Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
Add more filters

Publication year range
1.
Stroke ; 53(1): 298-302, 2022 01.
Article in English | MEDLINE | ID: mdl-34905943

ABSTRACT

We review the implications of the recently approved aducanumab amyloid-ß immunotherapy for treating Alzheimer disease with comorbid cerebral amyloid angiopathy. In clinical trials, amyloid-ß immunotherapy has been associated with a high rate of amyloid-related imaging abnormalities, potentially driven by coexisting cerebral amyloid angiopathy. Therefore, immunotherapy's efficacy in patients may be modified by coexisting cerebrovascular pathology. We discuss the contributions of cerebral amyloid angiopathy on the development of amyloid-related imaging abnormalities and propose strategies to identify cerebral amyloid angiopathy in patients considered for immunotherapy.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid/drug effects , Antibodies, Monoclonal, Humanized/therapeutic use , Cerebral Amyloid Angiopathy/drug therapy , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/complications , Humans , Time Factors
2.
Blood ; 135(21): 1899-1911, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32128578

ABSTRACT

Hemophilia A, an X-linked bleeding disorder caused by deficiency of factor VIII (FVIII), is treated by protein replacement. Unfortunately, this regimen is costly due to the expense of producing recombinant FVIII as a consequence of its low-level secretion from mammalian host cells. FVIII expression activates the endoplasmic reticulum (ER) stress response, causes oxidative stress, and induces apoptosis. Importantly, little is known about the factors that cause protein misfolding and aggregation in metazoans. Here, we identified intrinsic and extrinsic factors that cause FVIII to form aggregates. We show that FVIII forms amyloid-like fibrils within the ER lumen upon increased FVIII synthesis or inhibition of glucose metabolism. Significantly, FVIII amyloids can be dissolved upon restoration of glucose metabolism to produce functional secreted FVIII. Two ER chaperone families and their cochaperones, immunoglobulin binding protein (BiP) and calnexin/calreticulin, promote FVIII solubility in the ER, where the former is also required for disaggregation. A short aggregation motif in the FVIII A1 domain (termed Aggron) is necessary and sufficient to seed ß-sheet polymerization, and BiP binding to this Aggron prevents amyloidogenesis. Our findings provide novel insight into mechanisms that limit FVIII secretion and ER protein aggregation in general and have implication for ongoing hemophilia A gene-therapy clinical trials.


Subject(s)
Amyloid/chemistry , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/drug effects , Factor VIII/metabolism , Glucose/pharmacology , Molecular Chaperones/metabolism , Amyloid/drug effects , Endoplasmic Reticulum/metabolism , Factor VIII/genetics , Hemostatics , Hep G2 Cells , Humans , Molecular Chaperones/genetics , Sweetening Agents/pharmacology
3.
FASEB J ; 35(2): e21318, 2021 02.
Article in English | MEDLINE | ID: mdl-33508158

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects cognition and memory. Recent advances have helped identify many clinical sub-types in AD. Mounting evidence point toward structural polymorphism among fibrillar aggregates of amyloid-ß (Aß) to being responsible for the phenotypes and clinical manifestations. In the emerging paradigm of polymorphism and prion-like propagation of aggregates in AD, the role of low molecular weight soluble oligomers, which are long known to be the primary toxic agents, in effecting phenotypes remains inconspicuous. In this study, we present the characterization of three soluble oligomers of Aß42, namely 14LPOs, 16LPOs, and GM1Os with discreet biophysical and biochemical properties generated using lysophosphatidyl glycerols and GM1 gangliosides. The results indicate that the oligomers share some biophysical similarities but display distinctive differences with GM1Os. Unlike the other two, GM1Os were observed to be complexed with the lipid upon isolation. It also differs mainly in detection by conformation-sensitive dyes and conformation-specific antibodies, temperature and enzymatic stability, and in the ability to propagate morphologically-distinct fibrils. GM1Os also show distinguishable biochemical behavior with pronounced neuronal toxicity. Furthermore, all the oligomers induce cerebral amyloid angiopathy (CAA) and plaque burden in transgenic AD mice, which seems to be a consistent feature among all lipid-derived oligomers, but 16LPOs and GM1Os displayed significantly higher effect than the others. These results establish a correlation between molecular features of Aß42 oligomers and their distinguishable effects in transgenic AD mice attuned by lipid characteristics, and therefore help bridge the knowledge gap in understanding how oligomer conformers could elicit AD phenotypes.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Lipids/pharmacology , Amyloid/drug effects , Animals , Cell Line, Tumor , Cell Survival/physiology , Circular Dichroism , Dynamic Light Scattering , G(M1) Ganglioside/pharmacology , Immunohistochemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Transgenic , Microscopy, Atomic Force , Phosphatidylglycerols/pharmacology , Plaque, Amyloid/metabolism , Spectrometry, Mass, Electrospray Ionization , Spectroscopy, Fourier Transform Infrared
4.
Nature ; 537(7618): 50-6, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27582220

ABSTRACT

Alzheimer's disease (AD) is characterized by deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aß to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aß. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aß, and reduce soluble and insoluble Aß in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aß in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating-Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Brain/drug effects , Brain/metabolism , Clinical Trials, Phase III as Topic , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Models, Biological , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/drug therapy , Solubility
5.
PLoS Pathog ; 15(8): e1007978, 2019 08.
Article in English | MEDLINE | ID: mdl-31469892

ABSTRACT

Curli amyloid fibrils secreted by Enterobacteriaceae mediate host cell adhesion and contribute to biofilm formation, thereby promoting bacterial resistance to environmental stressors. Here, we present crystal structures of amyloid-forming segments from the major curli subunit, CsgA, revealing steric zipper fibrils of tightly mated ß-sheets, demonstrating a structural link between curli and human pathological amyloids. D-enantiomeric peptides, originally developed to interfere with Alzheimer's disease-associated amyloid-ß, inhibited CsgA fibrillation and reduced biofilm formation in Salmonella typhimurium. Moreover, as previously shown, CsgA fibrils cross-seeded fibrillation of amyloid-ß, providing support for the proposed structural resemblance and potential for cross-species amyloid interactions. The presented findings provide structural insights into amyloidogenic regions important for curli formation, suggest a novel strategy for disrupting amyloid-structured biofilms, and hypothesize on the formation of self-propagating prion-like species originating from a microbial source that could influence neurodegenerative diseases.


Subject(s)
Amyloid/antagonists & inhibitors , Biofilms/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Peptide Fragments/pharmacology , Amyloid/drug effects , Biofilms/drug effects , Crystallography, X-Ray , Humans , Microscopy, Electron, Transmission , Peptide Fragments/chemistry , Protein Binding , Salmonella typhimurium/drug effects , Salmonella typhimurium/physiology
6.
Arch Biochem Biophys ; 714: 109077, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34728171

ABSTRACT

Neurodegenerative diseases are a group of debilitating maladies involving protein aggregation. To this day, all advances in neurodegenerative disease therapeutics have helped symptomatically but have not prevented the root cause of the disease, i.e., the aggregation of involved proteins. Antibiotics are becoming increasingly obsolete due to the rising multidrug resistance strains of bacteria. Thus, antibiotics, if put to different use as therapeutics against other diseases, could pave a new direction to the world of antibiotics. Hence, we studied the antibiotic levofloxacin for its potential anti-amyloidogenic behavior using human lysozyme, a protein involved in non-systemic amyloidosis, as a model system. At the sub-stoichiometric level, levofloxacin was able to inhibit amyloid formation in human lysozyme as observed by various spectroscopic and microscopic methods, with IC50 values as low as 8.8 ± 0.1 µM. Levofloxacin also displayed a retarding effect on seeding phenomena by elongating the lag-phase (from 0 to 88 h) at lower concentration, and arresting lysozyme fibrillation at the lag stage in sub-stoichiometric concentrations. Structural and computational analyses provided mechanistic insight showing that levofloxacin stabilizes the lysozyme in the native state by binding to the aggregation-prone residues, and thereby inhibiting amyloid fibrillation. Levofloxacin also showed the property of disrupting amyloid fibrils into a smaller polymeric form of proteins which were less cytotoxic as confirmed by hemolytic assay. Therefore, we throw new light on levofloxacin as an amyloid inhibitor and disruptor which could pave way to utilization of levofloxacin as a potential therapeutic against non-systemic amyloidosis and neurodegenerative diseases.


Subject(s)
Amyloid/drug effects , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Levofloxacin/pharmacology , Amyloid/biosynthesis , Circular Dichroism , Drug Resistance, Multiple, Bacterial/genetics , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Point Mutation , Spectrometry, Fluorescence
7.
J Cardiovasc Pharmacol ; 78(5): e641-e647, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34321398

ABSTRACT

ABSTRACT: The transthyretin (TTR) amyloidoses result from misfolding of the protein leading to fibril formation and aggregation as amyloid deposits in predominantly the cardiovascular and nervous systems. Cardiac involvement can manifest as heart failure, arrhythmias, and valvular disease. Neurologic involvement can cause sensorimotor polyneuropathies, mononeuropathies, and dysautonomia. Previously, treatment has focused on management of these symptoms and disease sequelae, with a high rate of mortality due to the absence of disease-modifying therapies. In this article, we review novel treatments focusing on 3 mechanistic pathways: (1) silencing of the TTR gene to suppress production, (2) stabilizing of TTR tetramers to prevent misfolding, or (3) disrupting of existing TTR amyloid fibrils to promote reabsorption.


Subject(s)
Amyloid Neuropathies, Familial/therapy , Amyloid/drug effects , Cardiomyopathies/therapy , Cardiovascular Agents/therapeutic use , Genetic Therapy , Myocytes, Cardiac/drug effects , Prealbumin/genetics , Amyloid/metabolism , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/metabolism , Amyloid Neuropathies, Familial/pathology , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiovascular Agents/adverse effects , Gene Silencing , Genetic Predisposition to Disease , Humans , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Prealbumin/metabolism , Protein Stability
8.
Nature ; 523(7562): 607-11, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26200341

ABSTRACT

The human lens is comprised largely of crystallin proteins assembled into a highly ordered, interactive macro-structure essential for lens transparency and refractive index. Any disruption of intra- or inter-protein interactions will alter this delicate structure, exposing hydrophobic surfaces, with consequent protein aggregation and cataract formation. Cataracts are the most common cause of blindness worldwide, affecting tens of millions of people, and currently the only treatment is surgical removal of cataractous lenses. The precise mechanisms by which lens proteins both prevent aggregation and maintain lens transparency are largely unknown. Lanosterol is an amphipathic molecule enriched in the lens. It is synthesized by lanosterol synthase (LSS) in a key cyclization reaction of a cholesterol synthesis pathway. Here we identify two distinct homozygous LSS missense mutations (W581R and G588S) in two families with extensive congenital cataracts. Both of these mutations affect highly conserved amino acid residues and impair key catalytic functions of LSS. Engineered expression of wild-type, but not mutant, LSS prevents intracellular protein aggregation of various cataract-causing mutant crystallins. Treatment by lanosterol, but not cholesterol, significantly decreased preformed protein aggregates both in vitro and in cell-transfection experiments. We further show that lanosterol treatment could reduce cataract severity and increase transparency in dissected rabbit cataractous lenses in vitro and cataract severity in vivo in dogs. Our study identifies lanosterol as a key molecule in the prevention of lens protein aggregation and points to a novel strategy for cataract prevention and treatment.


Subject(s)
Cataract/drug therapy , Cataract/metabolism , Lanosterol/pharmacology , Lanosterol/therapeutic use , Protein Aggregates/drug effects , Protein Aggregation, Pathological/drug therapy , Adult , Amino Acid Sequence , Amyloid/chemistry , Amyloid/drug effects , Amyloid/metabolism , Amyloid/ultrastructure , Animals , Base Sequence , Cataract/congenital , Cataract/genetics , Cataract/pathology , Cell Line , Child , Crystallins/chemistry , Crystallins/genetics , Crystallins/metabolism , Crystallins/ultrastructure , Dogs , Female , Humans , Lanosterol/administration & dosage , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Male , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutant Proteins/ultrastructure , Pedigree , Protein Aggregation, Pathological/pathology
9.
Biochem J ; 477(3): 645-670, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31939603

ABSTRACT

The aggregation of the protein alpha synuclein (α-Syn), a known contributor in Parkinson's disease (PD) pathogenesis is triggered by transition metal ions through occupational exposure and disrupted metal ion homeostasis. Naturally occurring small molecules such as polyphenols have emerged as promising inhibitors of α-Syn fibrillation and toxicity and could be potential therapeutic agents against PD. Here, using an array of biophysical tools combined with cellular assays, we demonstrate that the novel polyphenolic compound scutellarin efficiently inhibits the uninduced and metal-induced fibrillation of α-Syn by acting at the nucleation stage and stabilizes a partially folded intermediate of α-Syn to form SDS-resistant, higher-order oligomers (∼680 kDa) and also disaggregates preformed fibrils of α-Syn into similar type of higher-order oligomers. ANS binding assay, fluorescence lifetime measurements and cell-toxicity experiments reveal scutellarin-generated oligomers as compact, low hydrophobicity structures with modulated surface properties and significantly reduced cytotoxicity than the fibrillation intermediates of α-Syn control. Fluorescence spectroscopy and isothermal titration calorimetry establish the binding between scutellarin and α-Syn to be non-covalent in nature and of moderate affinity (Ka ∼ 105 M-1). Molecular docking approaches suggest binding of scutellarin to the residues present in the NAC region and C-terminus of monomeric α-Syn and the C-terminal residues of fibrillar α-Syn, demonstrating inhibition of fibrillation upon binding to these residues and possible stabilization of the autoinhibitory conformation of α-Syn. These findings reveal interesting insights into the mechanism of scutellarin action and establish it as an efficient modulator of uninduced as well as metal-induced α-Syn fibrillation and toxicity.


Subject(s)
Apigenin/pharmacology , Glucuronates/pharmacology , Parkinson Disease , Protein Aggregates/drug effects , Protein Aggregation, Pathological/drug therapy , alpha-Synuclein/metabolism , Amyloid/drug effects , Amyloid/metabolism , Humans , Molecular Docking Simulation , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/drug effects
10.
Proc Natl Acad Sci U S A ; 115(41): 10481-10486, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30249646

ABSTRACT

Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons, a process that current therapeutic approaches cannot prevent. In PD, the typical pathological hallmark is the accumulation of intracellular protein inclusions, known as Lewy bodies and Lewy neurites, which are mainly composed of α-synuclein. Here, we exploited a high-throughput screening methodology to identify a small molecule (SynuClean-D) able to inhibit α-synuclein aggregation. SynuClean-D significantly reduces the in vitro aggregation of wild-type α-synuclein and the familiar A30P and H50Q variants in a substoichiometric molar ratio. This compound prevents fibril propagation in protein-misfolding cyclic amplification assays and decreases the number of α-synuclein inclusions in human neuroglioma cells. Computational analysis suggests that SynuClean-D can bind to cavities in mature α-synuclein fibrils and, indeed, it displays a strong fibril disaggregation activity. The treatment with SynuClean-D of two PD Caenorhabditis elegans models, expressing α-synuclein either in muscle or in dopaminergic neurons, significantly reduces the toxicity exerted by α-synuclein. SynuClean-D-treated worms show decreased α-synuclein aggregation in muscle and a concomitant motility recovery. More importantly, this compound is able to rescue dopaminergic neurons from α-synuclein-induced degeneration. Overall, SynuClean-D appears to be a promising molecule for therapeutic intervention in Parkinson's disease.


Subject(s)
Amyloid/drug effects , Caenorhabditis elegans/drug effects , Dopaminergic Neurons/drug effects , Parkinson Disease/drug therapy , Protein Aggregation, Pathological/drug therapy , Small Molecule Libraries/pharmacology , alpha-Synuclein/antagonists & inhibitors , Amyloid/metabolism , Animals , Caenorhabditis elegans/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , High-Throughput Screening Assays , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Tumor Cells, Cultured , alpha-Synuclein/metabolism
11.
Int J Mol Sci ; 22(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34884584

ABSTRACT

Significant research on Alzheimer's disease (AD) has demonstrated that amyloid ß (Aß) oligomers are toxic molecules against neural cells. Thus, determining the generation mechanism of toxic Aß oligomers is crucial for understanding AD pathogenesis. Aß fibrils were reported to be disaggregated by treatment with small compounds, such as epigallocatechin gallate (EGCG) and dopamine (DA), and a loss of fibril shape and decrease in cytotoxicity were observed. However, the characteristics of intermediate products during the fibril disaggregation process are poorly understood. In this study, we found that cytotoxic Aß aggregates are generated during a moderate disaggregation process of Aß fibrils. A cytotoxicity assay revealed that Aß fibrils incubated with a low concentration of EGCG and DA showed higher cytotoxicity than Aß fibrils alone. Atomic force microscopy imaging and circular dichroism spectrometry showed that short and narrow protofilaments, which were highly stable in the ß-sheet structure, were abundant in these moderately disaggregated samples. These results indicate that toxic Aß protofilaments are generated during disaggregation from amyloid fibrils, suggesting that disaggregation of Aß fibrils by small compounds may be one of the possible mechanisms for the generation of toxic Aß aggregates in the brain.


Subject(s)
Amyloid beta-Peptides/pharmacology , Amyloid/chemistry , Catechin/analogs & derivatives , Dopamine/pharmacology , Neuroblastoma/pathology , Amyloid/drug effects , Antioxidants/pharmacology , Catechin/pharmacology , Dopamine Agents/pharmacology , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Tumor Cells, Cultured
12.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923726

ABSTRACT

Alzheimer's disease (AD) is a complex multifactorial disorder, mainly characterized by the progressive loss of memory and cognitive, motor, and functional capacity. The absence of effective therapies available for AD alongside the consecutive failures in the central nervous system (CNS) drug development has been motivating the search for new disease-modifying therapeutic strategies for this disease. To address this issue, the multitarget directed ligands (MTDLs) are emerging as a therapeutic alternative to target the multiple AD-related factors. Following this concept, herein we describe the design, synthesis, and biological evaluation of a family of chromeno[3,4-b]xanthones as well as their (E)-2-[2-(propargyloxy)styryl]chromone precursors, as first-in-class acetylcholinesterase (AChE) and ß-amyloid (Aß) aggregation dual-inhibitors. Compounds 4b and 10 emerged as well-balanced dual-target inhibitors, with IC50 values of 3.9 and 2.9 µM for AChE and inhibitory percentages of 70 and 66% for Aß aggregation, respectively. The molecular docking showed that most of the compounds bound to AChE through hydrogen bonds with residues of the catalytic triad and π-stacking interactions between the main scaffold and the aromatic residues present in the binding pocket. The interesting well-balanced activities of these compounds makes them interesting templates for the development of new multitarget compounds for AD.


Subject(s)
Amyloid/drug effects , Cholinesterase Inhibitors/chemical synthesis , Neuroprotective Agents/chemical synthesis , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Amyloid/chemistry , Amyloid/metabolism , Binding Sites , Cholinesterase Inhibitors/pharmacology , Chromones/chemistry , Humans , Neuroprotective Agents/pharmacology , Protein Binding , Protein Multimerization , Xanthones/chemistry
13.
Int J Mol Sci ; 22(17)2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34502079

ABSTRACT

The formation of amyloid-like structures by metabolites is associated with several inborn errors of metabolism (IEMs). These structures display most of the biological, chemical and physical properties of protein amyloids. However, the molecular interactions underlying the assembly remain elusive, and so far, no modulating therapeutic agents are available for clinical use. Chemical chaperones are known to inhibit protein and peptide amyloid formation and stabilize misfolded enzymes. Here, we provide an in-depth characterization of the inhibitory effect of osmolytes and hydrophobic chemical chaperones on metabolite assemblies, thus extending their functional repertoire. We applied a combined in vivo-in vitro-in silico approach and show their ability to inhibit metabolite amyloid-induced toxicity and reduce cellular amyloid content in yeast. We further used various biophysical techniques demonstrating direct inhibition of adenine self-assembly and alteration of fibril morphology by chemical chaperones. Using a scaffold-based approach, we analyzed the physiochemical properties of various dimethyl sulfoxide derivatives and their role in inhibiting metabolite self-assembly. Lastly, we employed whole-atom molecular dynamics simulations to elucidate the role of hydrogen bonds in osmolyte inhibition. Our results imply a dual mode of action of chemical chaperones as IEMs therapeutics, that could be implemented in the rational design of novel lead-like molecules.


Subject(s)
Amyloid/drug effects , Dimethyl Sulfoxide/pharmacology , Adenine/chemistry , Adenine/metabolism , Amyloid/chemistry , Amyloid/metabolism , Dimethyl Sulfoxide/analogs & derivatives , Molecular Dynamics Simulation , Polymerization/drug effects , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
14.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768913

ABSTRACT

Aggregation of α-synuclein (α-Syn) is implicated in the pathogenesis of Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Therefore, the removal of α-Syn aggregation could lead to the development of many new therapeutic agents for neurodegenerative diseases. In the present study, we succeeded in generating a new α-Syn stably expressing cell line using a piggyBac transposon system to investigate the neuroprotective effect of the flavonoid kaempferol on α-Syn toxicity. We found that kaempferol provided significant protection against α-Syn-related neurotoxicity. Furthermore, kaempferol induced autophagy through an increase in the biogenesis of lysosomes by inducing the expression of transcription factor EB and reducing the accumulation of α-Syn; thus, kaempferol prevented neuronal cell death. Moreover, kaempferol directly blocked the amyloid fibril formation of α-Syn. These results support the therapeutic potential of kaempferol in diseases such as synucleinopathies that are characterized by α-Syn aggregates.


Subject(s)
Amyloid/drug effects , Autophagy , Kaempferols/pharmacology , Neuroblastoma/drug therapy , Neurotoxicity Syndromes/drug therapy , Protective Agents/pharmacology , alpha-Synuclein/toxicity , Amyloid/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology
15.
Int J Mol Sci ; 22(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809196

ABSTRACT

Deposition of amyloid ß (Aß) fibrils in the brain is a key pathologic hallmark of Alzheimer's disease. A class of polyphenolic biflavonoids is known to have anti-amyloidogenic effects by inhibiting aggregation of Aß and promoting disaggregation of Aß fibrils. In the present study, we further sought to investigate the structural basis of the Aß disaggregating activity of biflavonoids and their interactions at the atomic level. A thioflavin T (ThT) fluorescence assay revealed that amentoflavone-type biflavonoids promote disaggregation of Aß fibrils with varying potency due to specific structural differences. The computational analysis herein provides the first atomistic details for the mechanism of Aß disaggregation by biflavonoids. Molecular docking analysis showed that biflavonoids preferentially bind to the aromatic-rich, partially ordered N-termini of Aß fibril via the π-π interactions. Moreover, docking scores correlate well with the ThT EC50 values. Molecular dynamic simulations revealed that biflavonoids decrease the content of ß-sheet in Aß fibril in a structure-dependent manner. Hydrogen bond analysis further supported that the substitution of hydroxyl groups capable of hydrogen bond formation at two positions on the biflavonoid scaffold leads to significantly disaggregation of Aß fibrils. Taken together, our data indicate that biflavonoids promote disaggregation of Aß fibrils due to their ability to disrupt the fibril structure, suggesting biflavonoids as a lead class of compounds to develop a therapeutic agent for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Benzothiazoles/pharmacology , Biflavonoids/pharmacology , Protein Aggregation, Pathological/drug therapy , Alzheimer Disease/pathology , Amyloid/antagonists & inhibitors , Amyloid/drug effects , Amyloid/ultrastructure , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/ultrastructure , Biflavonoids/chemistry , Brain/drug effects , Brain/pathology , Humans , Hydrogen Bonding/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology
16.
Int J Mol Sci ; 22(11)2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34199427

ABSTRACT

The interplay between α-synuclein and dopamine derivatives is associated with oxidative stress-dependent neurodegeneration in Parkinson's disease (PD). The formation in the dopaminergic neurons of intraneuronal inclusions containing aggregates of α-synuclein is a typical hallmark of PD. Even though the biochemical events underlying the aberrant aggregation of α-synuclein are not completely understood, strong evidence correlates this process with the levels of dopamine metabolites. In vitro, 3,4-dihydroxyphenylacetaldehyde (DOPAL) and the other two metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and 3,4-dihydroxyphenylethanol (DOPET), share the property to inhibit the growth of mature amyloid fibrils of α-synuclein. Although this effect occurs with the formation of differently toxic products, the molecular basis of this inhibition is still unclear. Here, we provide information on the effect of DOPAC on the aggregation properties of α-synuclein and its ability to interact with membranes. DOPAC inhibits α-synuclein aggregation, stabilizing monomer and inducing the formation of dimers and trimers. DOPAC-induced oligomers did not undergo conformational transition in the presence of membranes, and penetrated the cell, where they triggered autophagic processes. Cellular assays showed that DOPAC reduced cytotoxicity and ROS production induced by α-synuclein aggregates. Our findings show that the early radicals resulting from DOPAC autoxidation produced covalent modifications of the protein, which were not by themselves a primary cause of either fibrillation or membrane binding inhibition. These findings are discussed in the light of the potential mechanism of DOPAC protection against the toxicity of α-synuclein aggregates to better understand protein and catecholamine biology and to eventually suggest a scaffold that can help in the design of candidate molecules able to interfere in α-synuclein aggregation.


Subject(s)
Cell Proliferation/drug effects , Parkinson Disease/genetics , Protein Aggregation, Pathological/genetics , alpha-Synuclein/genetics , 3,4-Dihydroxyphenylacetic Acid/analogs & derivatives , 3,4-Dihydroxyphenylacetic Acid/pharmacology , Amyloid/drug effects , Amyloid/genetics , Dopamine/genetics , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Oxidative Stress/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Protein Aggregation, Pathological/drug therapy , Protein Multimerization/genetics , alpha-Synuclein/antagonists & inhibitors
17.
Biochemistry ; 59(3): 290-296, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31702899

ABSTRACT

Amyloid-ß (Aß) oligomers are well-known toxic molecular species associated with Alzheimer's disease. Recent discoveries of the ability of amyloid fibril surfaces to convert soluble proteins into toxic oligomers suggested that these surfaces could serve as therapeutic targets for intervention. We have shown previously that a short helical peptide could be a key structural motif that can specifically recognize the K16-E22 region of the Aß40 fibril surface with an affinity at the level of several micromolar. Here, we demonstrate that in-tether chiral center-induced helical stabilized peptides could also recognize the fibril surfaces, effectively inhibiting the surface-mediated oligomerization of Aß40. Moreover, through extensive computational sampling, we observed two distinct ways in which the peptide inhibitors recognize the fibril surface. Apart from a binding mode that, in accord with the original design, involves hydrophobic side chains at the binding interface, we observed much more frequently another binding mode in which the hydrophobic staple interacts directly with the fibril surface. The affinity of the peptides for the fibril surface could be adjusted by tuning the hydrophobicity of the staple. The best candidate investigated here exhibits a submicromolar affinity (∼0.75 µM). Collectively, this work opens an avenue for the rational design of candidate drugs with stapled peptides for amyloid-related disease.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid/drug effects , Peptides/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptides/antagonists & inhibitors , Peptides/therapeutic use , Protein Multimerization/drug effects
18.
J Biol Chem ; 294(29): 11259-11275, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31167790

ABSTRACT

Transthyretin (TTR) is a major amyloidogenic protein associated with hereditary (ATTRm) and nonhereditary (ATTRwt) intractable systemic transthyretin amyloidosis. The pathological mechanisms of ATTR-associated amyloid fibril formation are incompletely understood, and there is a need for identifying compounds that target ATTR. C-terminal TTR fragments are often present in amyloid-laden tissues of most patients with ATTR amyloidosis, and on the basis of in vitro studies, these fragments have been proposed to play important roles in amyloid formation. Here, we found that experimentally-formed aggregates of full-length TTR are cleaved into C-terminal fragments, which were also identified in patients' amyloid-laden tissues and in SH-SY5Y neuronal and U87MG glial cells. We observed that a 5-kDa C-terminal fragment of TTR, TTR81-127, is highly amyloidogenic in vitro, even at neutral pH. This fragment formed amyloid deposits and induced apoptosis and inflammatory gene expression also in cultured cells. Using the highly amyloidogenic TTR81-127 fragment, we developed a cell-based high-throughput screening method to discover compounds that disrupt TTR amyloid fibrils. Screening a library of 1280 off-patent drugs, we identified two candidate repositioning drugs, pyrvinium pamoate and apomorphine hydrochloride. Both drugs disrupted patient-derived TTR amyloid fibrils ex vivo, and pyrvinium pamoate also stabilized the tetrameric structure of TTR ex vivo in patient plasma. We conclude that our TTR81-127-based screening method is very useful for discovering therapeutic drugs that directly disrupt amyloid fibrils. We propose that repositioning pyrvinium pamoate and apomorphine hydrochloride as TTR amyloid-disrupting agents may enable evaluation of their clinical utility for managing ATTR amyloidosis.


Subject(s)
Amyloid/metabolism , High-Throughput Screening Assays/methods , Prealbumin/metabolism , Amyloid/drug effects , Amyloid Neuropathies, Familial/metabolism , Apomorphine/pharmacology , Cells, Cultured , Drug Repositioning , Humans , Hydrogen-Ion Concentration , Inflammation/genetics , Neuroglia/metabolism , Neurons/metabolism , Prealbumin/chemistry , Protein Conformation , Proteolysis , Pyrvinium Compounds/pharmacology , Trypsin/metabolism
19.
J Biol Chem ; 294(37): 13740-13754, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31346035

ABSTRACT

Seminal amyloid fibrils are made up of naturally occurring peptide fragments and are key targets for the development of combination microbicides or antiviral drugs. Previously, we reported that the polysulfonic compound ADS-J1 is a potential candidate microbicide that not only inhibits HIV-1 entry, but also seminal fibrils. However, the carcinogenic azo moieties in ADS-J1 preclude its clinical application. Here, we screened several ADS-J1-like analogs and found that the antiparasitic drug suramin most potently inhibited seminal amyloid fibrils. Using various biochemical methods, including Congo red staining, CD analysis, transmission EM, viral infection assays, surface plasmon resonance imaging, and molecular dynamics simulations, we investigated suramin's inhibitory effects and its putative mechanism of action. We found that by forming a multivalent interaction, suramin binds to proteolytic peptides and mature fibrils, thereby inhibiting seminal fibril formation and blocking fibril-mediated enhancement of viral infection. Of note, suramin exhibited potent anti-HIV activities, and combining suramin with several antiretroviral drugs produced synergistic effects against HIV-1 in semen. Suramin also displayed a good safety profile for vaginal application. Moreover, suramin inhibited the semen-derived enhancer of viral infection (SEVI)/semen-mediated enhancement of HIV-1 transcytosis through genital epithelial cells and the subsequent infection of target cells. Collectively, suramin has great potential for further development as a combination microbicide to reduce the spread of the AIDS pandemic by targeting both viral and host factors involved in HIV-1 sexual transmission.


Subject(s)
Amyloid/drug effects , Semen/drug effects , Suramin/pharmacology , Adult , Animals , Anti-HIV Agents/pharmacology , Anti-Retroviral Agents/pharmacology , HIV Infections/metabolism , HIV-1/metabolism , Healthy Volunteers , Humans , Male , Peptide Fragments/metabolism , Peptides/metabolism , Rabbits , Semen/metabolism , Suramin/metabolism
20.
Biochem Biophys Res Commun ; 524(3): 764-771, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32037089

ABSTRACT

ß-Amyloid (Aß) plaque in the brains of patients with Alzheimer's disease (AD) is mainly caused by impaired clearance of Aß by glial cells, including microglia and astrocytes. Because microglia play an important protective role in the central nervous system, many efforts have been made to identify agents that effectively improve microglial Aß phagocytosis. This study found that TLQP-21, which is cleaved from VGF (VGF nerve growth factor inducible) precursor protein, enhanced Aß phagocytosis and degradation by microglial BV2 cells. TLQP-21 also improved microglial phagocytic activity and promoted fibrillar amyloid-ß (fAß) uptake by microglial BV2 cells via a C3AR1-dependent mechanism. Moreover, TLQP-21 stimulated Aß degradation by enhancing lysosome activity, thereby enhancing fAß clearance. These results suggest that treatment with TLQP-21 may be a novel therapeutic strategy to efficiently enhance microglial Aß clearance in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Extracellular Space/metabolism , Microglia/metabolism , Peptide Fragments/pharmacology , Amyloid/drug effects , Animals , Cell Line , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Microglia/drug effects , Neuropeptides/pharmacology , Phagocytosis/drug effects , Proteolysis/drug effects , Receptors, Complement/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL