Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Neurosci Lett ; 837: 137914, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39032802

ABSTRACT

ß-amyloid42 (Aß42) in Alzheimer's disease (AD) and orexin in narcolepsy are considered crucial biomarkers for diagnosis and therapeutic targets. Recently, orexin and Aß cerebral dynamics have been studied in both pathologies, but how they interact with each other remains further to be known. In this study, we investigated the reliability of using the correlation between orexin-A and Aß42 CSF levels as a candidate marker to explain the chain of events leading to narcolepsy or AD pathology. In order to test the correlation between these biomarkers, patients diagnosed with AD (n = 76), narcolepsy type 1 (NT1, n = 17), narcolepsy type 2 (NT2, n = 23) and healthy subjects (n = 91) were examined. Patients and healthy subjects underwent lumbar puncture between 8:00 and 10:00 am at the Neurology Unit of the University Hospital of Rome "Tor Vergata". CSF levels of Aß42, total-tau, phosphorylated-tau, and orexin-A were assessed. The results showed that CSF levels of Aß42 were significantly lower (p < 0.001) in AD (332.28 ± 237.36 pg/mL) compared to NT1 (569.88 ± 187.00 pg/mL), NT2 (691.00 ± 292.63 pg/mL) and healthy subjects (943.68 ± 198.12 pg/mL). CSF orexin-A levels were statistically different (p < 0.001) between AD (148.01 ± 29.49 pg/mL), NT1 (45.94 ± 13.63 pg/mL), NT2 (104.92 ± 25.55 pg/mL) and healthy subjects (145.18 ± 27.01 pg/mL). Moderate-severe AD patients (mini mental state examination < 21) showed the highest CSF orexin-A levels, whereas NT1 patients showed the lowest CSF orexin-A levels. Correlation between CSF levels of Aß42 and orexin-A was found only in healthy subjects (r = 0.26; p = 0.01), and not in narcolepsy or AD patients. This lack of correlation in both diseases may be explained by the pathology itself since the correlation between these two biomarkers is evident only in the healthy subjects. This study adds to the present literature by further documenting the interplay between orexinergic neurotransmission and cerebral Aß dynamics, possibly sustained by sleep.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Narcolepsy , Orexins , Peptide Fragments , Humans , Orexins/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Alzheimer Disease/cerebrospinal fluid , Narcolepsy/cerebrospinal fluid , Male , Female , Middle Aged , Aged , Peptide Fragments/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Adult , tau Proteins/cerebrospinal fluid
2.
Biomedica ; 44(Sp. 1): 42-46, 2024 05 31.
Article in English, Spanish | MEDLINE | ID: mdl-39079148

ABSTRACT

Alzheimer's disease is the leading cause of dementia worldwide and a critical public health problem. While deaths from cardiovascular diseases have decreased, those attributed to Alzheimer's disease have increased in recent years with no curative treatment to date. In this context, effective treatment development has become a global priority. Aducanumab is a human anti-amyloid ß monoclonal antibody approved by the FDA in June 2021 for the treatment of Alzheimer's disease but failed to show the expected clinical efficacy in phase III trials. This review analyzes the history of its controversial acceptance, implications, and prospects for future treatment.


La enfermedad de Alzheimer es la principal causa de demencia en todo el mundo y representa un importante problema de salud pública. Si bien las muertes por enfermedades cardiovasculares han disminuido, las atribuidas a la enfermedad de Alzheimer han aumentado en los últimos años y hasta la fecha no existe tratamiento curativo. Por este motivo, el desarrollo de un tratamiento eficaz se ha convertido en una prioridad mundial. Aducanumab es un anticuerpo monoclonal anti-amiloide ß humano aprobado para el tratamiento de la enfermedad de Alzheimer en junio de 2021 por la FDA, sin la eficacia clínica esperada en los ensayos de fase III. Esta revisión analiza la historia de su controvertida aceptación, implicaciones y perspectivas para el tratamiento futuro.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal, Humanized , Drug Approval , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Alzheimer Disease/drug therapy , United States
3.
ACS Appl Bio Mater ; 7(4): 2218-2239, 2024 04 15.
Article in English | MEDLINE | ID: mdl-38527228

ABSTRACT

The prompt detection of diseases hinges on the accessibility and the capability to identify relevant biomarkers. The integration of aptamers and the incorporation of nanomaterials into signal transducers have not only expedited but also enhanced the development of nanoaptasensors, enabling heightened sensitivity and selectivity. Here, the bimetallic nickel-cobalt-porphyrin metal-organic framework ((Ni + Cu)TPyP MOF) is regarded as an electron mediator, immobilization platform for an Alzheimer aptamer and to increase the electrochemical signal for the detection of the main biomarker of Alzheimer's disease (AD), amyloid ß (Aß-42). Furthermore, the ((Ni + Cu)TPyP MOF) was combined with reduced graphene oxide (rGO) and gold nanoparticles (AuNPs), on a gold electrode (GE) to provide an efficient interface for immobilizing aptamer strands. Concurrently, the incorporation of rGO and AuNPs imparts enhanced electrical conductivity and efficacious catalytic activity, establishing them as adept electrochemical indicators. Owing to the superior excellent electrical conductivity of rGO and AuNPs, coupled with the presence of ample mesoporous channels and numerous Ni and Cu metal sites within (Ni + Cu)TPyP MOF, this nanostructure with abundant functional groups is proficient in immobilizing a substantial quantity of aptamer. These interactions are achieved through robust π-π stacking and electrostatic interactions, alongside the high affinity between the thiol group of the aptamer and AuNPs concurrently. The as-prepared ternary (Au@(Ni + Cu)TPyP MOF/rGO) nanostructure electrode exhibited an enhancement in its electrochemically active surface area of about 7 times, compared with the bare electrode and the Aß-42 redox process is highly accelerated, so the peak currents are significantly higher than those obtained with bare GE substrate. Under the optimized conditions, the designed aptasensor had the quantitative detection of Aß-42 with a low detection limit of 48.6 fg mL-1 within the linear range of 0.05 pg mL-1 to 5 ng mL-1 by differential pulse voltammetry (DPV), accompanied by precise reproducibility, satisfactory stability (95.6% of the initial activity after 10 days), and minimal impact of interfering agents. Recorded results in human blood plasma demonstrated the high efficacy of porphyrin MOF system sensing even in the clinical matrix. The great performance of this aptasensor indicates that our new design of Au@(Ni + Cu)TPyP MOF/rGO nanostructure provides more opportunities for the detection of chemical signals in early diagnosis of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Aptamers, Nucleotide , Biosensing Techniques , Graphite , Metal Nanoparticles , Humans , Gold/chemistry , Amyloid beta-Peptides , Metal Nanoparticles/chemistry , Reproducibility of Results , Aptamers, Nucleotide/chemistry , Electrochemical Techniques/methods , Biosensing Techniques/methods
4.
Amino Acids ; 55(12): 1991-1997, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37904049

ABSTRACT

Alzheimer's disease is characterized by the presence of senile plaques composed of ß-amyloid peptide (Aß) aggregates with toxic effects that are still not fully understood. Recently, it was discovered that Aß(1-42) fibrils possess catalytic activity on acetylcholine hydrolysis. Catalytic amyloids are an emerging and exciting field of research. In this study, we examined the catalytic activity of the fibrils formed by Aß(1-40), the most abundant Aß variant, on acetylcholine hydrolysis. Our findings reveal that Aß(1-40) fibrils exhibit moderate enzymatic activity, indicating that natural peptide aggregates could serve as biocatalysts and provide new insights into the potential role of Aß in neurological disorders.


Subject(s)
Acetylcholine , Alzheimer Disease , Humans , Hydrolysis , Amyloid beta-Peptides , Peptide Fragments/chemistry , Amyloid
5.
Biomolecules ; 13(9)2023 08 24.
Article in English | MEDLINE | ID: mdl-37759689

ABSTRACT

N-nitrosodiethylamine (NDEA) is a potential carcinogen known to cause liver tumors and chronic inflammation, diabetes, cognitive problems, and signs like Alzheimer's disease (AD) in animals. This compound is classified as probably carcinogenic to humans. Usual sources of exposure include food, beer, tobacco, personal care products, water, and medications. AD is characterized by cognitive decline, amyloid-ß (Aß) deposit, tau hyperphosphorylation, and cell loss. This is accompanied by neuroinflammation, which involves release of microglial cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin 1ß (IL-1ß), by nuclear factor kappa B (NF-κB) upregulation; each are linked to AD progression. Weak PI3K/Akt insulin-signaling inhibits IRS-1 phosphorylation, activates GSK3ß and promotes tau hyperphosphorylation. Metformin, an antihyperglycemic agent, has potent anti-inflammatory efficacy. It reduces proinflammatory cytokines such as IL-6, IL-1ß, and TNF-α via NF-κB inhibition. Metformin also reduces reactive oxidative species (ROS) and modulates cognitive disorders reported due to brain insulin resistance links. Our study examined how NDEA affects spatial memory in Wistar rats. We found that all NDEA doses tested impaired memory. The 80 µg/kg dose of NDEA increased levels of Aß1-42, TNF-α, and IL-6 in the hippocampus, which correlated with memory loss. Nonetheless, treatment with 100 mg/kg of metformin attenuated the levels of pro-inflammatory cytokines and Aß1-42, and enhanced memory. It suggests that metformin may protect against NDEA-triggered memory issues and brain inflammation.


Subject(s)
Alzheimer Disease , Metformin , Animals , Rats , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Carcinogens , Cytokines , Diethylnitrosamine , Hippocampus , Interleukin-6 , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Metformin/pharmacology , Metformin/therapeutic use , NF-kappa B , Phosphatidylinositol 3-Kinases , Rats, Wistar , Tumor Necrosis Factor-alpha
6.
IBRO Neurosci Rep ; 14: 264-272, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36926592

ABSTRACT

Melatonin is a hormone secreted by the pineal gland, it can be associated with circadian rhythms, aging and neuroprotection. Melatonin levels are decreased in sporadic Alzheimer's disease (sAD) patients, which suggests a relationship between the melatonergic system and sAD. Melatonin may reduce inflammation, oxidative stress, TAU protein hyperphosphorylation, and the formation of ß-amyloid (Aß) aggregates. Therefore, the objective of this work was to investigate the impact of treatment with 10 mg/kg of melatonin (i.p) in the animal model of sAD induced by the intracerebroventricular (ICV) infusion of 3 mg/kg of streptozotocin (STZ). ICV-STZ causes changes in the brain of rats similar to those found in patients with sAD. These changes include; progressive memory decline, the formation of neurofibrillary tangles, senile plaques, disturbances in glucose metabolism, insulin resistance and even reactive astrogliosis characterized by the upregulation of glucose levels and glial fibrillary acidic protein (GFAP). The results show that ICV-STZ caused short-term spatial memory impairment in rats after 30 days of STZ infusion without locomotor impairment which was evaluated on day 27 post-injury. Furthermore, we observed that a prolonged 30-day treatment with melatonin can improve the cognitive impairment of animals in the Y-maze test, but not in the object location test. Finally, we demonstrated that animals receiving ICV-STZ have high levels of Aß and GFAP in the hippocampus and that treatment with melatonin reduces Aß levels but does not reduce GFAP levels, concluding that melatonin may be useful to control the progression of amyloid pathology in the brain.

7.
Nutrients ; 15(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36839291

ABSTRACT

Currently, there is an increasing number of people with mild cognitive (MCI) impairment and dementia (D). In the present work we studied the role of tau protein, ß-amyloid, LPS (lipopolysaccharide), and curli protein of elderly adults with MCI or D and the contribution of gut microbiota. Four groups were studied: young subjects, healthy adults older than 60 years (A), elderly adults with MCI (MCI), and elderly adults with dementia (D). A preclinical study was conducted in old male Wistar rats to evaluate the impact of gut microbiota on curli protein abundance in feces and brain. The results showed that with increasing age, tau protein, ß-amyloid, and LPS significantly increased in serum during MCI and D, and this was associated with an increase in the abundance of E. coli that synthesize the amyloid protein curli, that may promote the aggregation of amyloid proteins. Rats showed a clear increase in the abundance of curli protein in the brain during aging. Thus, cognitive impairment and dementia are in part due to an alteration in the gut microbiota-brain axis via increase in curli protein and LPS leading to an increase in tau and ß-amyloid protein.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Gastrointestinal Microbiome , Male , Rats , Animals , Amyloid beta-Peptides/metabolism , Lipopolysaccharides , Escherichia coli/metabolism , Rats, Wistar , Aging , Brain/metabolism , Feces
8.
Pharmaceuticals (Basel) ; 16(1)2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36678626

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder whose pathophysiology includes the abnormal accumulation of proteins (e.g., ß-amyloid), oxidative stress, and alterations in neurotransmitter levels, mainly acetylcholine. Here we present a comparative study of the effect of extracts obtained from endemic Argentinian species of valerians, namely V. carnosa Sm., V. clarionifolia Phil. and V. macrorhiza Poepp. ex DC from Patagonia and V. ferax (Griseb.) Höck and V. effusa Griseb., on different AD-related biological targets. Of these anxiolytic, sedative and sleep-inducing valerians, V. carnosa proved the most promising and was assayed in vivo. All valerians inhibited acetylcholinesterase (IC50 between 1.08-12.69 mg/mL) and butyrylcholinesterase (IC50 between 0.0019-1.46 mg/mL). They also inhibited the aggregation of ß-amyloid peptide, were able to chelate Fe2+ ions, and exhibited a direct relationship between antioxidant capacity and phenolic content. Moreover, V. carnosa was able to inhibit human monoamine oxidase A (IC50: 0.286 mg/mL (0.213-0.384)). A daily intake of aqueous V. carnosa extract by male Swiss mice (50 and 150 mg/kg/day) resulted in anxiolytic and antidepressant-like behavior and improved spatial memory. In addition, decreased AChE activity and oxidative stress markers were observed in treated mouse brains. Our studies contribute to the development of indigenous herbal medicines as therapeutic agents for AD.

9.
Front Cell Neurosci ; 16: 987212, 2022.
Article in English | MEDLINE | ID: mdl-36212691

ABSTRACT

Down syndrome (DS) arises from the triplication of human chromosome 21 and is considered the most common genetic cause of intellectual disability. Glial cells, specifically astroglia and microglia, display pathological alterations that might contribute to DS neuropathological alterations. Further, in middle adulthood, people with DS develop clinical symptoms associated with premature aging and Alzheimer's disease (AD). Overexpression of the amyloid precursor protein (APP) gene, encoded on chromosome 21, leads to increased amyloid-ß (Aß) levels and subsequent formation of Aß plaques in the brains of individuals with DS. Amyloid-ß deposition might contribute to astroglial and microglial reactivity, leading to neurotoxic effects and elevated secretion of inflammatory mediators. This review discusses evidence of astroglial and microglial alterations that might be associated with the AD continuum in DS.

10.
Nanomedicine ; 44: 102569, 2022 08.
Article in English | MEDLINE | ID: mdl-35595016

ABSTRACT

The ß-amyloid (Aß) peptide is one of the key etiological agents in Alzheimer's disease (AD). The in vivo detection of Aß species is challenging in all stages of the illness. Currently, the development of fluorescent probes allows the detection of Aß in animal models in the near-infrared region (NIR). However, considering future applications in biomedicine, it is relevant to develop strategies to improve detection of amyloid aggregates using NIR probes. An innovative approach to increase the fluorescence signal of these fluorophores is the use of plasmonic gold nanoparticles (surface-enhanced fluorescence effect). In this work, we improved the detection of Aß aggregates in C. elegans and mouse models of AD by co-administering functionalized gold nanorods (GNRs-PEG-D1) with the fluorescent probes CRANAD-2 or CRANAD-58, which bind selectively to different amyloid species (soluble and insoluble). This work shows that GNRs improve the detection of Aß using NIR probes in vivo.


Subject(s)
Alzheimer Disease , Metal Nanoparticles , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Animals , Caenorhabditis elegans , Fluorescent Dyes/chemistry , Gold , Metal Nanoparticles/chemistry , Mice
11.
J Biomol Struct Dyn ; 40(20): 9602-9612, 2022.
Article in English | MEDLINE | ID: mdl-34042019

ABSTRACT

In this study, comparable molecular dynamic (MD) simulations of 1.2 microseconds were performed to clarify the prevention of the ß-amyloid peptide (Aß1-42) aggregation by cucurbit[7]uril (CB[7]). The accumulation of this peptide in the brain is one of the most harmful in Alzheimer's disease. The inhibition mechanism of Aß1-42 aggregation by different molecules is attributed to preventing of Aß1-42 conformational transition from α-helix to the ß-sheet structure. However, our structural analysis shows that the pure water and aqueous solution of the CB[7] denature the native Aß1-42 α-helix structure forming different compactness and unfolded conformations, not in ß-sheet form. On the other hand, in the three CB[7]@Aß1-42 complexes, it was observed the encapsulation of N-terminal (Asp1), Lys16, and Val36 by CB[7] along the MD trajectory, and not with aromatic residues as suggested by the literature. Only in one CB[7]@Aß1-42 complex was observed stable Asp23-Lys28 salt bridge with an average distance of 0.36 nm. All CB[7]@Aß1-42 complexes are very stable with binding free energy lowest than ∼-50 kcal/mol between the CB[7] and Aß1-42 monomer from MM/PBSA calculation. Therefore, herein we show that the mechanism of the prevention of elongation protofibril by CB[7] is due to the disruption of the Asp23-Lys28 salt bridge and steric effects of CB[7]@Aß1-42 complex with the fibril lattice, and not due to the transition from α-helix to ß-sheet following the dock-lock mechanism.Communicated by Ramaswamy H. Sarma.


Subject(s)
Amyloid beta-Peptides , Humans , Amyloid beta-Peptides/chemistry , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Protein Conformation, beta-Strand
12.
Int J Mol Sci ; 21(20)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050466

ABSTRACT

Amyloid-beta oligomers (AßO) have been proposed as the most potent neurotoxic and inflammation inducers in Alzheimer's disease (AD). AßO contribute to AD pathogenesis by impairing the production of several cytokines and inflammation-related signaling pathways, such as the Janus kinases/signal transducer of transcription factor-3 (JAK/STAT3) pathway. STAT3 modulates glial activation, indirectly regulates Aß deposition, and induces cognitive decline in AD transgenic models. However, in vivo studies using an AßO microinjection rat model have not yet explored STAT3 role. The main purpose of this study was to elucidate if a single microinjection of AßO could promote an increased expression of STAT3 in glial cells favoring neuroinflammation and neurodegeneration. We designed a model of intrahippocampal microinjection and assessed glial activation, cytokines production, STAT3 expression, and neurodegeneration in time. Our results showed robust expression of STAT3 in glial cells (mainly in astrocytes) and neurons, correlating with neuronal death in response to AßO administration. A STAT3 inhibition assay conducted in rat primary hippocampal cultures, suggested that the induction of the transcription factor by AßO in astrocytes leads them to an activation state that may favor neuronal death. Notwithstanding, pharmacological inhibition of the JAK2/STAT3 pathway should be focused on astrocytes because it is also essential in neurons survival. Overall, these findings strongly suggest the participation of STAT3 in the development of neurodegeneration.


Subject(s)
Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Gliosis/etiology , Gliosis/metabolism , Neurons/metabolism , STAT3 Transcription Factor/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Biomarkers , Cell Death , Disease Models, Animal , Disease Susceptibility , Fluorescent Antibody Technique , Gliosis/pathology , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Protein Aggregates , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Multimerization , Rats , STAT3 Transcription Factor/genetics
13.
Bioorg Chem ; 103: 104201, 2020 10.
Article in English | MEDLINE | ID: mdl-32890999

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative process that compromises cognitive functions. The physiopathology of AD is multifactorial and is mainly supported by the cholinergic and amyloid hypotheses, which allows the identification the fundamental role of some markers, such as the enzymes acetylcholinesterase (AChE) and ß-secretase (BACE-1), and the ß-amyloid peptide (Aß). In this work, we prepared a series of chalcones and 2'-aminochalcones, which were tested against AChE and BACE-1 enzymes and on the aggregation of Aß. All compounds inhibited AChE activity with different potencies. We have found that the majority of chalcones having the amino group are able to inhibit BACE-1, which was not observed for chalcones without this group. The most active compound is the one derived from 2,3-dichlorobenzaldeyde, having an IC50 value of 2.71 µM. A molecular docking study supported this result, showing a good interaction of the amino group with aspartic acid residues of the catalytic diade of BACE-1. Thioflavin-T fluorescence emission is reduced in 30 - 40%, when Aß42 is incubated in the presence of some chalcones under aggregation conditions. In vitro cytotoxicity and in silico prediction of pharmacokinetic properties were also conducted in this study.


Subject(s)
Chalcones/pharmacology , Cholinesterase Inhibitors/pharmacology , Protease Inhibitors/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , Chalcones/chemical synthesis , Chalcones/metabolism , Chalcones/pharmacokinetics , Chlorocebus aethiops , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Electrophorus , Humans , Mice , Molecular Docking Simulation , Peptide Fragments/metabolism , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Protein Binding , Protein Multimerization/drug effects , Vero Cells
14.
Curr Pharm Des ; 26(12): 1365-1376, 2020.
Article in English | MEDLINE | ID: mdl-31931693

ABSTRACT

The ß-amyloid peptide (1-42) is a molecule capable of aggregating into neurotoxic structures that have been implicated as potential etiological factors of Alzheimer's Disease. The aim of this study was to evaluate the inhibition of ß-amyloid aggregation of ethyl acetate and ethanolic extracts obtained from Ugni molinae leaves on neurotoxic actions of ß-amyloid aggregates. Chemical analyses were carried out with the extracts in order to determine their phenolic profile and its quantification. Both extracts showed a tendency to reduce neuronal deaths caused by ß-amyloid. This tendency was inversely proportional to the evaluated concentrations. Moreover, the effect of EAE and ETE on ß-amyloid aggregation was studied by fluorimetric T Thioflavin assay and transmission electronic microscopy (TEM); the extracts showed a modulation in the aggregation process. Partly, it is believed that these effects can be attributed to the polyphenolic compounds present in the extracts.


Subject(s)
Alzheimer Disease , Myrtaceae , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Humans , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phenols/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry
15.
Electron. j. biotechnol ; Electron. j. biotechnol;40: 1-9, July. 2019. tab, graf, ilus
Article in English | LILACS | ID: biblio-1053195

ABSTRACT

BACKGROUND: Microalgae are aquatic chlorophyll-containing organisms comprising unicellular microscopic forms, and their biomasses are potential sources of bioactive compounds, biofuels and food-based products. However, the neuroprotective effects of microalgal biomass have not been fully explored. In this study, biomass from two Chlorella species was characterized, and their antioxidant, anticholinesterase and anti-amyloidogenic activities were investigated. RESULTS: GC­MS analysis of the extracts revealed the presence of some phenols, sterols, steroids, fatty acids and terpenes. Ethanol extract of Chlorella sorokiniana (14.21 mg GAE/g) and dichloromethane extract of Chlorella minutissima (20.65 mg QE/g) had the highest total phenol and flavonoid contents, respectively. All the extracts scavenged 2,2-diphenyl-1-picrylhydrazyl, 2,2-azinobis (3-ethylbenzothiazoline-6-sulfonate) and hydroxyl radicals. The highest metal chelating activity of the extracts was observed in the ethanol extracts of C. minutissima (102.60 µg/mL) and C. sorokiniana (107.84 µg/mL). Furthermore, the cholinesterase inhibitory activities of the extracts showed that ethanol extract of C. sorokiniana (13.34 µg/mL) exhibited the highest acetylcholinesterase inhibitory activity, while dichloromethane extract of C. minutissima (11.78 µg/mL) showed the highest butyrylcholinesterase inhibitory activity. Incubation of the ß-amyloid protein increased the aggregation of amyloid fibrils after 96 h. However, ethanol extract of C. sorokiniana and C. minutissima inhibited further aggregation of Aß1­42 and caused disaggregation of matured protein fibrils compared to the control. This study reveals the modulatory effects of C. sorokiniana and C. minutissima extracts on some mediators of Alzheimer's disease and provides insights into their potential benefits as functional food, nutraceutics or therapeutic agent for the management of this neurodegenerative disease.


Subject(s)
Chlorella/chemistry , Cholinesterase Inhibitors/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Antioxidants/pharmacology , Phenols/analysis , Steroids/analysis , Sterols/analysis , Terpenes/analysis , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cholinesterase Inhibitors/chemistry , Spectroscopy, Fourier Transform Infrared , Neuroprotective Agents , Biomass , Ethanol , Fatty Acids/analysis , Microalgae , Alzheimer Disease/prevention & control , Amyloid/drug effects , Gas Chromatography-Mass Spectrometry , Antioxidants/chemistry
16.
Neurosci Lett ; 708: 134339, 2019 08 24.
Article in English | MEDLINE | ID: mdl-31226364

ABSTRACT

This study aimed to evaluate behavioral and neurochemical parameters in adult (180-day-old) and young (60-day-old) rats subjected to sepsis. Sepsis was induced by cecal ligation and perforation (CLP). Thirty days after surgery, behavioral tests were performed, and the ß-amyloid content, oxidative damage, and cytokine levels were measured in the hippocampus and prefrontal cortex. In both adult and young rats, sepsis impaired the inhibitory avoidance task performance and increased immobility time in the forced swimming test. However, the adult septic rats had a higher immobility time compared to the young rats. Both sepsis and aging induced brain inflammation and oxidative damage and increased Aß content. Sepsis along with aging had additive effects on hippocampal interleukin-1 levels and prefrontal carbonyl levels. Taken together, our results suggest that age has a minor influence on brain inflammation and behavioral alterations observed in septic rats.


Subject(s)
Behavior, Animal , Sepsis/metabolism , Sepsis/psychology , Age Factors , Amyloid beta-Peptides/metabolism , Animals , Cognition , Depression/psychology , Hippocampus/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Oxidative Stress , Prefrontal Cortex/metabolism , Rats , Time Factors
17.
J Pept Sci ; 25(6): e3170, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31006946

ABSTRACT

Glutamic acid-rich peptides are crucial to a variety of biological processes, including glutamatergic neurotransmission and immunological defense. Glutamic acid sequences often exhibit unusual organization into ß2 -type sheets, where bifurcated H bonds formed between glutamic acid side chains and NH in amide bonds on adjacent ß-strands play a paramount role for stabilizing the molecular assembly. Herein, we investigate the self-assembly and supramolecular structure of simplified models consisting of alternating glutamic acid/phenylalanine residues. Small-angle X-ray scattering and atomic force microscopy show that the aggregation pathway is characterized by the formation of small oligomers, followed by coalescence into nanofibrils and nanotapes. Amyloidogenic features are further demonstrated through fiber X-ray diffraction, which reveal molecular packing according to cross-ß patterns, where ß-strands appear perpendicularly oriented to the long axis of nanofibrils and nanotapes. Nanoscale infrared spectroscopy from individual nanoparticles on dried samples shows a remarkable decrease of ß2 -sheet content, accompanied by growth of standard ß-sheet fractions, indicating a ß2 -to-ß1 transition as a consequence of the release of solvent from the interstices of peptide assemblies. Our findings highlight the key role played by water molecules in mediating H-bond formation in ß2 -sheets commonly found in amyloidogenic glutamic acid-rich aggregates.


Subject(s)
Amyloid/chemistry , Glutamic Acid/chemistry , Nanostructures/chemistry , Microscopy, Atomic Force , Models, Molecular , Protein Conformation, beta-Strand , Scattering, Small Angle , X-Ray Diffraction
18.
Brain Behav Immun ; 79: 284-293, 2019 07.
Article in English | MEDLINE | ID: mdl-30797044

ABSTRACT

The consumption of saturated fatty acids is one of the leading risk factors for Alzheimer's Disease (AD) development. Indeed, the short-term consumption of a high-fat diet (HFD) is related to increased inflammatory signals in the hippocampus; however, the potential molecular mechanisms linking it to AD pathogenesis are not fully elucidated. In our study, we investigated the effects of short-term HFD feeding (within 3, 7 and 10 days) in AD markers and neuroinflammation in the hippocampus of mice. The short period of HFD increased fasting glucose and HOMA-IR. Also, mice fed HFD increased the protein content of ß-Amyloid, pTau, TNFα, IL1ß, pJNK, PTP1B, peIF2α, CHOP, Caspase3, Cleaved-Caspase3 and Alzheimer-related genes (Bax, PS1, PEN2, Aph1b). At 10 days, both neuronal (N2a) and microglial (BV2) cells presented higher expression of inflammatory and apoptotic genes when stimulated with palmitate. These findings suggest that a short period of consumption of a diet rich in saturated fat is associated with activation of inflammatory, ER stress and apoptotic signals in the hippocampus of young mice.


Subject(s)
Alzheimer Disease/etiology , Diet, High-Fat/adverse effects , Hippocampus/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Apoptosis , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Inflammation/metabolism , Interleukin-1beta , Mice , Mice, Transgenic , Microglia/metabolism , Neurons/metabolism , Phosphorylation , Signal Transduction/drug effects , Temporal Lobe/metabolism , tau Proteins/metabolism
19.
Curr Radiopharm ; 12(1): 58-71, 2019.
Article in English | MEDLINE | ID: mdl-30605068

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common form of dementia. Neuroimaging methods have widened the horizons for AD diagnosis and therapy. The goals of this work are the synthesis of 2-(3-fluoropropyl)-6-methoxynaphthalene (5) and its [18F]-radiolabeled counterpart ([18F]Amylovis), the in silico and in vitro comparative evaluations of [18F]Amylovis and [11C]Pittsburg compound B (PIB) and the in vivo preclinical evaluation of [18F]Amylovis in transgenic and wild mice. METHODS: Iron-catalysis cross coupling reaction, followed by fluorination and radiofluorination steps were carried out to obtain 5 and 18F-Amylovis. Protein/Aß plaques binding, biodistribution, PET/CT Imaging and immunohistochemical studies were conducted in healthy/transgenic mice. RESULTS: The synthesis of 5 was successful obtained. Comparative in silico studies predicting that 5 should have affinity to the Aß-peptide, mainly through π-π interactions. According to a dynamic simulation study the ligand-Aß peptide complexes are stable in simulation-time (ΔG = -5.31 kcal/mol). [18F]Amylovis was obtained with satisfactory yield, high radiochemical purity and specific activity. The [18F]Amylovis log Poct/PBS value suggests its potential ability for crossing the blood brain barrier (BBB). According to in vitro assays, [18F]Amylovis has an adequate stability in time. Higher affinity to Aß plaques were found for [18F]Amylovis (Kd 0.16 nmol/L) than PIB (Kd 8.86 nmol/L) in brain serial sections of 3xTg-AD mice. Biodistribution in healthy mice showed that [18F]Amylovis crosses the BBB with rapid uptake (7 %ID/g at 5 min) and good washout (0.11±0.03 %ID/g at 60 min). Comparative PET dynamic studies of [18F]Amylovis in healthy and transgenic APPSwe/PS1dE9 mice, revealed a significant high uptake in the mice model. CONCLUSION: The in silico, in vitro and in vivo results justify that [18F]Amylovis should be studied as a promissory PET imaging agent to detect the presence of Aß senile plaques.


Subject(s)
Carbon Radioisotopes/chemistry , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/pharmacology , Naphthalenes/chemistry , Neuroimaging/methods , Plaque, Amyloid/diagnostic imaging , Positron Emission Tomography Computed Tomography , Radiochemistry/methods , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology , Animals , Computer Simulation , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tissue Distribution
20.
Bio Protoc ; 9(20): e3397, 2019 Oct 20.
Article in English | MEDLINE | ID: mdl-33654898

ABSTRACT

Animal models have promoted meaningful contribution to science including Alzheimer's disease (AD) research. Several animal models for AD have been used, most of them related to genetic mutations observed in familial AD. However, sporadic form of AD, also named late-onset is the most frequent form of the disease, which is multifactorial, being influenced by genetic, environmental and lifestyle factors. Here, we describe a protocol of an AD-like pathology of the sporadic form using Wistar rats by a single bilateral intracerebroventricular (icv) injection of streptozotocin (STZ, 2 mg/kg). Icv injection of STZ induces brain resistance to insulin and other pathological alterations related to those observed in AD, such as cognitive impairment and accumulation of phosphorylated tau protein and ß-amyloid in the brain. Thus, icv injection of STZ is a useful tool to investigate the pathological mechanisms and the metabolic alterations involved in AD and to propose new therapeutic approaches and neuroprotective drugs.

SELECTION OF CITATIONS
SEARCH DETAIL