Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 556
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166928, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38660915

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disorder with clinical presentations of moderate to severe cognitive, motor, and psychiatric disturbances. HD is caused by the trinucleotide repeat expansion of CAG of the huntingtin (HTT) gene. The mutant HTT protein containing pathological polyglutamine (polyQ) extension is prone to misfolding and aggregation in the brain. It has previously been observed that copper and iron concentrations are increased in the striata of post-mortem human HD brains. Although it has been shown that the accumulation of mutant HTT protein can interact with copper, the underlying HD progressive phenotypes due to copper overload remains elusive. Here, in a Drosophila model of HD, we showed that copper induces dose-dependent aggregational toxicity and enhancement of Htt-induced neurodegeneration. Specifically, we found that copper increases mutant Htt aggregation, enhances the accumulation of Thioflavin S positive ß-amyloid structures within Htt aggregates, and consequently alters autophagy in the brain. Administration of copper chelator D-penicillamine (DPA) through feeding significantly decreases ß-amyloid aggregates in the HD pathological model. These findings reveal a direct role of copper in potentiating mutant Htt protein-induced aggregational toxicity, and further indicate the potential impact of environmental copper exposure in the disease onset and progression of HD.


Subject(s)
Copper , Huntingtin Protein , Huntington Disease , Animals , Humans , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Autophagy/drug effects , Autophagy/genetics , Brain/metabolism , Brain/pathology , Brain/drug effects , Copper/metabolism , Copper/toxicity , Disease Models, Animal , Drosophila melanogaster/drug effects , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mutation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology
2.
Langmuir ; 39(51): 18923-18934, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38079396

ABSTRACT

Alzheimer's disease (AD) is related to the fibrillation of the Aß peptides at neuronal membranes, a process that depends on the lipid composition and may impart different physical states to the membrane. In the present work, we study the properties of the Aß peptide when mixed with a zwitterionic lipid (DMPC), using the Langmuir monolayer technique as an approach to control membrane physical conditions. First, we build on previous characterizations of pure Aß monolayers and observe that, in addition to high shear, these films present a pronounced compressional hysteresis. When Aß is assembled with DMPC in a binary film, the resulting membranes become heterogeneous, with a peptide-enriched phase distributed in a network-like pattern, and they exhibit a lateral transition that depends on the Aß content. At lower peptide proportions, the films segregate into two well-defined phases: one consisting of lipids and another enriched with peptides. The reflectivity of these phases differs from that obtained for pure Aß films. Thus, the formed fibers effectively cover most of the interface area and remain stable at higher pressures (from 20 to 30 mN m-1 depending on Aß content) compared to pure peptide films (17 mN m-1). Furthermore, such structures induce a compressional hysteresis in the film, similar to that of pure peptide films (which is nonexistent in the pure lipid monolayer), even at low peptide proportions. We claim that the mechanical properties at the interface are governed by the size of the fibril-like structures. Based on the low molar fractions and surface packing at which these phenomena were observed, we postulate that as a consequence of peptide intermolecular interactions, Aß may have drastic effects on the molecular arrangement and mechanical properties of a lipid membrane.


Subject(s)
Amyloid beta-Peptides , Mechanical Phenomena , Membrane Lipids , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Membrane Lipids/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Microscopy, Electron, Scanning , Protein Aggregation, Pathological/pathology , Humans
3.
Biomed Pharmacother ; 148: 112773, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35245734

ABSTRACT

Growing evidence suggests that neuronal dysfunction in the endo-lysosomal and autophagic processes contributes to the onset and progression of neurodegenerative diseases such as Alzheimer's disease (AD). Since they are the primary cellular systems involved in the production and clearance of aggregated amyloid plaques, endo-lysosomal or autophagic equilibrium must be maintained throughout life. As a result, variations in the autophagic and endo-lysosomal torrent, as a measure of degenerative function in these sections or pathways, may have a direct impact on disease-related processes, such as Aß clearance from the brain and interneuronal deposition of Aß and tau aggregates, thus disrupting synaptic plasticity. The discovery of several chromosomal factors for Alzheimer's disease that are clinically linked to regulation of the endocytic pathway, including protein aggregation and removal, supports the theory that the endo-lysosomal/autophagic torrent is more susceptible to impairment, especially as people age, thus catalysing the onset of disease. Although the role of endo-lysosomal/autophagic dysfunction in neurodegeneration has progressed in recent years, the field remains underdeveloped. Because of its possible therapeutic implications in Alzheimer's disease, further study is needed to explain the possibilities for effective autophagy regulation.


Subject(s)
Alzheimer Disease/pathology , Endosomes/metabolism , Lysosomes/metabolism , rab5 GTP-Binding Proteins/metabolism , Aging/pathology , Endocytosis/physiology , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/pathology , tau Proteins/metabolism
4.
Molecules ; 27(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35164285

ABSTRACT

Amyloidosis is a common pathological event in which proteins self-assemble into misfolded soluble and insoluble molecular forms, oligomers and fibrils that are often toxic to cells. Notably, aggregation-prone human islet amyloid polypeptide (hIAPP), or amylin, is a pancreatic hormone linked to islet ß-cells demise in diabetics. The unifying mechanism by which amyloid proteins, including hIAPP, aggregate and kill cells is still matter of debate. The pathology of type-2 diabetes mellitus (T2DM) is characterized by extracellular and intracellular accumulation of toxic hIAPP species, soluble oligomers and insoluble fibrils in pancreatic human islets, eventually leading to loss of ß-cell mass. This review focuses on molecular, biochemical and cell-biology studies exploring molecular mechanisms of hIAPP synthesis, trafficking and degradation in the pancreas. In addition to hIAPP turnover, the dynamics and the mechanisms of IAPP-membrane interactions; hIAPP aggregation and toxicity in vitro and in situ; and the regulatory role of diabetic factors, such as lipids and cholesterol, in these processes are also discussed.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Islet Amyloid Polypeptide/metabolism , Pancreas/pathology , Protein Aggregation, Pathological/pathology , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Islet Amyloid Polypeptide/analysis , Pancreas/metabolism , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Protein Conformation , Protein Folding , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology
5.
FEBS J ; 289(2): 494-506, 2022 01.
Article in English | MEDLINE | ID: mdl-34482629

ABSTRACT

Light-chain (AL) amyloidosis is characterized by deposition of immunoglobulin light chains (LC) as fibrils in target organs. Alongside the full-length protein, abundant LC fragments are always present in AL deposits. Herein, by combining gel-based and mass spectrometry analyses, we identified and compared the fragmentation sites of amyloid LCs from multiple organs of an AL λ amyloidosis patient (AL-55). The positions pinpointed here in kidney and subcutaneous fat, alongside those previously detected in heart of the same patient, were aligned and mapped on the LC's dimeric and fibrillar states. All tissues contain fragmented LCs along with the full-length protein; the fragment pattern is coincident across organs, although microheterogeneity exists. Multiple cleavage positions were detected; some are shared, whereas some are organ-specific, likely due to a complex of proteases. Cleavage sites are concentrated in 'proteolysis-prone' regions, common to all tissues. Several proteolytic sites are not accessible on native dimers, while they are compatible with fibrils. Overall, data suggest that the heterogeneous ensemble of LC fragments originates in tissues and is consistent with digestion of preformed fibrils, or with the hypothesis that initial proteolytic cleavage of the constant domain triggers the amyloidogenic potential of LCs, followed by subsequent proteolytic degradation. This work provides a unique set of molecular data on proteolysis from ex vivo amyloid, which allows discussing hypotheses on role and timing of proteolytic events occurring along amyloid formation and accumulation in AL patients.


Subject(s)
Amyloid Neuropathies/genetics , Amyloid/genetics , Amyloidogenic Proteins/genetics , Amyloidosis/genetics , Immunoglobulin Light Chains/genetics , Amyloid/metabolism , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Amyloidosis/metabolism , Amyloidosis/pathology , Endopeptidases/genetics , Humans , Immunoglobulin Light Chains/metabolism , Kinetics , Peptide Hydrolases/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteolysis , Thermodynamics
6.
Br J Pharmacol ; 179(1): 23-45, 2022 01.
Article in English | MEDLINE | ID: mdl-34528272

ABSTRACT

Lewy bodies that contain aggregated α-synuclein in dopamine neurons are the main culprit for neurodegeneration in Parkinson's disease. However, mitochondrial dysfunction has a well-established and prominent role in the pathogenesis of Parkinson's disease. The exact mechanism by which α-synuclein causes dopamine neuronal loss is unclear. Recent evidence suggests that aggregated α-synuclein localises in the mitochondria contributes to oxidative stress-mediated apoptosis in neurons. Therefore, the involvement of aggregated α-synuclein in mitochondrial dysfunction-mediated neuronal loss has made it an emerging drug target for the treatment of Parkinson's disease. However, the exact mechanism by which α-synuclein permeabilises through the mitochondrial membrane and affects the electron transport chain remains under investigation. In the present study, we describe mitochondria-α-synuclein interactions and how α-synuclein aggregation modulates mitochondrial homeostasis in Parkinson's disease pathogenesis. We also discuss recent therapeutic interventions targeting α-synuclein aggregation that may help researchers to design novel therapeutic treatments for Parkinson's disease.


Subject(s)
Dopaminergic Neurons , Mitochondria , Parkinson Disease , Protein Aggregation, Pathological , alpha-Synuclein , Apoptosis , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Oxidative Stress , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , alpha-Synuclein/metabolism
7.
Cells ; 10(12)2021 12 13.
Article in English | MEDLINE | ID: mdl-34944028

ABSTRACT

A hallmark of Alzheimer's disease (AD) is the accumulation of tau protein in the brain. Compelling evidence indicates that the presence of tau aggregates causes irreversible neuronal destruction, eventually leading to synaptic loss. So far, the inhibition of tau aggregation has been recognized as one of the most effective therapeutic strategies. Cannabidiol (CBD), a major component found in Cannabis sativa L., has antioxidant activities as well as numerous neuroprotective features. Therefore, we hypothesize that CBD may serve as a potent substance to hamper tau aggregation in AD. In this study, we aim to investigate the CBD effect on the aggregation of recombinant human tau protein 1N/4R isoform using biochemical methods in vitro and in silico. Using Thioflavin T (ThT) assay, circular dichroism (CD), and atomic force microscopy (AFM), we demonstrated that CBD can suppress tau fibrils formation. Moreover, by quenching assay, docking, and job's plot, we further demonstrated that one molecule of CBD interacts with one molecule of tau protein through a spontaneous binding. Experiments performed by quenching assay, docking, and Thioflavin T assay further established that the main forces are hydrogen Van der Waals and some non-negligible hydrophobic forces, affecting the lag phase of tau protein kinetics. Taken together, this study provides new insights about a natural substance, CBD, for tau therapy which may offer new hope for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cannabidiol/pharmacology , Neurons/drug effects , Protein Aggregation, Pathological/drug therapy , tau Proteins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Benzothiazoles/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cannabidiol/chemistry , Humans , Kinetics , Microscopy, Atomic Force , Neurons/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Isoforms/drug effects , Protein Isoforms/genetics , tau Proteins/antagonists & inhibitors , tau Proteins/ultrastructure
8.
Int J Mol Sci ; 22(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34948096

ABSTRACT

Gerstmann-Sträussler-Scheinker syndrome (GSS) is a hereditary neurodegenerative disease characterized by extracellular aggregations of pathological prion protein (PrP) forming characteristic plaques. Our study aimed to evaluate the micromorphology and protein composition of these plaques in relation to age, disease duration, and co-expression of other pathogenic proteins related to other neurodegenerations. Hippocampal regions of nine clinically, neuropathologically, and genetically confirmed GSS subjects were investigated using immunohistochemistry and multichannel confocal fluorescent microscopy. Most pathognomic prion protein plaques were small (2-10 µm), condensed, globous, and did not contain any of the other investigated proteinaceous components, particularly dystrophic neurites. Equally rare (in two cases out of nine) were plaques over 50 µm having predominantly fibrillar structure and exhibit the presence of dystrophic neuritic structures; in one case, the plaques also included bulbous dystrophic neurites. Co-expression with hyperphosphorylated protein tau protein or amyloid beta-peptide (Aß) in GSS PrP plaques is generally a rare observation, even in cases with comorbid neuropathology. The dominant picture of the GSS brain is small, condensed plaques, often multicentric, while presence of dystrophic neuritic changes accumulating hyperphosphorylated protein tau or Aß in the PrP plaques are rare and, thus, their presence probably constitutes a trivial observation without any relationship to GSS development and progression.


Subject(s)
Gerstmann-Straussler-Scheinker Disease , Mutation, Missense , Prion Proteins , Protein Aggregation, Pathological , Adult , Aged , Female , Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Male , Middle Aged , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology
9.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884786

ABSTRACT

The aggregation of α-synuclein is the hallmark of a collective of neurodegenerative disorders known as synucleinopathies. The tendency to aggregate of this protein, the toxicity of its aggregation intermediates and the ability of the cellular protein quality control system to clear these intermediates seems to be regulated, among other factors, by post-translational modifications (PTMs). Among these modifications, we consider herein proteolysis at both the N- and C-terminal regions of α-synuclein as a factor that could modulate disassembly of toxic amyloids by the human disaggregase, a combination of the chaperones Hsc70, DnaJB1 and Apg2. We find that, in contrast to aggregates of the protein lacking the N-terminus, which can be solubilized as efficiently as those of the WT protein, the deletion of the C-terminal domain, either in a recombinant context or as a consequence of calpain treatment, impaired Hsc70-mediated amyloid disassembly. Progressive removal of the negative charges at the C-terminal region induces lateral association of fibrils and type B* oligomers, precluding chaperone action. We propose that truncation-driven aggregate clumping impairs the mechanical action of chaperones, which includes fast protofilament unzipping coupled to depolymerization. Inhibition of the chaperone-mediated clearance of C-truncated species could explain their exacerbated toxicity and higher propensity to deposit found in vivo.


Subject(s)
Amyloid/metabolism , HSP70 Heat-Shock Proteins/metabolism , Protein Aggregation, Pathological/pathology , Synucleinopathies/pathology , alpha-Synuclein/metabolism , Calpain/pharmacology , HSC70 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/metabolism , Protein Aggregates/physiology , Protein Processing, Post-Translational/physiology , Proteolysis
10.
PLoS One ; 16(11): e0260519, 2021.
Article in English | MEDLINE | ID: mdl-34847196

ABSTRACT

Endoplasmic reticulum (ER) stress is associated with acute kidney injury (AKI) caused by various mechanisms, including antibiotics, non-steroidal anti-inflammatory drugs, cisplatin, and radiocontrast. Tunicamycin (TM) is a nucleoside antibiotic that induces ER stress and is a commonly used model of AKI. 4-phenylbutyrate (4-PBA) is a chemical chaperone and histone deacetylase (HDAC) inhibitor and has been shown to protect the kidney from ER stress, apoptosis, and structural damage in a tunicamycin model of AKI. The renal protection provided by 4-PBA is attributed to its ability to prevent misfolded protein aggregation and inhibit ER stress; however, the HDAC inhibitor effects of 4-PBA have not been examined in the TM-induced model of AKI. As such, the main objective of this study was to determine if histone hyperacetylation provides any protective effects against TM-mediated AKI. The FDA-approved HDAC inhibitor vorinostat was used, as it has no ER stress inhibitory effects and therefore the histone hyperacetylation properties alone could be investigated. In vitro work demonstrated that vorinostat inhibited histone deacetylation in cultured proximal tubular cells but did not prevent ER stress or protein aggregation induced by TM. Vorinostat induced a significant increase in cell death, and exacerbated TM-mediated total cell death and apoptotic cell death. Wild type male mice were treated with TM (0.5 mg/kg, intraperitoneal injection), with or without vorinostat (50 mg/kg/day) or 4-PBA (1 g/kg/day). Mice treated with 4-PBA or vorinostat exhibited similar levels of histone hyperacetylation. Expression of the pro-apoptotic protein CHOP was induced with TM, and not inhibited by vorinostat. Further, vorinostat did not prevent any renal damage or decline in renal function caused by tunicamycin. These data suggest that the protective mechanisms found by 4-PBA are primarily due to its molecular chaperone properties, and the HDAC inhibitors used did not provide any protection against renal injury caused by ER stress.


Subject(s)
Acute Kidney Injury , Endoplasmic Reticulum Stress/drug effects , Histone Deacetylase Inhibitors/pharmacology , Tunicamycin/adverse effects , Vorinostat/pharmacology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Animals , Cell Line , Disease Models, Animal , Male , Mice , Protein Aggregation, Pathological/chemically induced , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/prevention & control , Tunicamycin/pharmacology
11.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34830074

ABSTRACT

Amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease and similar neurodegenerative disorders take their toll on patients, caregivers and society. A common denominator for these disorders is the accumulation of aggregated proteins in nerve cells, yet the triggers for these aggregation processes are currently unknown. In ALS, protein aggregation has been described for the SOD1, C9orf72, FUS and TDP-43 proteins. The latter is a nuclear protein normally binding to both DNA and RNA, contributing to gene expression and mRNA life cycle regulation. TDP-43 seems to have a specific role in ALS pathogenesis, and ubiquitinated and hyperphosphorylated cytoplasmic inclusions of aggregated TDP-43 are present in nerve cells in almost all sporadic ALS cases. ALS pathology appears to include metal imbalances, and environmental metal exposure is a known risk factor in ALS. However, studies on metal-to-TDP-43 interactions are scarce, even though this protein seems to have the capacity to bind to metals. This review discusses the possible role of metals in TDP-43 aggregation, with respect to ALS pathology.


Subject(s)
Amyotrophic Lateral Sclerosis , Environmental Exposure/adverse effects , Metals , Mutation , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Metals/metabolism , Metals/toxicity , Protein Aggregation, Pathological/etiology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
12.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830391

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder. An important hallmark of PD involves the pathological aggregation of proteins in structures known as Lewy bodies. The major component of these proteinaceous inclusions is alpha (α)-synuclein. In different conditions, α-synuclein can assume conformations rich in either α-helix or ß-sheets. The mechanisms of α-synuclein misfolding, aggregation, and fibrillation remain unknown, but it is thought that ß-sheet conformation of α-synuclein is responsible for its associated toxic mechanisms. To gain fundamental insights into the process of α-synuclein misfolding and aggregation, the secondary structure of this protein in the presence of charged and non-charged surfactant solutions was characterized. The selected surfactants were (anionic) sodium dodecyl sulphate (SDS), (cationic) cetyltrimethylammonium chloride (CTAC), and (uncharged) octyl ß-D-glucopyranoside (OG). The effect of surfactants in α-synuclein misfolding was assessed by ultra-structural analyses, in vitro aggregation assays, and secondary structure analyses. The α-synuclein aggregation in the presence of negatively charged SDS suggests that SDS-monomer complexes stimulate the aggregation process. A reduction in the electrostatic repulsion between N- and C-terminal and in the hydrophobic interactions between the NAC (non-amyloid beta component) region and the C-terminal seems to be important to undergo aggregation. Fourier transform infrared spectroscopy (FTIR) measurements show that ß-sheet structures comprise the assembly of the fibrils.


Subject(s)
Neurodegenerative Diseases/drug therapy , Parkinson Disease/drug therapy , Protein Aggregation, Pathological/drug therapy , alpha-Synuclein/genetics , Amyloid/antagonists & inhibitors , Amyloid/genetics , Cetrimonium/pharmacology , Circular Dichroism , Galactosides/pharmacology , Humans , Lewy Bodies/drug effects , Lewy Bodies/ultrastructure , Neurodegenerative Diseases/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Conformation , Protein Conformation, beta-Strand/genetics , Protein Folding/drug effects , Protein Structure, Secondary/drug effects , Sodium Dodecyl Sulfate/pharmacology , Spectroscopy, Fourier Transform Infrared , alpha-Synuclein/antagonists & inhibitors
13.
PLoS Genet ; 17(11): e1009921, 2021 11.
Article in English | MEDLINE | ID: mdl-34788284

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by alpha-synuclein (αSyn) aggregation and associated with abnormalities in lipid metabolism. The accumulation of lipids in cytoplasmic organelles called lipid droplets (LDs) was observed in cellular models of PD. To investigate the pathophysiological consequences of interactions between αSyn and proteins that regulate the homeostasis of LDs, we used a transgenic Drosophila model of PD, in which human αSyn is specifically expressed in photoreceptor neurons. We first found that overexpression of the LD-coating proteins Perilipin 1 or 2 (dPlin1/2), which limit the access of lipases to LDs, markedly increased triacylglyclerol (TG) loaded LDs in neurons. However, dPlin-induced-LDs in neurons are independent of lipid anabolic (diacylglycerol acyltransferase 1/midway, fatty acid transport protein/dFatp) and catabolic (brummer TG lipase) enzymes, indicating that alternative mechanisms regulate neuronal LD homeostasis. Interestingly, the accumulation of LDs induced by various LD proteins (dPlin1, dPlin2, CG7900 or KlarsichtLD-BD) was synergistically amplified by the co-expression of αSyn, which localized to LDs in both Drosophila photoreceptor neurons and in human neuroblastoma cells. Finally, the accumulation of LDs increased the resistance of αSyn to proteolytic digestion, a characteristic of αSyn aggregation in human neurons. We propose that αSyn cooperates with LD proteins to inhibit lipolysis and that binding of αSyn to LDs contributes to the pathogenic misfolding and aggregation of αSyn in neurons.


Subject(s)
Lipid Metabolism/genetics , Neurons/metabolism , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Animals, Genetically Modified/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Humans , Lipid Droplets/metabolism , Lipolysis/genetics , Membrane Transport Proteins/genetics , Neuroblastoma/genetics , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Perilipin-2/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Proteolysis
14.
PLoS Genet ; 17(11): e1009911, 2021 11.
Article in English | MEDLINE | ID: mdl-34780474

ABSTRACT

The capacity of a cell to maintain proteostasis progressively declines during aging. Virtually all age-associated neurodegenerative disorders associated with aggregation of neurotoxic proteins are linked to defects in the cellular proteostasis network, including insufficient lysosomal hydrolysis. Here, we report that proteotoxicity in yeast and Drosophila models for Parkinson's disease can be prevented by increasing the bioavailability of Ca2+, which adjusts intracellular Ca2+ handling and boosts lysosomal proteolysis. Heterologous expression of human α-synuclein (αSyn), a protein critically linked to Parkinson's disease, selectively increases total cellular Ca2+ content, while the levels of manganese and iron remain unchanged. Disrupted Ca2+ homeostasis results in inhibition of the lysosomal protease cathepsin D and triggers premature cellular and organismal death. External administration of Ca2+ reduces αSyn oligomerization, stimulates cathepsin D activity and in consequence restores survival, which critically depends on the Ca2+/calmodulin-dependent phosphatase calcineurin. In flies, increasing the availability of Ca2+ discloses a neuroprotective role of αSyn upon manganese overload. In sum, we establish a molecular interplay between cathepsin D and calcineurin that can be activated by Ca2+ administration to counteract αSyn proteotoxicity.


Subject(s)
Calcineurin/genetics , Cathepsin D/genetics , Parkinson Disease/genetics , alpha-Synuclein/genetics , Aging/drug effects , Aging/genetics , Animals , Animals, Genetically Modified/genetics , Calcium/metabolism , Calcium/pharmacology , Cell Death/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation/drug effects , Humans , Lysosomes/drug effects , Lysosomes/genetics , Neurons/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Proteolysis/drug effects , Saccharomyces cerevisiae/genetics
15.
Biomolecules ; 11(10)2021 10 06.
Article in English | MEDLINE | ID: mdl-34680099

ABSTRACT

Glycation of α-synuclein (αSyn), as occurs with aging, has been linked to the progression of Parkinson's disease (PD) through the promotion of advanced glycation end-products and the formation of toxic oligomers that cannot be properly cleared from neurons. DJ-1, an antioxidative protein that plays a critical role in PD pathology, has been proposed to repair glycation in proteins, yet a mechanism has not been elucidated. In this study, we integrate solution nuclear magnetic resonance (NMR) spectroscopy and liquid atomic force microscopy (AFM) techniques to characterize glycated N-terminally acetylated-αSyn (glyc-ac-αSyn) and its interaction with DJ-1. Glycation of ac-αSyn by methylglyoxal increases oligomer formation, as visualized by AFM in solution, resulting in decreased dynamics of the monomer amide backbone around the Lys residues, as measured using NMR. Upon addition of DJ-1, this NMR signature of glyc-ac-αSyn monomers reverts to a native ac-αSyn-like character. This phenomenon is reversible upon removal of DJ-1 from the solution. Using relaxation-based NMR, we have identified the binding site on DJ-1 for glycated and native ac-αSyn as the catalytic pocket and established that the oxidation state of the catalytic cysteine is imperative for binding. Based on our results, we propose a novel mechanism by which DJ-1 scavenges glyc-ac-αSyn oligomers without chemical deglycation, suppresses glyc-ac-αSyn monomer-oligomer interactions, and releases free glyc-ac-αSyn monomers in solution. The interference of DJ-1 with ac-αSyn oligomers may promote free ac-αSyn monomer in solution and suppress the propagation of toxic oligomer and fibril species. These results expand the understanding of the role of DJ-1 in PD pathology by acting as a scavenger for aggregated αSyn.


Subject(s)
Glycation End Products, Advanced/genetics , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , alpha-Synuclein/genetics , Acetylation , Cysteine/metabolism , Humans , Magnetic Resonance Spectroscopy , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Multimerization/genetics
16.
Nat Commun ; 12(1): 5739, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667166

ABSTRACT

Protein aggregates associated with neurodegenerative diseases have the ability to transmit to unaffected cells, thereby templating their own aberrant conformation onto soluble homotypic proteins. Proteopathic seeds can be released into the extracellular space, secreted in association with extracellular vesicles (EV) or exchanged by direct cell-to-cell contact. The extent to which each of these pathways contribute to the prion-like spreading of protein misfolding is unclear. Exchange of cellular cargo by both direct cell contact or via EV depends on receptor-ligand interactions. We hypothesized that enabling these interactions through viral ligands enhances intercellular proteopathic seed transmission. Using different cellular models propagating prions or pathogenic Tau aggregates, we demonstrate that vesicular stomatitis virus glycoprotein and SARS-CoV-2 spike S increase aggregate induction by cell contact or ligand-decorated EV. Thus, receptor-ligand interactions are important determinants of intercellular aggregate dissemination. Our data raise the possibility that viral infections contribute to proteopathic seed spreading by facilitating intercellular cargo transfer.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Extracellular Vesicles/metabolism , Membrane Glycoproteins/metabolism , Protein Aggregation, Pathological/virology , Spike Glycoprotein, Coronavirus/metabolism , Viral Envelope Proteins/metabolism , Adult , Aged , Brain/pathology , Case-Control Studies , Cell Line , Endocytosis , Female , Humans , Intravital Microscopy , Male , Middle Aged , Prions/metabolism , Protein Aggregation, Pathological/pathology , Protein Folding , tau Proteins/metabolism
17.
Cell Mol Life Sci ; 78(23): 7185-7198, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34643743

ABSTRACT

A review of the multidisciplinary scientific literature reveals a large variety of amyloid-ß (Aß) oligomeric species, differing in molecular weight, conformation and morphology. These species, which may assemble via either on- or off-aggregation pathways, exhibit differences in stability, function and neurotoxicity, according to different experimental settings. The conformations of the different Aß species are stabilized by intra- and inter-molecular hydrogen bonds and by electrostatic and hydrophobic interactions, all depending on the chemical and physical environment (e.g., solvent, ions, pH) and interactions with other molecules, such as lipids and proteins. This complexity and the lack of a complete understanding of the relationship between the different Aß species and their toxicity is currently dictating the nature of the inhibitor (or inducer)-based approaches that are under development for interfering with (or inducing) the formation of specific species and Aß oligomerization, and for interfering with the associated downstream neurotoxic effects. Here, we review the principles that underlie the involvement of different Aß oligomeric species in neurodegeneration, both in vitro and in preclinical studies. In addition, we provide an overview of the existing inhibitors (or inducers) of Aß oligomerization that serve as potential therapeutics for neurodegenerative diseases. The review, which covers the exciting studies that have been published in the past few years, comprises three main parts: 1) on- and off-fibrillar assembly mechanisms and Aß structural polymorphism; 2) interactions of Aß with other molecules and cell components that dictate the Aß aggregation pathway; and 3) targeting the on-fibrillar Aß assembly pathway as a therapeutic approach.


Subject(s)
Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid/chemistry , Neurodegenerative Diseases/pathology , Peptide Fragments/metabolism , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , Peptide Fragments/genetics , Protein Aggregation, Pathological/pathology , Protein Conformation
18.
Sci Rep ; 11(1): 19262, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34584131

ABSTRACT

Amyloid ß (Aß) peptide aggregation plays a central role in Alzheimer's disease (AD) etiology. AD drug candidates have included small molecules or peptides directed towards inhibition of Aß fibrillogenesis. Although some Aß-derived peptide fragments suppress Aß fibril growth, comprehensive analysis of inhibitory potencies of peptide fragments along the whole Aß sequence has not been reported. The aim of this work is (a) to identify the region(s) of Aß with highest propensities for aggregation and (b) to use those fragments to inhibit Aß fibrillogenesis. Structural and aggregation properties of the parent Aß1-42 peptide and seven overlapping peptide fragments have been studied, i.e. Aß1-10 (P1), Aß6-15 (P2), Aß11-20 (P3), Aß16-25 (P4), Aß21-30 (P5), Aß26-36 (P6), and Aß31-42 (P7). Structural transitions of the peptides in aqueous buffer have been monitored by circular dichroism and Fourier transform infrared spectroscopy. Aggregation and fibrillogenesis were analyzed by light scattering and thioflavin-T fluorescence. The mode of peptide-peptide interactions was characterized by fluorescence resonance energy transfer. Three peptide fragments, P3, P6, and P7, exhibited exceptionally high propensity for ß-sheet formation and aggregation. Remarkably, only P3 and P6 exerted strong inhibitory effect on the aggregation of Aß1-42, whereas P7 and P2 displayed moderate inhibitory potency. It is proposed that P3 and P6 intercalate between Aß1-42 molecules and thereby inhibit Aß1-42 aggregation. These findings may facilitate therapeutic strategies of inhibition of Aß fibrillogenesis by Aß-derived peptides.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Peptide Fragments/pharmacology , Protein Aggregation, Pathological/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Circular Dichroism , Humans , Peptide Fragments/therapeutic use , Peptide Fragments/ultrastructure , Protein Aggregation, Pathological/pathology
19.
Neurobiol Dis ; 160: 105515, 2021 12.
Article in English | MEDLINE | ID: mdl-34571136

ABSTRACT

Brain inclusions mainly composed of misfolded and aggregated TAR DNA binding protein 43 (TDP-43), are characteristic hallmarks of amyotrophic lateral sclerosis (ALS). Irrespective of the role played by the inclusions, their reduction represents an important therapeutic pathway that is worth exploring. Their removal can either lead to the recovery of TDP-43 function by removing the self-templating conformers that sequester the protein in the inclusions, and/or eliminate any potential intrinsic toxicity of the aggregates. The search for curative therapies has been hampered by the lack of ALS models for use in high-throughput screening. We adapted, optimised, and extensively characterised our previous ALS cellular model for such use. The model demonstrated efficient aggregation of endogenous TDP-43, and concomitant loss of its splicing regulation function. We provided a proof-of-principle for its eventual use in high-throughput screening using compounds of the tricyclic family and showed that recovery of TDP-43 function can be achieved by the enhanced removal of TDP-43 aggregates by these compounds. We observed that the degradation of the aggregates occurs independent of the autophagy pathway beyond autophagosome-lysosome fusion, but requires a functional proteasome pathway. The in vivo translational effect of the cellular model was tested with two of these compounds in a Drosophila model expressing a construct analogous to the cellular model, where thioridazine significantly improved the locomotive defect. Our findings have important implications as thioridazine cleared TDP-43 aggregates and recovered TDP-43 functionality. This study also highlights the importance of a two-stage, in vitro and in vivo model system to cross-check the search for small molecules that can clear TDP-43 aggregates in TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , DNA-Binding Proteins/metabolism , Dopamine Antagonists/therapeutic use , Drosophila Proteins/metabolism , Protein Aggregation, Pathological/drug therapy , Thioridazine/therapeutic use , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy/drug effects , Cell Line , Disease Models, Animal , Dopamine Antagonists/pharmacology , Drosophila , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Thioridazine/pharmacology
20.
Int J Biol Macromol ; 188: 207-214, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34364939

ABSTRACT

Protein aggregation is suggested as a reversible, wide-spread physiological process used by cells to regulate their growth and adapt to different stress conditions. Nucleophosmin 1(NPM1) protein is an abundant multifunctional nucleolar chaperone and its gene is the most frequently mutated in Acute Myeloid Leukemia (AML) patients. So far, the role of NPM1 mutations in leukemogenesis has remained largely elusive considering that they have the double effect of unfolding the C-terminal domain (CTD) and delocalizing the protein in the cytosol (NPM1c+). This mislocalization heavily impacts on cell cycle regulation. Our recent investigations unequivocally demonstrated an amyloid aggregation propensity introduced by AML mutations. Herein, employing complementary biophysical assays, we have characterized a N-terminal extended version of type F AML mutation of CTD and proved that it is able to form assemblies with amyloid character and fibrillar morphology. The present study represents an additional phase of knowledge to deepen the roles exerted by different types of cytoplasmatic NPM1c+ forms to develop in the future potential therapeutics for their selective targeting.


Subject(s)
Carcinogenesis/genetics , Leukemia, Myeloid, Acute/genetics , Nucleophosmin/genetics , Protein Aggregation, Pathological/genetics , Amyloidosis/genetics , Amyloidosis/pathology , Cell Line, Tumor , Cytoplasm/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Mutation/genetics , Nuclear Proteins/genetics , Protein Aggregates/genetics , Protein Aggregation, Pathological/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...