Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26.705
Filter
1.
Biochemistry ; 63(18): 2245-2256, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39222658

ABSTRACT

Type 1 diabetes results from the autoimmune destruction of pancreatic insulin-producing ß-cells, primarily targeted by autoreactive T cells that recognize insulin B9-23 peptides as antigens. Using drift tube ion mobility spectrometry-mass spectrometry, transmission electron microscopy, and two-dimensional infrared spectroscopy, we characterized mouse insulin 1 B9-23 (Ins1 B9-23), insulin 2 B9-23 (Ins2 B9-23), along with two of their mutants, Ins2 B9-23 Y16A and Ins2 B9-23 C19S. Our findings indicate that Ins1 B9-23 and the Ins2 Y16A mutant exhibit rapid fibril formation, whereas Ins2 B9-23 and the Ins2 C19S mutant show slower fibrillization and a structural rearrangement from globular protofibrils to fibrillar aggregates. These differences in aggregation behaviors also manifest in interactions with (-)epigallocatechin gallate (EGCG), a canonical amyloid inhibitor. EGCG effectively disrupts the fibrils formed by Ins1 B9-23 and the Y16A mutant. However, it proves ineffective in preventing fibril formation of Ins2 B9-23 and the C19S mutant. These results establish a strong correlation between the aggregation behaviors of these peptides and their divergent effects on anti-islet autoimmunity.


Subject(s)
Insulin , Peptide Fragments , Animals , Mice , Insulin/chemistry , Insulin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/genetics , Peptide Fragments/immunology , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Catechin/metabolism , Amyloid/chemistry , Amyloid/metabolism
2.
J Mass Spectrom ; 59(10): e5090, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39328006

ABSTRACT

The study of metal ion's role in the biological processes of Alzheimer's disease has spurred investigations into the coordination chemistry of amyloid beta peptide and its fragments. Nano-electrospray ionization mass spectrometry (nESI-MS) has been utilized to examine the stabilization of bound anions on multiprotein complexes without bulk solvent. However, the effects of anions on metal ion binding interactions with amyloid beta peptide have not been explored. This study directly examined metal-peptide complexes using nESI-MS and investigated the effects of various anions on the binding ratio and stability of these complexes from ammonium salt solutions. The results indicate that different anions have distinct effects on the binding ratio and stability of various metal-peptide complexes. Of these, the bicarbonate ion exhibits the highest binding ratios for metal-peptide complexes, while binding ratios for these complexes in phosphate are comparatively low. Our results suggest that acetate, formate, bicarbonate, and phosphate have weak affinities and act as weak stabilizers of the metal-peptide complex structure in the gas phase. Intriguingly, chloride and sulfate act as stabilizers of the metal-peptide complex in the gas phase. The rank order determined from these data is substantially different from the Hofmeister salt series in solution. Although this outcome was anticipated due to the reduced influence of anions and water solvation, our findings correlate well with expected anion binding in solution and emphasize the importance of both hydration layer and anion-metal-peptide binding effects for Hofmeister-type stabilization in solution. This approach proved useful in examining the interactions between metal ions and amyloid beta peptide, which are relevant to Alzheimer's disease, using direct ESI-MS.


Subject(s)
Amyloid beta-Peptides , Anions , Ion Mobility Spectrometry , Spectrometry, Mass, Electrospray Ionization , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Anions/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Ion Mobility Spectrometry/methods , Protein Binding , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Metals/chemistry , Metals/metabolism , Humans
3.
Int J Mol Sci ; 25(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39337389

ABSTRACT

The life cycle of Ebola and Marburg viruses includes a step of the virion envelope fusion with the cell membrane. Here, we analyzed whether the fusion of liposome membranes under the action of fragments of fusion peptides of Ebola and Marburg viruses depends on the composition of lipid vesicles. A fluorescence assay and electron microscopy were used to quantify the fusogenic activity of the virus fusion peptides and to identify the lipid determinants affecting membrane merging. Differential scanning calorimetry of lipid phase transitions revealed alterations in the physical properties of the lipid matrix produced by virus fusion peptides. Additionally, we found that plant polyphenols, quercetin, and myricetin inhibited vesicle fusion induced by the Marburg virus fusion peptide.


Subject(s)
Ebolavirus , Flavonoids , Marburgvirus , Ebolavirus/drug effects , Marburgvirus/drug effects , Marburgvirus/chemistry , Flavonoids/chemistry , Flavonoids/pharmacology , Membrane Fusion/drug effects , Liposomes/chemistry , Quercetin/chemistry , Quercetin/pharmacology , Virus Internalization/drug effects , Hemorrhagic Fever, Ebola/virology , Polyphenols/chemistry , Polyphenols/pharmacology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Humans , Cell Membrane/metabolism , Peptides/chemistry , Peptides/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology
4.
J Am Soc Mass Spectrom ; 35(10): 2267-2271, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39304183

ABSTRACT

Apolipoprotein A-I (ApoA-I), one of the most abundant proteins in plasma and the major protein component of high-density lipoprotein (HDL), is naturally found in several proteoforms; two of them are ProApoA-I and mature ApoA-I. These two proteoforms of ApoA-I coexist in biological samples and differ only in their N-terminal end. Virtually, the only way to differentiate them is by detecting the proteoform-specific N-terminal proteolytic peptides (RHFWQQDEPPQSPWDR and DEPPQSPWDR, respectively) using liquid chromatography in multiple reaction monitoring mode mass spectrometry (LC-MRM-MS). We have developed a bottom-up LC-MRM-MS method to simultaneously detect proApoA-I and mature ApoA-I. To test the specificity of the method, we digested with trypsin purified mature ApoA-I and recombinant proApoA-I. As expected, only the N-term peptide corresponding to the mature ApoA-I proteoform (DEPPQSPWDR) was detected when digesting mature ApoA-I. However, the digestion of the proApoA-I produced not only the N-terminal peptide corresponding to proApoA-I (RHFWQQDEPPQSPWDR) but also the N-terminal tryptic peptide corresponding to mature ApoA-I (DEPPQSPWDR). This effect was produced by standard and high-specificity trypsin as well as by the Arg-C enzyme in a self-limited manner (approximately 10% of the total). The synthetic proApo-I peptide is not cleaved by trypsin, suggesting that the here reported effect is dependent on protein conformation. The effect is not negligible, as it can be detected by LC-MRM-MS, and correction calculations should be applied to accurately quantify proApoA-I and mature ApoA-I in biological samples where these two proteoforms may coexist.


Subject(s)
Apolipoprotein A-I , Mass Spectrometry , Trypsin , Apolipoprotein A-I/analysis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Trypsin/metabolism , Trypsin/chemistry , Humans , Chromatography, Liquid/methods , Mass Spectrometry/methods , Amino Acid Sequence , Peptide Fragments/analysis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Molecular Sequence Data
5.
Sci Rep ; 14(1): 22246, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333193

ABSTRACT

Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, impacting millions of individuals worldwide. Among its defining characteristics is the accumulation of senile plaques within the brain's gray matter, formed through the self-assembly of misfolded proteins contributing to the progressive symptoms of AD. This study investigates a polymorphic Aß fibril under static and oscillating electric fields using molecular dynamics simulation. Specifically, we utilized a polymorphic fibrillar complex composed of two intertwined pentamer-strands of the Aß1-40 peptide with the Osaka mutation (E22Δ), known for its toxicity and stable structure. Our findings demonstrate that a 0.3 and 0.4 V/nm electric field combined with a 0.20 GHz frequency effectively disrupts the polymorphic conformation of Aß fibrils. Furthermore, we elucidate the molecular mechanisms underlying this disruption, providing insights into the potential therapeutic use of oscillating electric fields for AD. This research offers valuable insights into novel therapeutic approaches for combating AD pathology.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Electricity , Molecular Dynamics Simulation , Mutation , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/chemistry , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Computer Simulation , Amyloid/chemistry , Amyloid/metabolism
6.
Biomolecules ; 14(9)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39334900

ABSTRACT

Neurotransmission is critical for brain function, allowing neurons to communicate through neurotransmitters and neuropeptides. RVD-hemopressin (RVD-Hp), a novel peptide identified in noradrenergic neurons, modulates cannabinoid receptors CB1 and CB2. Unlike hemopressin (Hp), which induces anxiogenic behaviors via transient receptor potential vanilloid 1 (TRPV1) activation, RVD-Hp counteracts these effects, suggesting that it may block TRPV1. This study investigates RVD-Hp's role as a TRPV1 channel blocker using HEK293 cells expressing TRPV1-GFP. Calcium imaging and patch-clamp recordings demonstrated that RVD-Hp reduces TRPV1-mediated calcium influx and TRPV1 ion currents. Molecular docking and dynamics simulations indicated that RVD-Hp interacts with TRPV1's selectivity filter, forming stable hydrogen bonds and van der Waals contacts, thus preventing ion permeation. These findings highlight RVD-Hp's potential as a therapeutic agent for conditions involving TRPV1 activation, such as pain and anxiety.


Subject(s)
Endocannabinoids , Molecular Docking Simulation , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , Humans , HEK293 Cells , Endocannabinoids/pharmacology , Endocannabinoids/metabolism , Endocannabinoids/chemistry , Calcium/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Molecular Dynamics Simulation , Hemoglobins
7.
ACS Chem Neurosci ; 15(18): 3344-3353, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39222387

ABSTRACT

Mono- and polyunsaturated fatty acids (FAs) are broadly used as food supplements. However, their effect on the aggregation of amyloidogenic proteins remains unclear. In this study, we investigated the effect of a large number of mono- and polyunsaturated, as well as fully saturated FAs on the aggregation of amyloid ß1-42 (Aß1-42) peptide. A progressive aggregation of this peptide is the expected molecular cause of Alzheimer's disease (AD), one of the most common neurodegenerative pathologies in the world. We found that arachidonic and stearic acids delayed the aggregation of Aß1-42. Using Nano-Infrared spectroscopy, we found that FAs caused very little if any changes in the secondary structure of Aß1-42 oligomers and fibrils formed at different stages of protein aggregation. However, the analyzed mono- and polyunsaturated, as well as fully saturated FAs uniquely altered the toxicity of Aß1-42 fibrils. We found a direct relationship between the degree of FAs unsaturation and toxicity of Aß1-42 fibrils formed in their presence. Specifically, with an increase in the degree of unsaturation, the toxicity Aß1-42/FA fibrils increased. These results indicate that fully saturated or monounsaturated FAs could be used to decrease the toxicity of amyloid aggregates and, consequently, decelerate the development of AD.


Subject(s)
Amyloid beta-Peptides , Fatty Acids , Peptide Fragments , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Peptide Fragments/metabolism , Peptide Fragments/chemistry , Fatty Acids/metabolism , Fatty Acids/chemistry , Humans , Amyloid/metabolism , Amyloid/chemistry , Protein Structure, Secondary
8.
J Phys Chem Lett ; 15(37): 9543-9547, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39265045

ABSTRACT

This study investigates the lasing effects in a Fabry-Perot cavity to discern the binding interactions of thioflavin T (ThT) with various peptides associated with Alzheimer's disease, including Aß(1-42), KLVFFA, and diphenylalanine (FF) in the condensed phase. Utilizing kinetic lasing measurements, the research explores ThT emission enhancements due to specific groove binding in ß-sheet structures and highlights additional contributions from weak surface interactions and solvent-solute interactions. Lasing spectroscopy reveals a lack of transition of the FF system from its native state to an amyloid-like structure, challenging traditional ThT assay interpretations. These findings show the potential of lasing spectroscopy in elucidating the molecular basis of amyloid fibril formation and the development of diagnostic tools for amyloidogenic diseases.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Benzothiazoles , Benzothiazoles/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Protein Conformation, beta-Strand , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Humans , Phenylalanine/chemistry , Dipeptides/chemistry , Dipeptides/metabolism , Protein Binding , Kinetics
9.
J Phys Chem B ; 128(38): 9143-9150, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39283804

ABSTRACT

We developed a coarse-grained model for the protic ionic liquid, triethylammonium mesylate ([TEA]+[Ms]-), to characterize its inhibitory effects on amyloid aggregation using the K16LVFFAE22 fragment of the amyloid-ß (Aß16-22) as a model amyloidogenic peptide. In agreement with previous experiments, coarse-grained molecular dynamics simulations showed that increasing concentrations of [TEA]+[Ms]- in aqueous media led to increasingly small Aß16-22 aggregates with low beta-sheet contents. The cause of [TEA]+[Ms]-'s inhibition of peptide aggregation was found to be a result of two interrelated effects. At a local scale, the enrichment of interactions between [TEA]+ cations and hydrophobic phenylalanine side chains weakened the hydrophobic cores of amyloid aggregates, resulting in poorly ordered structures. At a global level, peptides tended to localize at the interfaces of IL-rich nanostructures with water. At high IL concentrations, when the IL-water interface was large or fragmented, Aß16-22 peptides were dispersed in the simulation cell, sometimes sequestered at unaggregated monomeric states. Together, these phenomena underlie [TEA]+[Ms]-'s inhibition of amyloid aggregation. This work addresses the critical lack of knowledge on the mechanisms of protein-ionic liquid interactions and may have broader implications for industrial applications.


Subject(s)
Amyloid beta-Peptides , Hydrophobic and Hydrophilic Interactions , Ionic Liquids , Molecular Dynamics Simulation , Peptide Fragments , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Ionic Liquids/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Nanostructures/chemistry
10.
Inorg Chem ; 63(39): 17983-17992, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39287976

ABSTRACT

Two dual fluorescent/phosphorescent tris-heteroleptic mononuclear Ru(ΙΙ) complexes (2 and 3) were designed and applied in amyloid-ß (Aß) sensing. These complexes have a general formula of [Ru(phen)(dppz)(L)](PF6)2, where L is (2-pyrazinyl)(2-pyridyl)(methyl)amine (H-L) with different substituents (-OMe for 2, -H for 3), phen is 1,10-phenanthroline, and dppz is dipyridophenazine, respectively. Compared with the previously reported ratiometric probe 1 with a di(pyrid-2-yl)(methyl)amine ligand, complex 2 can be employed for not only ratiometric emissive detection of Aß aggregation but also ratiometric imaging detection of Aß fibrils. In ratiometric emissive detection, as the incubation time of the Aß sample (Aß40 and Aß42) was prolonged, a new phosphorescence emission band appeared with gradual enhancement of the emission intensity, while the fluorescence emission was basically unchanged, which could be treated as an intrinsic internal reference signal. In comparison, a larger ratiometric photoluminescence enhancement (I640/I440) was observed for Aß40 aggregation with respect to Aß42. In ratiometric imaging detection, the imaging signals obtained from the phosphorescence emission are much brighter than the fluorescence emission in both Aß40 and Aß42 fibrils. As indicated by molecular docking results, stronger interactions were found between complex 2 with Aß40 fibrils, which included π/π, π/C-H, and π/H interactions between bidentate ligands dppz and phen with amino acid residues. Moreover, computational calculations were carried out to assist the interpretation of these experimental findings.


Subject(s)
Amyloid beta-Peptides , Coordination Complexes , Ruthenium , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/analysis , Ruthenium/chemistry , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Humans , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Molecular Structure , Molecular Docking Simulation , Optical Imaging , Peptide Fragments/chemistry , Peptide Fragments/analysis
11.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337556

ABSTRACT

Alzheimer's disease (AD) is one of the most common causes of dementia, accounting for more than 60% of all cases. It is a neurodegenerative disease in which symptoms such as a decline in memory, thinking, learning, and organizing skills develop gradually over many years and eventually become more severe. To date, there is no effective treatment for the cause of Alzheimer's disease, and the existing pharmacological options primarily help manage symptoms. Treatment is mainly based on acetylcholinesterase (AChE) inhibitors such as donepezil, rivastigmine, and galantamine, which exhibit numerous adverse cardiovascular and gastrointestinal effects due to excessive stimulation of peripheral cholinergic activity involving muscarinic receptors. Therefore, in addition to the obvious drugs that act on the cause of the disease, new drugs based on AChE inhibition that show the fewest side effects are needed. One potential drug could be a new compound under study, tetrahydroacridine derivative (CHDA), which showed significant potential to inhibit the AChE enzyme in previous in vitro studies. The present study shows that while having very potent AChE inhibitory properties, CHDA is a compound with low toxicity to nerve cell culture and living organisms. In addition, it exhibits dissociative activity against amyloid ß fibrils, which is extremely important for applications in Alzheimer's disease therapy.


Subject(s)
Acetylcholinesterase , Amyloid beta-Peptides , Cholinesterase Inhibitors , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemistry , Amyloid beta-Peptides/metabolism , Humans , Acetylcholinesterase/metabolism , Acetylcholinesterase/chemistry , Acridines/pharmacology , Acridines/chemistry , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amyloid/metabolism
12.
Cell Mol Life Sci ; 81(1): 409, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289189

ABSTRACT

Autophagy is an evolutionarily ancient catabolic pathway and has recently emerged as an integral part of the innate immune system. While the core machinery of autophagy is well defined, the physiological regulation of autophagy is less understood. Here, we identify a C-terminal fragment of human hemoglobin A (HBA1, amino acids 111-132) in human bone marrow as a fast-acting non-inflammatory inhibitor of autophagy initiation. It is proteolytically released from full-length HBA1 by cathepsin E, trypsin or pepsin. Biochemical characterization revealed that HBA1(111-132) has an in vitro stability of 52 min in human plasma and adopts a flexible monomeric conformation in solution. Structure-activity relationship studies revealed that the C-terminal 13 amino acids of HBA1(120-132) are sufficient to inhibit autophagy, two charged amino acids (D127, K128) mediate solubility, and two serines (S125, S132) are required for function. Successful viruses like human immunodeficiency virus 1 (HIV-1) evolved strategies to subvert autophagy for virion production. Our results show that HBA1(120-132) reduced virus yields of lab-adapted and primary HIV-1. Summarizing, our data identifies naturally occurring HBA1(111-132) as a physiological, non-inflammatory antagonist of autophagy. Optimized derivatives of HBA1(111-132) may offer perspectives to restrict autophagy-dependent viruses.


Subject(s)
Autophagy , HIV-1 , Humans , HIV-1/metabolism , HIV-1/physiology , Structure-Activity Relationship , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Amino Acid Sequence
13.
Sci Rep ; 14(1): 22633, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349560

ABSTRACT

The deposition of the amyloid-ß (Aß) peptide into amyloid fibrils is a hallmark of Alzheimer's disease. Recently, it has been reported that some proteins can aggregate and form amyloids through an intermediate pathway involving a liquid-like condensed phase. These observations prompted us to investigate the phase space of Aß. We thus explored the ability of Aß to undergo liquid-liquid phase separation, and the subsequent liquid-to-solid transition that takes place within the resulting condensates. Through the use of microfluidic approaches, we observed that the 40-residue form of Αß (Αß40) can undergo liquid-liquid phase separation, and that accessing a liquid-like intermediate state enables Αß40 to self-assemble and aggregate into amyloid fibrils through this pathway. These results prompt further studies to investigate the possible role of Αß liquid-liquid phase separation and its subsequent aggregation in the context of Alzheimer's disease and more generally on neurodegenerative processes.


Subject(s)
Amyloid beta-Peptides , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Humans , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phase Transition , Protein Aggregates , Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloid/metabolism , Protein Aggregation, Pathological/metabolism , Liquid-Liquid Extraction/methods , Phase Separation
14.
Biochemistry ; 63(19): 2397-2413, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39255071

ABSTRACT

Amyloidosis of amyloid-ß (Aß) triggers a cascade of events, leading to oxidative damage and neuronal death. Therefore, inhibiting Aß amyloidosis or disrupting the matured fibrils is the primary target to combat progressive Alzheimer's disease (AD) pathogenesis. Here, we undertake optimization strategies to improve the antiamyloid efficiency of our previously reported NF11 (NAVRWSLMRPF) peptide. Among the series of peptides tested, nontoxic and serum-stable peptide 1 or P1 containing an anthranilic acid residue shows immense potential in not only inhibiting the Aß42 amyloid formation but also disrupting the mature Aß42 fibrils into nontoxic small molecular weight soluble species. Our studies provide high-resolution characterization of the peptide's mechanism of action. With a binding affinity within the micromolar range for both the monomer and aggregated Aß42, this α/ß hybrid peptide can efficiently modulate Aß amyloidosis while facilitating the clearance of toxic aggregates and enforcing protection from apoptosis. Thus, our studies highlight that incorporating a ß-amino acid not only imparts protection from proteolytic degradation and improved stability but also functions effectively as a ß breaker, redirecting the aggregation kinetics toward off-pathway fibrillation.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Peptide Fragments , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amyloid/metabolism , Amyloid/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/prevention & control , Protein Aggregation, Pathological/drug therapy , Peptides/chemistry , Peptides/pharmacology , Animals
15.
ACS Chem Neurosci ; 15(17): 3136-3151, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39158263

ABSTRACT

The inhibition of amyloid-ß (Aß) fibrillation and clearance of Aß aggregates have emerged as a potential pharmacological strategy to alleviate Aß aggregate-induced neurotoxicity in Alzheimer's disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged N-substituted oligopyrrolamides for its notable ability to inhibit Aß fibrillation, disintegrate intracellular cytotoxic Aß oligomers, and alleviate Aß-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aß42 fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aß42 fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of ß-strands and the emergence of a helix conformation, indicating a loss of the well-organized ß-sheet-rich structure of the disease-relevant Aß42 fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aß42 fibril (ΔGbinding= -142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged N-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aß42 fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aß peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aß42 fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aß42 fibril. The illumination of key interactions responsible for the destabilization of the Aß42 fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aß aggregates in AD.


Subject(s)
Amyloid beta-Peptides , Molecular Dynamics Simulation , Peptide Fragments , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Humans , Peptide Fragments/metabolism , Peptide Fragments/chemistry , Amyloid/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy
16.
J Am Chem Soc ; 146(35): 24426-24439, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39177153

ABSTRACT

The two most abundant isoforms of amyloid-ß (Aß) are the 40- (Aß40) and 42-residue (Aß42) peptides. Since they coexist and there is a correlation between toxicity and the ratio of the two isoforms, quantitative characterization of their interactions is crucial for understanding the Aß aggregation mechanism. In this work, we follow the aggregation of individual isoforms in a mixture using single-molecule FRET spectroscopy by labeling Aß42 and Aß40 with the donor and acceptor fluorophores, respectively. We found that there are two phases of aggregation. The first phase consists of coaggregation of Aß42 with a small amount of Aß40, while the second phase results mostly from aggregation of Aß40. We also found that the aggregation of Aß42 is slowed by Aß40 while the aggregation of Aß40 is accelerated by Aß42 in a concentration-dependent manner. The formation of oligomers was monitored by incubating mixtures in a plate reader and performing a single-molecule free-diffusion experiment at several different stages of aggregation. The detailed properties of the oligomers were obtained by maximum likelihood analysis of fluorescence bursts. The FRET efficiency distribution is much broader than that of the Aß42 oligomers, indicating the diversity in isoform composition of the oligomers. Pulsed interleaved excitation experiments estimate that the fraction of Aß40 in the co-oligomers in a 1:1 mixture of Aß42 and Aß40 varies between 0 and 20%. The detected oligomers were mostly co-oligomers especially at the physiological ratio of Aß42 and Aß40 (1:10), suggesting the critical role of Aß40 in oligomer formation and aggregation.


Subject(s)
Amyloid beta-Peptides , Fluorescence Resonance Energy Transfer , Peptide Fragments , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregates , Humans , Single Molecule Imaging
17.
Int J Mol Sci ; 25(16)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39201616

ABSTRACT

Amyloid ß peptide (Aß) aggregation and deposition are considered the main causes of Alzheimer's disease. In a previous study, we demonstrated that anionic Zn-phthalocyanine (ZnPc) can interact with the Aß peptide and inhibit the fibril-formation process. However, due to the inability of anionic ZnPc to cross the intact blood-brain barrier, we decided to explore the interaction of cationic methylated Zn-phthalocyanine (cZnPc) with the peptide. Using a ThT fluorescence assay, we observed that cZnPc dose-dependently and time-dependently inhibited Aß1-42 fibril levels under in vitro fibril-formation conditions. Electron microscopy revealed that it caused Aß1-42 peptides to form small aggregates. Western blotting and dot immunoblot oligomer experiments demonstrated that cZnPc increased rather than decreased the levels of oligomers from the very early stages of incubation. A binding assay confirmed that cZnPc could bind with the peptide. Docking simulations indicated that the oligomer species of Aß1-42 had a higher ability to interact with cZnPc. ANS fluorescence assay results indicated that cZnPc did not affect the hydrophobicity of the peptide. However, cZnPc significantly increased intrinsic tyrosine fluorescence of the peptide after 8 h of incubation in fibril-formation conditions. Importantly, cell culture experiments demonstrated that cZnPc did not exhibit any toxicity up to a concentration of 10 µM. Instead, it protected a neuronal cell line from Aß1-42-induced toxicity. Thus, our results suggest that cZnPc can affect the aggregation process of Aß1-42, rendering it non-toxic, which could be crucial for the therapy of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Indoles , Isoindoles , Organometallic Compounds , Peptide Fragments , Zinc Compounds , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Indoles/chemistry , Indoles/pharmacology , Humans , Zinc Compounds/chemistry , Zinc Compounds/pharmacology , Organometallic Compounds/pharmacology , Organometallic Compounds/chemistry , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Protein Aggregates/drug effects , Animals , Molecular Docking Simulation , Neurons/drug effects , Neurons/metabolism
18.
J Biochem Mol Toxicol ; 38(8): e23800, 2024 08.
Article in English | MEDLINE | ID: mdl-39132781

ABSTRACT

Pyroglutamate (pE)-modified amyloid-ß (Aß) peptides play a crucial role in the development of Alzheimer's disease. pEAß3-42 can rapidly form oligomers that gradually elongate hydrophobic segments to form ß-sheet-rich amyloid intermediates, ultimately resulting in the formation of mature amyloid fibrils. pEAß3-42 can also catalyze the aggregation of Aß species and subsequently accelerate the formation of amyloid senile plaques. Considering the recent clinical success of the pEAß3-42-targeting antibody donanemab, molecules that strongly bind pEAß3-42 and prevent its aggregation and catalytic effect on Aßs may also provide potential therapeutic options for Alzheimer's disease. Here, we demonstrate that the natural antibiotic cyclopeptide tyrocidine A (TA) not only strongly inhibits the aggregation of Aß1-42 as previously reported, but also interacts with the hydrophobic C-terminus and middle domain of pEAß3-42 to maintain an unordered conformation, effectively impeding the formation of initial oligomers and subsequently halting the aggregation of pEAß3-42. Furthermore, TA can disrupt the "catalytic effect" of pEAß3-42 on amyloid aggregates, effectively suppressing Aß aggregation and ultimately preventing the pathological events induced by Aßs.


Subject(s)
Amyloid beta-Peptides , Hydrophobic and Hydrophilic Interactions , Pyrrolidonecarboxylic Acid , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/pharmacology , Humans , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy
19.
Int J Nanomedicine ; 19: 8271-8284, 2024.
Article in English | MEDLINE | ID: mdl-39161360

ABSTRACT

Purpose: Development of SERS-based Raman nanoprobes can detect the misfolding of Amyloid beta (Aß) 42 peptides, making them a viable diagnostic technique for Alzheimer's disease (AD). The detection and imaging of amyloid peptides and fibrils are expected to help in the early identification of AD. Methods: Here, we propose a fast, easy-to-use, and simple scheme based on the selective adsorption of Aß42 molecules on SERS active gold nanoprobe (RB-AuNPs) of diameter 29 ± 3 nm for Detection of Alzheimer's Disease Biomarkers. Binding with the peptides results in a spectrum shift, which correlates with the target peptide. We also demonstrated the possibility of using silver nanoparticles (AgNPs) as precursors for the preparation of a SERS active nanoprobe with carbocyanine (CC) dye and AgNPs known as silver nanoprobe (CC-AgNPs) of diameter 25 ± 4 nm. Results: RB-AuNPs probe binding with the peptides results in a spectrum shift, which correlates with the target peptide. Arginine peak appears after the conjugation confirms the binding of Aß 42 with the nanoprobe. Tyrosine peaks appear after conjugated Aß42 with CC-AgNPs providing binding of the peptide with the probe. The nanoprobe produced a strong, stable SERS signal. Further molecular docking was utilized to analyse the interaction and propose a structural hypothesis for the process of binding the nanoprobe to Aß42 and Tau protein. Conclusion: This peptide-probe interaction provides a general enhancement factor and the molecular structure of the misfolded peptides. Secondary structural information may be obtained at the molecular level for specific residues owing to isotope shifts in the Raman spectra. Conjugation of the nanoprobe with Aß42 selectively detected AD in bodily fluids. The proposed nanoprobes can be easily applied to the detection of Aß plaques in blood, saliva, and sweat samples.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Gold , Metal Nanoparticles , Molecular Docking Simulation , Peptide Fragments , Silver , Spectrum Analysis, Raman , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Spectrum Analysis, Raman/methods , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/chemistry , Metal Nanoparticles/chemistry , Gold/chemistry , Silver/chemistry , Humans , Biomarkers/analysis , Adsorption , Peptide Fragments/analysis , Peptide Fragments/chemistry
20.
Colloids Surf B Biointerfaces ; 244: 114182, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39216441

ABSTRACT

Abnormal amyloid ß-protein (Aß42) fibrillation is a key event in Alzheimer's disease (AD), and photodynamic therapy (PDT) possesses great potential in modulating Aß42 self-assembly. However, the poor blood-brain barrier (BBB) penetration, low biocompatibility, and limited tissue penetration depth of existing photosensitizers limit the progress of photo-oxidation strategies. In this paper, novel indocyanine green-modified graphene quantum dot nano-assemblies (NBGQDs-ICGs) were synthesized based on a molecular assembly strategy of electrostatic interactions for PDT inhibition of Aß42 self-assembly process and decomposition of preformed fibrils under near-infrared light. Combining the small-size structure of graphene quantum dots and the near-infrared light-responsive properties of ICGs, the NBGQDs-ICGs could achieve BBB penetration under 808 nm irradiation. More importantly, the neuroprotective mechanism of NBGQDs-ICG was studied for the first time by AFM, which effectively weakened the adhesion of Aß42 aggregates to the cell surface by blocking the interaction between Aß42 and the cell membrane, and restored the mechanical stability and adhesion of the neuron membrane. Meanwhile, NBGQDs-ICG promoted phagocytosis of Aß42 by microglia. In addition, the good biocompatibility and stability ensured the biosafety of NBGQDs-ICG in future clinical applications. We anticipate that such multifunctional nanocomponents may provide promising avenues for the development of novel AD inhibitors.


Subject(s)
Amyloid beta-Peptides , Blood-Brain Barrier , Quantum Dots , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Quantum Dots/chemistry , Humans , Animals , Graphite/chemistry , Graphite/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Particle Size , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Phagocytosis/drug effects , Carbon/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL