Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.522
Filter
1.
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
2.
Talanta ; 280: 126699, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39142131

ABSTRACT

Accurate and sensitive detection of amyloid-ß 42 oligomer (Aß42O) is of great significance for early diagnosis of Alzheimer's disease (AD). Herein, a signal on-off ratiometric electrochemical immunosensor was developed for highly selective and quantitative determination of Aß42O by using novel covalent organic frameworks (COFs) composites as the sensing platform. This immunosensor produced two independent electrochemical signals from the [Fe(CN)6]3-/4- and methylene blue (MB) probes at different potentials based on the electrocatalytic activity of gold nanoparticle-functionalized porphyrinyl COFs nanocomposites toward [Fe(CN)6]3-/4- and the signal probe of MB encapsulated in the aptamer-modified alkynyl COFs. Because the two signals of [Fe(CN)6]3-/4- and MB changed in opposite directions, a signal on-off mode was generated which can correct the results by introducing a reference signal and effectively eliminate background interference. Under optimal experimental conditions, the current ratio (IMB/I[Fe(CN)6]3-/4-) was well linearly related to the logarithmic value of Aß42O concentrations in the range of 10 pM to 1 µM, and the detection limit was 5.1 pM (S/N = 3). Additionally, the immunosensor exhibited satisfactory performance in case of real cerebrospinal fluid samples. The designed ratiometric electrochemical immunosensor provides a valuable route for early diagnosis of AD and our results also pave the way for designing of sensing platforms using COF-based nanomaterials and extending their functions and applications to bioanalysis.


Subject(s)
Amyloid beta-Peptides , Electrochemical Techniques , Metal-Organic Frameworks , Peptide Fragments , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/cerebrospinal fluid , Electrochemical Techniques/methods , Metal-Organic Frameworks/chemistry , Humans , Peptide Fragments/analysis , Peptide Fragments/chemistry , Limit of Detection , Biosensing Techniques/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Methylene Blue/chemistry , Immunoassay/methods , Alzheimer Disease/diagnosis
3.
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
5.
Eur J Med Chem ; 276: 116682, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39053190

ABSTRACT

Timely diagnosis and therapy of Alzheimer's disease remains one of the greatest questions in medicinal chemistry of neurodegenerative disease. The lack of low-cost sensors capable of reliable detection of structural changes in AD-related proteins is the driving factor for the development of novel molecules with affinity for AD hallmarks. The development of cheap, safe diagnostic methods is a highly sought-after area of research. Optical fluorescent probes are of great interest due to their non-radioactivity, low cost, and ability of the real-time visualization of AD hallmarks. Boron dipyrromethene (BODIPY)-based fluorophore is one promising fluorescent unit for in vivo labeling due to its high photostability, easy modification, low toxicity, and cell-permeability. In recent years, many fluorescent BODIPY-based probes capable of Aß plaque, Aß soluble oligomers, neurofibrillary tangles (NFT) optical detection, as well as probes with copper ion chelating units and viscosity sensors have been developed. In this review, we summarized BODIPY derivatives as fluorescent sensors capable of detecting pathological features of Alzheimer's disease, published from 2009 to 2023, as well as their design strategies, optical properties, and in vitro and in vivo activities.


Subject(s)
Alzheimer Disease , Boron Compounds , Fluorescent Dyes , Alzheimer Disease/diagnosis , Boron Compounds/chemistry , Boron Compounds/chemical synthesis , Humans , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Animals , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Molecular Structure
6.
Sci Rep ; 14(1): 16084, 2024 07 12.
Article in English | MEDLINE | ID: mdl-38992063

ABSTRACT

Cerebrospinal fluid (CSF) core biomarkers of Alzheimer's disease (AD), including amyloid peptide beta-42 (Aß42), Aß42/40 ratio, and phosphorylated tau (pTau), are precious tools for supporting AD diagnosis. However, their use in clinical practice is limited due to the invasiveness of CSF collection. Thus, there is intensive research to find alternative, noninvasive, and widely accessible biological matrices to measure AD core biomarkers. In this study, we measured AD core biomarkers in saliva and plasma by a fully automated platform. We enrolled all consecutive patients with cognitive decline. For each patient, we measured Aß42, Aß40, and pTau levels in CSF, saliva, and plasma by Lumipulse G1200 (Fujirebio). We included forty-two patients, of whom 27 had AD. Levels of all biomarkers significantly differed in the three biofluids, with saliva having the lowest and CSF the highest levels of Aß42, Aß40, and pTau. A positive correlation of pTau, Aß42/40 ratio, and pTau/Aß42 ratio levels in CSF and plasma was detected, while no correlation between any biomarker in CSF and saliva was found. Our findings suggest that plasma but not saliva could represent a surrogate biofluid for measuring core AD biomarkers. Specifically, plasma Aß42/40 ratio, pTau/Aß42 ratio, and pTau could serve as surrogates of the corresponding CSF biomarkers.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Saliva , tau Proteins , Humans , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Saliva/metabolism , Saliva/chemistry , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Female , Male , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/analysis , tau Proteins/cerebrospinal fluid , tau Proteins/blood , tau Proteins/analysis , Middle Aged , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Peptide Fragments/analysis , Luminescent Measurements/methods , Aged, 80 and over
8.
Bioorg Chem ; 150: 107514, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38870704

ABSTRACT

Alzheimer's disease (AD), a progressive neurodegenerative disorder, has garnered increased attention due to its substantial economic burden and the escalating global aging phenomenon. Amyloid-ß deposition is a key pathogenic marker observed in the brains of Alzheimer's sufferers. Based on real-time, safe, low-cost, and commonly used, near-infrared fluorescence (NIRF) imaging technology have become an essential technique for the detection of AD in recent years. In this work, NIRF probes with hemicyanine structure were designed, synthesized and evaluated for imaging Aß aggregates in the brain. We use the hemicyanine structure as the parent nucleus to enhance the probe's optical properties. The introduction of PEG chain is to improve the probe's brain dynamice properties, and the alkyl chain on the N atom is to enhance the fluorescence intensity of the probe after binding to the Aß aggregates as much as possible. Among these probes, Z2, Z3, Z6, X3, X6 and T1 showed excellent optical properties and high affinity to Aß aggregates (Kd = 24.31 âˆ¼ 59.60 nM). In vitro brain section staining and in vivo NIRF imaging demonstrated that X6 exhibited superior discrimination between Tg mice and WT mice, and X6 has the best brain clearance rate. As a result, X6 was identified as the optimal probe. Furthermore, the docking theory calculation results aided in describing X6's binding behavior with Aß aggregates. As a high-affinity, high-selectivity, safe and effective probe of targeting Aß aggregates, X6 is a promising NIRF probe for in vivo detection of Aß aggregates in the AD brain.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Carbocyanines , Drug Design , Fluorescent Dyes , Optical Imaging , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Animals , Carbocyanines/chemistry , Carbocyanines/chemical synthesis , Mice , Molecular Structure , Humans , Protein Aggregates , Structure-Activity Relationship , Brain/diagnostic imaging , Brain/metabolism , Dose-Response Relationship, Drug
9.
Talanta ; 277: 126362, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38843773

ABSTRACT

Alzheimer's disease (AD), gradually recognized as an untreatable neurodegenerative disorder, has been considered to be closely associated with Aß plaques, which consist of ß-amyloid protein (Aß) and is one of the crucial pathological features of AD. There are no obvious symptoms in the initial phase of AD, and thus the therapeutic interventions are important for early diagnosis of AD. Moreover, recent researches have indicated that lipid droplets might serve as a potential ancillary biomarker, and its viscosity changes are closely associated to the pathological process of AD. Herein, two newly fluorescent probes 5QSZ and BQSZ have been developed and synthesized for identifying Aß aggregates and detecting the viscosity of lipid droplet. After selectively binding to Aß aggregates, 5QSZ and BQSZ exhibited linear and obvious fluorescence enhancements (32.58 and 36.70 folds), moderate affinity (Kd = 268.0 and 148.6 nM) and low detection limits (30.11 and 65.37 nM) in aqueous solutions. Further fluorescence staining of 5QSZ on brain tissue sections of APP/PS1 transgenic mouse exhibited the higher selectivity of 5QSZ towards Aß aggregates locating at the core of the plaques. Furthermore, 5QSZ and BQSZ displayed strong linear fluorescence emission enhancements towards viscosity changes and would be utilized to monitor variation in cellular viscosity induced by LPS or monensin. These two probes were non-cytotoxic and showed good localization in lipid droplets. Therefore, 5QSZ and BQSZ could serve as potential bi-functional fluorescent probes to image Aß aggregates and monitor the viscosity of lipid droplets, which have significant implications for the early diagnosis and progression of AD.


Subject(s)
Amyloid beta-Peptides , Boron Compounds , Fluorescent Dyes , Lipid Droplets , Protein Aggregates , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Viscosity , Boron Compounds/chemistry , Boron Compounds/chemical synthesis , Animals , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Mice , Humans , Mice, Transgenic , Alzheimer Disease/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/diagnostic imaging , Optical Imaging
10.
ACS Sens ; 9(6): 3387-3393, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38850514

ABSTRACT

Fatty acid amide hydrolase (FAAH) plays a crucial role in the metabolism of the endocannabinoid system by hydrolyzing a series of bioactive amides, whose abnormal levels are associated with neuronal disorders including Alzheimer's disease (AD). However, due to the lack of suitable quantitative sensing tools, real-time and accurate monitoring of the activity of FAAH in living systems remains unresolved. Herein, a novel enzyme-activated near-infrared two-photon ratiometric fluorescent probe (CANP) based on a naphthylvinylpyridine monofluorophore is successfully developed, in which the electron-withdrawing amide moiety is prone to be hydrolyzed to an electron-donating amine group under the catalysis of FAAH, leading to the activation of the intramolecular charge transfer process and the emergence of a new 80 nm red-shifted emission, thereby achieving a ratiometric luminescence response. Benefiting from the high selectivity, high sensitivity, and ratiometric response to FAAH, the probe CANP is successfully used to quantitatively monitor and image the FAAH levels in living neurons, by which an amyloid ß (Aß)-induced upregulation of endogenous FAAH activity is observed. Similar increases in FAAH activity are found in various brain regions of AD model mice, indicating a potential fatty acid amide metabolite-involved pathway for the pathological deterioration of AD. Moreover, our quantitative FAAH inhibition experiments further demonstrate the great value of CANP as an efficient visual probe for in situ and precise assessment of FAAH inhibitors in complex living systems, assisting the discovery of FAAH-related therapeutic agents.


Subject(s)
Amidohydrolases , Brain , Fluorescent Dyes , Neurons , Amidohydrolases/metabolism , Animals , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Brain/diagnostic imaging , Brain/metabolism , Neurons/metabolism , Mice , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Humans , Pyridines/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Photons
11.
Anal Chem ; 96(24): 9799-9807, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38830618

ABSTRACT

Cerebral accumulation of amyloid-ß (Aß) initiates molecular and cellular cascades that lead to Alzheimer's disease (AD). However, amyloid deposition does not invariably lead to dementia. Amyloid-positive but cognitively unaffected (AP-CU) individuals present widespread amyloid pathology, suggesting that molecular signatures more complex than the total amyloid burden are required to better differentiate AD from AP-CU cases. Motivated by the essential role of Aß and the key lipid involvement in AD pathogenesis, we applied multimodal mass spectrometry imaging (MSI) and machine learning (ML) to investigate amyloid plaque heterogeneity, regarding Aß and lipid composition, in AP-CU versus AD brain samples at the single-plaque level. Instead of focusing on a population mean, our analytical approach allowed the investigation of large populations of plaques at the single-plaque level. We found that different (sub)populations of amyloid plaques, differing in Aß and lipid composition, coexist in the brain samples studied. The integration of MSI data with ML-based feature extraction further revealed that plaque-associated gangliosides GM2 and GM1, as well as Aß1-38, but not Aß1-42, are relevant differentiators between the investigated pathologies. The pinpointed differences may guide further fundamental research investigating the role of amyloid plaque heterogeneity in AD pathogenesis/progression and may provide molecular clues for further development of emerging immunotherapies to effectively target toxic amyloid assemblies in AD therapy. Our study exemplifies how an integrative analytical strategy facilitates the unraveling of complex biochemical phenomena, advancing our understanding of AD from an analytical perspective and offering potential avenues for the refinement of diagnostic tools.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Plaque, Amyloid , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/diagnosis , Humans , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/chemistry , Brain/metabolism , Brain/pathology , Lipids/analysis , Lipids/chemistry , Machine Learning , Aged
12.
Anal Chim Acta ; 1314: 342779, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38876518

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease in the world and poses a huge challenge to global healthcare. Early and accurate detection of amyloid-ß (1-42) (Aß42), a key biomarker of AD, is crucial for effective diagnosis and intervention of AD. Specific or overexpressed proteins on extracellular vesicles (EVs) describe a close correlation with the occurrence and development of diseases. EVs are a very promising non-invasive biomarker for the diagnosis of AD and other diseases. As a sensitive, simple and rapid analytical method, fluorescence resonance energy transfer (FRET) has been widely applied in the detection of EVs. Herein, we developed a dual labelling strategy for simultaneously detecting EV membrane proteins of Aß42 and CD63 based on FRET pair consisting of Au nanoclusters (AuNCs) and polydopamine nanospheres (PDANSs). The constructed nanoprobe, termed EVMPFAP assay, could specifically measure the Aß42 and CD63 on EVs with excellent sensitivity, high specificity and satisfactory accuracy. The limit of detection of EVMPFAP assay was 1.4 × 103 particles mL-1 and the linear range was from 104 to 108 particles mL-1. EVMPFAP assay was successfully used to analyze plasma EVs to distinguish AD and healthy mice. We expect that EVMPFAP assay can be routinely applied for early diagnosis and development-monitoring of AD, thus facilitating the fight against AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Extracellular Vesicles , Fluorescence Resonance Energy Transfer , Gold , Metal Nanoparticles , Tetraspanin 30 , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Extracellular Vesicles/chemistry , Animals , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/blood , Mice , Humans , Tetraspanin 30/metabolism , Gold/chemistry , Metal Nanoparticles/chemistry , Peptide Fragments/analysis , Peptide Fragments/blood , Peptide Fragments/chemistry , Polymers/chemistry , Indoles/chemistry , Limit of Detection
13.
Anal Chem ; 96(28): 11397-11403, 2024 07 16.
Article in English | MEDLINE | ID: mdl-38940533

ABSTRACT

Low-mass soluble ß-amyloid peptide oligomers (LSAßOs) play a crucial role in the pathogenesis of Alzheimer's disease. However, these oligomers exhibit heterogeneity in terms of structure, stability, and stoichiometry, and their abundance in biofluids is low, making accurate identification challenging. In this study, we developed a DNA nanocage-assisted method for selective sizing and sensitive quantification of LSAßOs in serum. Using LSAßO less than 10 kDa (LSAßO10kD) and less than 30 kDa (LSAßO30kD) as models, the size-matching rules between DNA nanocages and LSAßOs were investigated, and two appropriate nanocages were selected for the detection of two LSAßOs, respectively. Both nanocages were functionalized by encapsulating oligomer's aptamer and a complementary sequence within their cavities. Once the LSAßO entered the corresponding nanocage cavity, the complementary sequence was released, triggering a hybridization chain reaction on an electrochemical sensing platform. The system achieved size-selective discrimination of LSAßO10kD with a linear range of 10-150 pM and LSAßO30kD with a linear range of 15-150 pM. Real sample testing confirmed the applicability of the method for blood-based diagnosis. The DNA nanocage-assisted electrochemical analysis platform provides an accurate, highly selective, and sensitive approach for oligomer analysis, which is significant for amyloid protein research and related disease diagnosis.


Subject(s)
Amyloid beta-Peptides , DNA , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/chemistry , DNA/chemistry , Humans , Nanostructures/chemistry , Particle Size , Electrochemical Techniques/methods , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Alzheimer Disease/blood , Biosensing Techniques/methods , Solubility , Aptamers, Nucleotide/chemistry , Molecular Weight
14.
Anal Sci ; 40(10): 1809-1821, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38884905

ABSTRACT

Accurate and timely diagnosis of Alzheimer's disease (AD) is necessary to maximize the effectiveness of treatment and using biomarkers for diagnosis is attracting attention as a minimally invasive method with few side effects. Electrochemical immunosensor (EI) is a method that is in the spotlight in the medical and bioanalytical fields due to its portability and field usability. Here, we quantified four AD specific biomarkers using EIs based on enzyme immunoassay. We selected and developed quantitative methods for the biomarkers using screen-printed gold electrodes. For three biomarkers, quantification was performed using competition immunoassays in which antigen-antibody premix mixtures were applied to antigen-immobilized electrodes and the limit of detection (LOD) values were secured, 1.20 ng/ml, 1.30 ng/ml, and 1.74 ng/ml, respectively. For the other, a sandwich immunoassay using antibody pair was selected for quantification and LOD was also achieved as 0.077 ng/ml. All four biomarkers in buffer samples were successfully quantified and reliable R2 values were obtained, and reliable calibration curves were secured for three biomarkers in spiked human serum samples. The immunosensors developed and will be optimized are expected to be used in various fields, including detection of biomarkers for not only AD but also related diseases.


Subject(s)
Alzheimer Disease , Biomarkers , Electrochemical Techniques , Alzheimer Disease/diagnosis , Alzheimer Disease/blood , Humans , Biomarkers/blood , Biomarkers/analysis , Biosensing Techniques , Immunoenzyme Techniques/methods , Electrodes , Limit of Detection , Gold/chemistry , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/immunology
15.
J Appl Lab Med ; 9(4): 789-802, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38712812

ABSTRACT

BACKGROUND: Standardizing cerebrospinal fluid (CSF) laboratory protocols will improve the reliability and availability of clinical biomarker testing required for prescription of novel Alzheimer disease (AD) therapies. This study evaluated several preanalytical handling and storage factors common to ß-amyloid1-42 (Aß1-42), ß-amyloid1-40 (Aß1-40), and phosphorylated tau (pTau181) concentrations including storage at different temperatures, extended cap contact, various mixing methods, and multiple freeze-thaw cycles. METHODS: Aß1-42, Aß1-40, and pTau181 concentrations were measured using LUMIPULSE G1200 automated assays. Samples were collected in polypropylene tubes of various volumes. Sample cap-contact was evaluated by storing samples in upright and inverted positions at either 4°C for 1 week or -80°C for 1 month. To assess mixing methods, samples were freeze-thawed and mixed by inversion, vortex, horizontal roller, or unmixed prior to assay sampling. The impact of successive freeze-thaw cycles was assessed through freezing, thawing, and analyzing CSF samples. RESULTS: Short-term storage at 4°C did not affect Aß1-42, Aß1-40, or pTau181 measurements in any tube type. Tube cap contact affected Aß1-42 in 2.5 mL tubes and pTau181 levels in 10 mL tubes. No difference was observed between mixing methods. After 4 freeze-thaw cycles, Aß1-42 significantly decreased but Aß1-40 remained unchanged. Utilizing the Aß1-42/Aß1-40 ratio, Aß1-42 values normalized, maintaining ratio values within ±5% of baseline measurements. CONCLUSIONS: Storage of CSF at 4°C for 1 week or -80°C for 1 month did not significantly affect Aß1-42, Aß1-40, pTau181, or associated ratio measurements. Tube cap-contact impacted pTau181 and pTau181/Aß1-42 values in larger tubes. Mixing methods are equivalent. The Aß1-42/Aß1-40 ratio compensates for freeze-thaw variability up to 4 cycles.


Subject(s)
Amyloid beta-Peptides , Peptide Fragments , tau Proteins , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/analysis , Humans , tau Proteins/cerebrospinal fluid , tau Proteins/analysis , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/analysis , Specimen Handling/methods , Specimen Handling/instrumentation , Luminescent Measurements/methods , Luminescent Measurements/instrumentation , Luminescent Measurements/standards , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Biomarkers/cerebrospinal fluid , Freezing , Phosphorylation
16.
Talanta ; 275: 126167, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38710128

ABSTRACT

The expression of metabotropic glutamate receptor 5 (mGluR5) is subject to developmental regulation and undergoes significant changes in neuropsychiatric disorders and diseases. Visualizing mGluR5 by fluorescence imaging is a highly desired innovative technology for biomedical applications. Nevertheless, there are substantial problems with the chemical probes that are presently accessible. In this study, we have successfully developed a two-photon fluorogenic probe, mGlu-5-TP, based on the structure of mGluR5 antagonist 6-methyl-2-(phenylethynyl)pyridine (MPEP). Due to this antagonist-based probe selectively recognizes mGluR5, high expression of mGluR5 on living SH-SY5Y human neuroblastoma cells has been detected during intracellular inflammation triggered by lipopolysaccharides (LPS). Of particular significance, the probe can be employed along with two-photon fluorescence microscopy to enable real-time visualization of the mGluR5 in Aß fiber-treated neuronal cells, thereby establishing a connection to the progression of Alzheimer's disease (AD). These results revealed that the probe can be a valuable imaging tool for studying mGluR5-related diseases in the nervous system.


Subject(s)
Fluorescent Dyes , Neurons , Pyridines , Receptor, Metabotropic Glutamate 5 , Receptor, Metabotropic Glutamate 5/metabolism , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Humans , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Neurons/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Cell Line, Tumor , Lipopolysaccharides/pharmacology , Photons , Optical Imaging , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis
17.
Anal Chim Acta ; 1308: 342575, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38740448

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a prevalent neurodegenerative disease with no effective treatment. Efficient and rapid detection plays a crucial role in mitigating and managing AD progression. Deep learning-assisted smartphone-based microfluidic paper analysis devices (µPADs) offer the advantages of low cost, good sensitivity, and rapid detection, providing a strategic pathway to address large-scale disease screening in resource-limited areas. However, existing smartphone-based detection platforms usually rely on large devices or cloud servers for data transfer and processing. Additionally, the implementation of automated colorimetric enzyme-linked immunoassay (c-ELISA) on µPADs can further facilitate the realization of smartphone µPADs platforms for efficient disease detection. RESULTS: This paper introduces a new deep learning-assisted offline smartphone platform for early AD screening, offering rapid disease detection in low-resource areas. The proposed platform features a simple mechanical rotating structure controlled by a smartphone, enabling fully automated c-ELISA on µPADs. Our platform successfully applied sandwich c-ELISA for detecting the ß-amyloid peptide 1-42 (Aß 1-42, a crucial AD biomarker) and demonstrated its efficacy in 38 artificial plasma samples (healthy: 19, unhealthy: 19, N = 6). Moreover, we employed the YOLOv5 deep learning model and achieved an impressive 97 % accuracy on a dataset of 1824 images, which is 10.16 % higher than the traditional method of curve-fitting results. The trained YOLOv5 model was seamlessly integrated into the smartphone using the NCNN (Tencent's Neural Network Inference Framework), enabling deep learning-assisted offline detection. A user-friendly smartphone application was developed to control the entire process, realizing a streamlined "samples in, answers out" approach. SIGNIFICANCE: This deep learning-assisted, low-cost, user-friendly, highly stable, and rapid-response automated offline smartphone-based detection platform represents a good advancement in point-of-care testing (POCT). Moreover, our platform provides a feasible approach for efficient AD detection by examining the level of Aß 1-42, particularly in areas with low resources and limited communication infrastructure.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Enzyme-Linked Immunosorbent Assay , Paper , Smartphone , Alzheimer Disease/diagnosis , Alzheimer Disease/blood , Humans , Biomarkers/blood , Biomarkers/analysis , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/blood , Peptide Fragments/blood , Peptide Fragments/analysis , Lab-On-A-Chip Devices , Deep Learning , Automation , Microfluidic Analytical Techniques/instrumentation
18.
ACS Sens ; 9(5): 2605-2613, 2024 05 24.
Article in English | MEDLINE | ID: mdl-38718161

ABSTRACT

Several new lines of research have demonstrated that a significant number of amyloid-ß peptides found in Alzheimer's disease (AD) are truncated and undergo post-translational modification by glutaminyl cyclase (QC) at the N-terminal. Notably, QC's products of Abeta-pE3 and Abeta-pE11 have been active targets for investigational drug development. This work describes the design, synthesis, characterization, and in vivo validation of a novel PET radioligand, [18F]PB0822, for targeted imaging of QC. We report herein a simplified and robust chemistry for the synthesis of the standard compound, [19F]PB0822, and the corresponding [18F]PB0822 radioligand. The PET probe was developed with 99.9% radiochemical purity, a molar activity of 965 Ci.mmol-1, and an IC50 of 56.3 nM, comparable to those of the parent PQ912 inhibitor (62.5 nM). Noninvasive PET imaging showed that the probe is distributed in the brain 5 min after intravenous injection. Further, in vivo PET imaging with [18F]PB0822 revealed that AD 5XFAD mice harbor significantly higher QC activity than WT counterparts. The data also suggested that QC activity is found across different brain regions of the tested animals.


Subject(s)
Alzheimer Disease , Aminoacyltransferases , Positron-Emission Tomography , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Positron-Emission Tomography/methods , Aminoacyltransferases/metabolism , Aminoacyltransferases/antagonists & inhibitors , Animals , Mice , Fluorine Radioisotopes/chemistry , Brain/diagnostic imaging , Brain/metabolism , Brain/enzymology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/chemical synthesis , Biomarkers/metabolism , Humans , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Ligands
19.
Biosens Bioelectron ; 259: 116405, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38776801

ABSTRACT

Alzheimer's disease (AD) is affecting more and more people worldwide without the effective treatment, while the existed pathological mechanism has been confirmed barely useful in the treatment. Amyloid-ß peptide (Aß), a main component of senile plaque, is regarded as the most promising target in AD treatment. Aß clearance from AD brain seems to be a reliably therapeutic strategy, as the two exited drugs, GV-971 and aducanumab, are both developed based on it. However, doubt still exists. To exhaustive expound on the pathological mechanism of Aß, rigorous analyses on the concentrations and aggregation forms are essential. Thus, it is attracting broad attention these years. However, most of the sensors have not been used in pathological studies, as the lack of the bridge between analytical chemist and pathologists. In this review, we made a brief introduce on Aß-related pathological mechanism included in ß-amyloid hypothesis to elucidate the detection conditions of sensor methods. Furthermore, a summary of the sensor methods was made, which were based on Aß concentrations and form detections that have been developed in the past 10 years. As the greatest number of the sensors were built on fluorescent spectroscopy, electrochemistry, and Roman spectroscopy, detailed elucidation on them was made. Notably, the aggregation process is another important factor in revealing the progress of AD and developing the treatment methods, so the sensors on monitoring Aß aggregation processes were also summarized.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biosensing Techniques , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Humans , Biosensing Techniques/methods , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Spectrometry, Fluorescence/methods , Electrochemical Techniques/methods , Antibodies, Monoclonal, Humanized
20.
Talanta ; 275: 126111, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38657362

ABSTRACT

Sensitive, accurate, and straightforward biosensors are pivotal in the battle against Alzheimer's disease, particularly in light of the escalating patient population. These biosensors enable early adjunctive diagnosis, thereby facilitating prompt intervention, alleviating socioeconomic burdens, and preserving individual well-being. In this study, we introduce the development of a highly sensitive add-drop dual-microring resonant microfluidic sensing chip boasting a sensitivity of 188.11 nm/RIU, marking a significant 20.7% enhancement over single microring systems. Leveraging ultra-thin Parylene C for streamlined antibody immobilization and non-destructive removal, this platform facilitates the precise quantification of the Alzheimer's disease biomarker Aß42. Employing an immune sensing strategy that amplifies and captures antigen signals using Au-labeled antibodies, we achieve an exceptional limit of detection of 9.02 pg/mL. The designed microring-based microfluidic biosensor chip exhibits outstanding specificity and sensitivity for Aß42 in serum samples, offering a promising avenue for the early adjunctive diagnosis of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides , Biosensing Techniques , Peptide Fragments , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/blood , Biosensing Techniques/methods , Humans , Peptide Fragments/blood , Peptide Fragments/analysis , Peptide Fragments/immunology , Alzheimer Disease/diagnosis , Alzheimer Disease/blood , Limit of Detection , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Antibodies, Immobilized/immunology , Antibodies, Immobilized/chemistry , Gold/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL