTRACT

Mutant KRAS is a major driver of oncogenesis in a multitude of cancers but remains a challenging target for classical small molecule drugs, motivating the exploration of alternative approaches. Here, we show that aggregation-prone regions (APRs) in the primary sequence of the oncoprotein constitute intrinsic vulnerabilities that can be exploited to misfold KRAS into protein aggregates. Conveniently, this propensity that is present in wild-type KRAS is increased in the common oncogenic mutations at positions 12 and 13. We show that synthetic peptides (Pept-ins™) derived from two distinct KRAS APRs could induce the misfolding and subsequent loss of function of oncogenic KRAS, both of recombinantly produced protein in solution, during cell-free translation and in cancer cells. The Pept-ins exerted antiproliferative activity against a range of mutant KRAS cell lines and abrogated tumor growth in a syngeneic lung adenocarcinoma mouse model driven by mutant KRAS G12V. These findings provide proof-of-concept that the intrinsic misfolding propensity of the KRAS oncoprotein can be exploited to cause its functional inactivation.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , Animals , Mice , Cell Line, Tumor , Lung Neoplasms/genetics , Mutation , Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Protein Folding
11.
EMBO J ; 40(21): e107568, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34617299

ABSTRACT

While aggregation-prone proteins are known to accelerate aging and cause age-related diseases, the cellular mechanisms that drive their cytotoxicity remain unresolved. The orthologous proteins MOAG-4, SERF1A, and SERF2 have recently been identified as cellular modifiers of such proteotoxicity. Using a peptide array screening approach on human amyloidogenic proteins, we found that SERF2 interacted with protein segments enriched in negatively charged and hydrophobic, aromatic amino acids. The absence of such segments, or the neutralization of the positive charge in SERF2, prevented these interactions and abolished the amyloid-promoting activity of SERF2. In protein aggregation models in the nematode worm Caenorhabditis elegans, protein aggregation and toxicity were suppressed by mutating the endogenous locus of MOAG-4 to neutralize charge. Our data indicate that MOAG-4 and SERF2 drive protein aggregation and toxicity by interactions with negatively charged segments in aggregation-prone proteins. Such charge interactions might accelerate primary nucleation of amyloid by initiating structural changes and by decreasing colloidal stability. Our study points at charge interactions between cellular modifiers and amyloidogenic proteins as potential targets for interventions to reduce age-related protein toxicity.


Subject(s)
Amyloid/chemistry , Amyloidogenic Proteins/chemistry , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Nerve Tissue Proteins/chemistry , alpha-Synuclein/chemistry , Amino Acid Sequence , Amyloid/genetics , Amyloid/metabolism , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Animals , Binding Sites , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Protein Aggregates , Protein Array Analysis , Protein Binding , Signal Transduction , Static Electricity , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
12.
Bioinformatics ; 40(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38662570

ABSTRACT

MOTIVATION: Proteins, the molecular workhorses of biological systems, execute a multitude of critical functions dictated by their precise three-dimensional structures. In a complex and dynamic cellular environment, proteins can undergo misfolding, leading to the formation of aggregates that take up various forms, including amorphous and ordered aggregation in the shape of amyloid fibrils. This phenomenon is closely linked to a spectrum of widespread debilitating pathologies, such as Alzheimer's disease, Parkinson's disease, type-II diabetes, and several other proteinopathies, but also hampers the engineering of soluble agents, as in the case of antibody development. As such, the accurate prediction of aggregation propensity within protein sequences has become pivotal due to profound implications in understanding disease mechanisms, as well as in improving biotechnological and therapeutic applications. RESULTS: We previously developed Cordax, a structure-based predictor that utilizes logistic regression to detect aggregation motifs in protein sequences based on their structural complementarity to the amyloid cross-beta architecture. Here, we present a dedicated web server interface for Cordax. This online platform combines several features including detailed scoring of sequence aggregation propensity, as well as 3D visualization with several customization options for topology models of the structural cores formed by predicted aggregation motifs. In addition, information is provided on experimentally determined aggregation-prone regions that exhibit sequence similarity to predicted motifs, scores, and links to other predictor outputs, as well as simultaneous predictions of relevant sequence propensities, such as solubility, hydrophobicity, and secondary structure propensity. AVAILABILITY AND IMPLEMENTATION: The Cordax webserver is freely accessible at https://cordax.switchlab.org/.


Subject(s)
Software , Protein Aggregates , Internet , Amyloid/chemistry , Proteins/chemistry , Amino Acid Motifs , Humans , Protein Conformation , Sequence Analysis, Protein/methods , Amino Acid Sequence
13.
Bioinformatics ; 40(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38514422

ABSTRACT

MOTIVATION: Deep learning algorithms applied to structural biology often struggle to converge to meaningful solutions when limited data is available, since they are required to learn complex physical rules from examples. State-of-the-art force-fields, however, cannot interface with deep learning algorithms due to their implementation. RESULTS: We present MadraX, a forcefield implemented as a differentiable PyTorch module, able to interact with deep learning algorithms in an end-to-end fashion. AVAILABILITY AND IMPLEMENTATION: MadraX documentation, together with tutorials and installation guide, is available at madrax.readthedocs.io.


Subject(s)
Deep Learning , Algorithms , Documentation
14.
Mol Cell ; 65(6): 1044-1055.e5, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28306503

ABSTRACT

Liquid-liquid phase separation (LLPS) of RNA-binding proteins plays an important role in the formation of multiple membrane-less organelles involved in RNA metabolism, including stress granules. Defects in stress granule homeostasis constitute a cornerstone of ALS/FTLD pathogenesis. Polar residues (tyrosine and glutamine) have been previously demonstrated to be critical for phase separation of ALS-linked stress granule proteins. We now identify an active role for arginine-rich domains in these phase separations. Moreover, arginine-rich dipeptide repeats (DPRs) derived from C9orf72 hexanucleotide repeat expansions similarly undergo LLPS and induce phase separation of a large set of proteins involved in RNA and stress granule metabolism. Expression of arginine-rich DPRs in cells induced spontaneous stress granule assembly that required both eIF2α phosphorylation and G3BP. Together with recent reports showing that DPRs affect nucleocytoplasmic transport, our results point to an important role for arginine-rich DPRs in the pathogenesis of C9orf72 ALS/FTLD.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Arginine/metabolism , Cytoplasmic Granules/metabolism , Dipeptides/metabolism , Intrinsically Disordered Proteins/metabolism , Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Arginine/chemistry , C9orf72 Protein , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytoplasmic Granules/pathology , DNA Helicases , Dipeptides/chemistry , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , HeLa Cells , Humans , Intrinsically Disordered Proteins/chemistry , Lipid Droplets/metabolism , Phosphorylation , Poly-ADP-Ribose Binding Proteins , Protein Domains , Proteins/chemistry , RNA/metabolism , RNA Helicases , RNA Recognition Motif Proteins , Time Factors , Transfection
15.
EMBO J ; 39(11): e102864, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32237079

ABSTRACT

Many chaperones favour binding to hydrophobic sequences that are flanked by basic residues while disfavouring acidic residues. However, the origin of this bias in protein quality control remains poorly understood. Here, we show that while acidic residues are the most efficient aggregation inhibitors, they are also less compatible with globular protein structure than basic amino acids. As a result, while acidic residues allow for chaperone-independent control of aggregation, their use is structurally limited. Conversely, we find that, while being more compatible with globular structure, basic residues are not sufficient to autonomously suppress protein aggregation. Using Hsp70, we show that chaperones with a bias towards basic residues are structurally adapted to prioritize aggregating sequences whose structural context forced the use of the less effective basic residues. The hypothesis that emerges from our analysis is that the bias of many chaperones for basic residues results from fundamental thermodynamic and kinetic constraints of globular structure. This also suggests the co-evolution of basic residues and chaperones allowed for an expansion of structural variety in the protein universe.


Subject(s)
Escherichia coli Proteins , Escherichia coli , HSP70 Heat-Shock Proteins , Protein Aggregates , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Hydrophobic and Hydrophilic Interactions
16.
BMC Bioinformatics ; 24(1): 287, 2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37464277

ABSTRACT

BACKGROUND: Next-generation sequencing technologies yield large numbers of genetic alterations, of which a subset are missense variants that alter an amino acid in the protein product. These variants can have a potentially destabilizing effect leading to an increased risk of misfolding and aggregation. Multiple software tools exist to predict the effect of single-nucleotide variants on proteins, however, a pipeline integrating these tools while starting from an NGS data output list of variants is lacking. RESULTS: The previous version SNPeffect 4.0 (De Baets in Nucleic Acids Res 40(D1):D935-D939, 2011) provided an online database containing pre-calculated variant effects and low-throughput custom variant analysis. Here, we built an automated and parallelized pipeline that analyzes the impact of missense variants on the aggregation propensity and structural stability of proteins starting from the Variant Call Format as input. The pipeline incorporates the AlphaFold Protein Structure Database to achieve high coverage for structural stability analyses using the FoldX force field. The effect on aggregation-propensity is analyzed using the established predictors TANGO and WALTZ. The pipeline focuses solely on the human proteome and can be used to analyze proteome stability/damage in a given sample based on sequencing results. CONCLUSION: We provide a bioinformatics pipeline that allows structural phenotyping from sequencing data using established stability and aggregation predictors including FoldX, TANGO, and WALTZ; and structural proteome coverage provided by the AlphaFold database. The pipeline and installation guide are freely available for academic users on https://github.com/vibbits/snpeffect and requires a computer cluster.


Subject(s)
Proteome , Software , Humans , Mutation , Mutant Proteins , Databases, Protein , High-Throughput Nucleotide Sequencing
17.
Bioinformatics ; 38(9): 2636-2638, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35199146

ABSTRACT

SUMMARY: Amyloid polymorphism is emerging as a key property that is differentially linked to various conformational diseases, including major neurodegenerative disorders, but also as a feature that potentially relates to complex structural mechanisms mediating transmissibility barriers and selective vulnerability of amyloids. In response to the rapidly expanding number of amyloid fibril structures formed by full-length proteins, we here have developed StAmP-DB, a public database that supports the curation and cross-comparison of experimentally determined three-dimensional amyloid polymorph structures. AVAILABILITY AND IMPLEMENTATION: StAmP-DB is freely accessible for queries and downloads at https://stamp.switchlab.org.


Subject(s)
Amyloid , Neurodegenerative Diseases , Humans , Amyloid/chemistry
18.
Brain ; 145(10): 3558-3570, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36270003

ABSTRACT

Alzheimer's disease is neuropathologically characterized by the deposition of the amyloid ß-peptide (Aß) as amyloid plaques. Aß plaque pathology starts in the neocortex before it propagates into further brain regions. Moreover, Aß aggregates undergo maturation indicated by the occurrence of post-translational modifications. Here, we show that propagation of Aß plaques is led by presumably non-modified Aß followed by Aß aggregate maturation. This sequence was seen neuropathologically in human brains and in amyloid precursor protein transgenic mice receiving intracerebral injections of human brain homogenates from cases varying in Aß phase, Aß load and Aß maturation stage. The speed of propagation after seeding in mice was best related to the Aß phase of the donor, the progression speed of maturation to the stage of Aß aggregate maturation. Thus, different forms of Aß can trigger propagation/maturation of Aß aggregates, which may explain the lack of success when therapeutically targeting only specific forms of Aß.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Animals , Humans , Mice , Amyloid beta-Peptides/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Plaque, Amyloid/metabolism , Mice, Transgenic , Brain/pathology , Disease Models, Animal
19.
Mol Cell ; 59(4): 615-27, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26257283

ABSTRACT

Excessive expansions of glutamine (Q)-rich repeats in various human proteins are known to result in severe neurodegenerative disorders such as Huntington's disease and several ataxias. However, the physiological role of these repeats and the consequences of more moderate repeat variation remain unknown. Here, we demonstrate that Q-rich domains are highly enriched in eukaryotic transcription factors where they act as functional modulators. Incremental changes in the number of repeats in the yeast transcriptional regulator Ssn6 (Cyc8) result in systematic, repeat-length-dependent variation in expression of target genes that result in direct phenotypic changes. The function of Ssn6 increases with its repeat number until a certain threshold where further expansion leads to aggregation. Quantitative proteomic analysis reveals that the Ssn6 repeats affect its solubility and interactions with Tup1 and other regulators. Thus, Q-rich repeats are dynamic functional domains that modulate a regulator's innate function, with the inherent risk of pathogenic repeat expansions.


Subject(s)
Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Base Sequence , DNA, Fungal/genetics , Gene Expression Regulation, Fungal , Glutamine/chemistry , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Stability , Repetitive Sequences, Amino Acid , Repressor Proteins/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Solubility
20.
Proc Natl Acad Sci U S A ; 117(46): 29090-29100, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33122432

ABSTRACT

TRPM3 channels play important roles in the detection of noxious heat and in inflammatory thermal hyperalgesia. The activity of these ion channels in somatosensory neurons is tightly regulated by µ-opioid receptors through the signaling of Gßγ proteins, thereby reducing TRPM3-mediated pain. We show here that Gßγ directly binds to a domain of 10 amino acids in TRPM3 and solve a cocrystal structure of this domain together with Gßγ. Using these data and mutational analysis of full-length proteins, we pinpoint three amino acids in TRPM3 and their interacting partners in Gß1 that are individually necessary for TRPM3 inhibition by Gßγ. The 10-amino-acid Gßγ-interacting domain in TRPM3 is subject to alternative splicing. Its inclusion in or exclusion from TRPM3 channel proteins therefore provides a mechanism for switching on or off the inhibitory action that Gßγ proteins exert on TRPM3 channels.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/pharmacology , TRPM Cation Channels/chemistry , TRPM Cation Channels/drug effects , TRPM Cation Channels/metabolism , Binding Sites , Calcium/metabolism , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , HEK293 Cells , Humans , Hyperalgesia/metabolism , Models, Molecular , Mutation , Neurons/metabolism , Pain/metabolism , Receptors, Opioid/metabolism , TRPM Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL