Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39000210

RESUMEN

Neurodegeneration diseases (NDs) are a group of complex diseases primarily characterized by progressive loss of neurons affecting mental function and movement. Oxidative stress is one of the factors contributing to the pathogenesis of NDs, including Alzheimer's disease (AD). These reactive species disturb mitochondrial function and accelerate other undesirable conditions including tau phosphorylation, inflammation, and cell death. Therefore, preventing oxidative stress is one of the imperative methods in the treatment of NDs. To accomplish this, we prepared hexane and ethyl acetate extracts of Anethum graveolens (dill) and identified the major phyto-components (apiol, carvone, and dihydrocarvone) by GC-MS. The extracts and major bioactives were assessed for neuroprotective potential and mechanism in hydrogen peroxide-induced oxidative stress in the SH-SY5Y neuroblastoma cell model and other biochemical assays. The dill (extracts and bioactives) provided statistically significant neuroprotection from 0.1 to 30 µg/mL by mitigating ROS levels, restoring mitochondrial membrane potential, reducing lipid peroxidation, and reviving the glutathione ratio. They moderately inhibited acetylcholine esterase (IC50 dill extracts 400-500 µg/mL; carvone 275.7 µg/mL; apiole 388.3 µg/mL), displayed mild anti-Aß1-42 fibrilization (DHC 26.6%) and good anti-oligomerization activity (>40% by dill-EA, carvone, and apiole). Such multifactorial neuroprotective displayed by dill and bioactives would help develop a safe, low-cost, and small-molecule drug for NDs.


Asunto(s)
Anethum graveolens , Neuroblastoma , Fármacos Neuroprotectores , Estrés Oxidativo , Extractos Vegetales , Semillas , Humanos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química , Línea Celular Tumoral , Extractos Vegetales/farmacología , Extractos Vegetales/química , Neuroblastoma/metabolismo , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Estrés Oxidativo/efectos de los fármacos , Anethum graveolens/química , Semillas/química , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno , Fitoquímicos/farmacología , Fitoquímicos/química , Supervivencia Celular/efectos de los fármacos , Acetilcolinesterasa/metabolismo
2.
Alzheimers Dement ; 19(6): 2479-2496, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36515320

RESUMEN

INTRODUCTION: Recent published clinical trial safety data showed that 41% of Alzheimer patients experienced amyloid-related imaging abnormalities (ARIA), marks of microhemorrhages and edema in the brain, following administration of Biogen's Aduhelm/aducanumab (amino acids 3-7 of the Aß peptide). Similarly, Janssen/Pfizer's Bapineuzumab (amino acids 1-5 of the Aß peptide) and Roche's Gantenerumab (amino acids 2-11/18-27 of the Aß peptide) also displayed ARIA in clinical trials, including microhemorrhage and focal areas of inflammation or vasogenic edema, respectively. The molecular mechanisms underlying ARIA caused by therapeutic anti-Aß antibodies remain largely unknown, however, recent reports demonstrated that therapeutic anti-prion antibodies activate neuronal allergenic proteomes following cross-linking cellular prion protein. METHODS: Here, we report that treatment of human induced pluripotent stem cells- derived neurons (HSCN) from a non-demented donor, co-cultured with human primary microglia with anti-Aß1-6, or anti-Aß17-23 antibodies activate a significant number of allergenic-related proteins as assessed by mass spectrometry. RESULTS: Interestingly, a large proportion of the identified proteins included cytokines such as interleukin (IL)-4, IL-12, and IL-13 suggesting a type-1 hypersensitivity response. Following flow cytometry analysis, several proinflammatory cytokines were significantly elevated following anti-Aß1-6, or anti-Aß17-23 antibody treatment. DISCUSSION: These results justify further and more robust investigation of the molecular mechanisms of ARIA during immunotherapy study trials of AD. HIGHLIGHTS: Allergenic-related proteins are often linked with Alzheimer's disease (AD). We investigated the effects of amyloid beta (Aß) immunotherapy on stem cell derived neurons and primary neuronal cells co-cultured with microglia. Anti-Aß antibody treatment of neurons or neurons co-cultured with microglia led to activation of a substantial number of allergenic-related genes. These allergenic-related genes are associated with endothelial dysfunction possibly responsible for ARIA.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/genética , Citocinas , Neuronas/metabolismo , Aminoácidos
3.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-35008983

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease with a multifactorial etiology. A multitarget treatment that modulates multifaceted biological functions might be more effective than a single-target approach. Here, the therapeutic efficacy of combination treatment using anti-Aß antibody NP106 and curcumin analog TML-6 versus monotherapy was investigated in an APP/PS1 mouse model of AD. Our data demonstrate that both combination treatment and monotherapy attenuated brain Aß and improved the nesting behavioral deficit to varying degrees. Importantly, the combination treatment group had the lowest Aß levels, and insoluble forms of Aß were reduced most effectively. The nesting performance of APP/PS1 mice receiving combination treatment was better than that of other APP/PS1 groups. Further findings indicate that enhanced microglial Aß phagocytosis and lower levels of proinflammatory cytokines were concurrent with the aforementioned effects of NP106 in combination with TML-6. Intriguingly, combination treatment also normalized the gut microbiota of APP/PS1 mice to levels resembling the wild-type control. Taken together, combination treatment outperformed NP106 or TML-6 monotherapy in ameliorating Aß pathology and the nesting behavioral deficit in APP/PS1 mice. The superior effect might result from a more potent modulation of microglial function, cerebral inflammation, and the gut microbiota. This innovative treatment paradigm confers a new avenue to develop more efficacious AD treatments.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/deficiencia , Anticuerpos Monoclonales/farmacología , Curcumina/farmacología , Presenilina-1/deficiencia , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores , Curcumina/análogos & derivados , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Inmunohistoquímica , Ratones , Ratones Noqueados , Microbiota/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Terapia Molecular Dirigida , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/patología
4.
Mar Drugs ; 19(8)2021 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-34436249

RESUMEN

Alzheimer's disease (AD) is an irreversible and progressive brain disorder that slowly destroys memory and thinking skills, and, eventually, the ability to perform simple tasks. As the aging population continues to increase exponentially, AD has become a big concern for society. Therefore, neuroprotective compounds are in the spotlight, as a means to tackle this problem. On the other hand, since it is believed-in many cultures-that marine organisms in an individual diet cannot only improve brain functioning, but also slow down its dysfunction, many researchers have focused on identifying neuroprotective compounds from marine resources. The fact that the marine environment is a rich source of structurally unique and biologically and pharmacologically active compounds, with unprecedented mechanisms of action, marine macroorganisms, such as tunicates, corals, sponges, algae, as well as microorganisms, such as marine-derived bacteria, actinomycetes, and fungi, have been the target sources of these compounds. Therefore, this literature review summarizes and categorizes various classes of marine-derived compounds that are able to inhibit key enzymes involved in AD, including acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), ß-secretase (BACE-1), and different kinases, together with the related pathways involved in the pathogenesis of AD. The compounds discussed herein are emerging as promising anti-AD activities for further in-depth in vitro and in vivo investigations, to gain more insight of their mechanisms of action and for the development of potential anti-AD drug leads.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Organismos Acuáticos/química , Inhibidores de la Colinesterasa/farmacología , Fármacos Neuroprotectores/farmacología , Encéfalo/metabolismo , Humanos , Simulación del Acoplamiento Molecular
5.
J Neurochem ; 155(2): 120-136, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32277473

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder with no approved disease-modifying therapy (DMT). In this review, we summarize the various past approaches taken in an attempt to find treatments capable of altering the long-term course for individuals with AD, including: translating epidemiological observations into potential treatment options; seeking a single-treatment approach across the continuum of AD severity; utilizing biomarkers for assessing target engagement; using biomarkers as early surrogates of clinical efficacy; and enriching study populations to demonstrate adequate placebo decline during the limited duration of clinical trials. Although targeting the amyloid-ß (Aß) pathway has been central to the search for an effective DMT, to date, trials of anti-Aß monoclonal antibodies have failed to consistently demonstrate significant clinical efficacy. Key learnings from these anti-Aß trials, as well as the trials that came before them, have shifted the focus within clinical development programs to identifying target populations thought most likely to benefit from treatments (i.e., individuals at an earlier stage of disease). Other learnings include strategies to increase the likelihood of showing measurable improvements within the clinical trial setting by better predicting decline in placebo participants, as well as developing measures to quantify the needed treatment exposure (e.g., higher doses). Given the complexity associated with AD pathology and progression, treatments targeting non-amyloid AD pathologies in combination with anti-amyloid therapies may offer an alternative for the successful development of DMTs.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos Monoclonales/uso terapéutico , Humanos , Factores de Riesgo
6.
J Comput Aided Mol Des ; 34(9): 983-1002, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32488355

RESUMEN

The cholinesterases are essential targets implicated in the pathogenesis of Alzheimer's disease (AD). In the present study, virtual screening and molecular docking are performed to identify the potential hits. Docking-post processing (DPP) and pose filtration protocols against AChE and BChE resulted in three hits (AW00308, HTS04089, and JFD03947). Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) and molecular dynamics simulation analysis affirmed the stability and binding pattern of the docked complex JFD03947, which was further synthesized and evaluated for in vitro cholinesterase inhibition (AChE, IC50 = 0.062 µM; BChE, IC50 = 1.482 µM) activity. The enzyme kinetics study of the JFD03947 against hAChE and hBChE suggested a mixed type of inhibition. The results of thioflavin T-assay also elicited anti-Aß aggregation activity by JFD03947. Further, biological evaluation of identified compound JFD03947 also showed neuroprotective ability against the SH-SY5Y neuroblastoma cell lines.


Asunto(s)
Acetilcolinesterasa/química , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Butirilcolinesterasa/química , Inhibidores de la Colinesterasa/farmacología , Biología Computacional/métodos , Diseño de Fármacos , Dominio Catalítico , Inhibidores de la Colinesterasa/química , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Agregado de Proteínas
7.
Bioorg Med Chem ; 28(8): 115400, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32146060

RESUMEN

A series of phthalide alkyl tertiary amine derivatives were designed, synthesized and evaluated as potential multi-target agents against Alzheimer's disease (AD). The results indicated that almost all the compounds displayed significant AChE inhibitory and selective activities. Besides, most of the derivatives exhibited increased self-induced Aß1-42 aggregation inhibitory activity compared to the lead compound dl-NBP, and some compounds also exerted good antioxidant activity. Specifically, compound I-8 showed the highest inhibitory potency toward AChE (IC50 = 2.66 nM), which was significantly better than Donepezil (IC50 = 26.4 nM). Moreover, molecular docking studies revealed that compound I-8 could bind to both the catalytic active site and peripheral anionic site of AChE. Furthermore, compound I-8 displayed excellent BBB permeability in vitro. Importantly, the step-down passive avoidance test indicated that I-8 significantly reversed scopolamine-induced memory deficit in mice. Collectively, these results suggested that I-8 might be a potent and selective AChE inhibitor for further anti-AD drug development.


Asunto(s)
Aminas/química , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/farmacología , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Butirilcolinesterasa/metabolismo , Diseño de Fármacos , Femenino , Masculino , Membranas Artificiales , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Permeabilidad , Agregación Patológica de Proteínas , Distribución Aleatoria , Ratas
8.
Biologicals ; 65: 25-32, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32165080

RESUMEN

Immunoglobulin preparations are one of the promising drugs for Alzheimer's disease (AD). Anti-ß-amyloid (Aß) oligomers antibodies in immunoglobulin preparations are considered to be critical for the therapeutic effect against Alzheimer's disease. However, the antibodies content in immunoglobulin preparations varies greatly. In order to determine which factor contributes to the difference of the antibodies content, the content of anti-Aß oligomers antibodies in multiple batches of immunoglobulin preparations from two manufacturers were measured by enzyme-linked immunosorbent assay. The results showed that no significant difference was found in the antibodies content among different bathes of normal immunoglobulin preparations prepared by the same process from the same manufacturer, whereas significant difference was found in the antibodies content between normal immunoglobulin preparations prepared by ethanol fractionation and those by chromatography process from the same manufacturer. In addition, significant variation existed in the antibodies content between normal immunoglobulin preparations and specific immunoglobulin preparations that are produced by plasma pool of immunized donors. Based on analysis of these results, the preparation process and raw plasma could be the main contributing factors affecting the content of anti-Aß oligomers antibodies in immunoglobulin preparations. This finding might help to develop AD-specific immunoglobulin preparation containing higher content of anti-Aß oligomers antibodies.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Anticuerpos/análisis , Productos Biológicos/química , Inmunoglobulinas/química , Fragmentos de Péptidos/inmunología , Enfermedad de Alzheimer/tratamiento farmacológico , Anticuerpos/uso terapéutico , Productos Biológicos/uso terapéutico , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos
9.
J Comput Aided Mol Des ; 33(5): 521-530, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30989573

RESUMEN

Although the mechanism of Alzheimer's disease (AD) is still not fully understood, the development of multifunctional AChE inhibitors remains a research focus for AD treatment. In this study, 48 AChE candidate inhibitors were picked out from SPECS database through a pharmacophore- and molecular docking-based virtual screening. The biological evaluation results indicated that four compounds 7, 29, 41 and 48 with different scaffolds exhibited potent and selective AChE inhibitory activity, with the best IC50 value of 1.62 ± 0.11 µM obtained for 48. Then their mechanism of action, the inhibition on Aß aggregation, neurotoxicity, and neuroprotective activity against Aß-induced nerve cell injury were well studied. The binding mode of 48 with AChE was also proposed. The present bioassay results indicated that these multifunctional AChE inhibitors were worth for further structural derivatization to make them the anti-AD lead compounds.


Asunto(s)
Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/farmacología , Acetilcolinesterasa/química , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular , Diseño de Fármacos , Electrophorus , Humanos , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Agregado de Proteínas/efectos de los fármacos
10.
Molecules ; 24(14)2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31311169

RESUMEN

In this study, a series of selective butyrylcholinesterase (BChE) inhibitors was designed and synthesized from the structural optimization of hit 1, a 4-((3,4-dihydroisoquinolin-2(1H)-yl)methyl)benzoic acid derivative identified by virtual screening our compound library. The in vitro enzyme assay results showed that compounds 9 ((4-((3,4-dihydroisoquinolin-2(1H)-yl)methyl)phenyl)(pyrrolidin-1-yl)methanone) and 23 (N-(2-bromophenyl)-4-((3,4-dihydroisoquinolin-2(1H)-yl)methyl)benzamide) displayed improved BChE inhibitory activity and good selectivity towards BChE versus AChE. Their binding modes were probed by molecular docking and further validated by molecular dynamics simulation. Kinetic analysis together with molecular modeling studies suggested that these derivatives could target both the catalytic active site (CAS) and peripheral anionic site (PAS) of BChE. In addition, the selected compounds 9 and 23 displayed anti-Aß1-42 aggregation activity in a dose-dependent manner, and they did not show obvious cytotoxicity towards SH-SY5Y neuroblastoma cells. Also, both compounds showed significantly protective activity against Aß1-42-induced toxicity in a SH-SY5Y cell model. The present results provided a new valuable chemical template for the development of selective BChE inhibitors.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Benzoatos/síntesis química , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/síntesis química , Péptidos beta-Amiloides/metabolismo , Benzoatos/química , Benzoatos/farmacología , Butirilcolinesterasa/química , Dominio Catalítico/efectos de los fármacos , Línea Celular Tumoral , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Fenoles , Agregado de Proteínas/efectos de los fármacos , Relación Estructura-Actividad
11.
J Enzyme Inhib Med Chem ; 33(1): 1212-1224, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30160188

RESUMEN

A new series of multifunctional hybrids, based on the structure of the donepezil (DNP) drug, have been developed and evaluated as potential anti Alzheimer's disease (AD) agents. The rationale of this study was the conjugation of a benzylpiperidine/benzylpiperazine moiety with derivatives of bioactive heterocyclics (benzimidazole or benzofuran), to mimic the main structure of DNP and to endow the hybrids with additional relevant properties such as inhibition of amyloid beta (Aß) peptide aggregation, antioxidant activity and metal chelation. Overall, they showed good activity for AChE inhibition (IC50=4.0-30.0 µΜ) and moderate ability for inhibition of Aß1-42 self-mediated aggregation. The hybrids containing chelating groups showed improvement in the inhibition of Cu-induced Aß42 aggregation and the antioxidant capacity. Moreover, neuroprotective effects of these compounds were evidenced in neuroblastoma cells after Aß1-42 induced toxicity. Structure-activity relationship allowed the identification of some promising compounds and the main determinant structural features for the targeted properties.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Antioxidantes/farmacología , Inhibidores de la Colinesterasa/farmacología , Indanos/farmacología , Piperidinas/farmacología , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Antioxidantes/síntesis química , Antioxidantes/química , Células CACO-2 , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/química , Donepezilo , Relación Dosis-Respuesta a Droga , Humanos , Indanos/síntesis química , Indanos/química , Modelos Moleculares , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Agregado de Proteínas/efectos de los fármacos , Relación Estructura-Actividad
12.
Bioorg Med Chem ; 25(6): 1997-2009, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28237559

RESUMEN

A series of 1-hydroxyl-3-aminoalkoxy-thioxanthone derivatives were designed, synthesized and evaluated as potential multifunctional agents against Alzheimer's disease (AD). The results indicated that most of these compounds exhibited good AChE and MAOs inhibitory activities, significant inhibition of self- and Cu2+-induced Aß1-42 aggregation, and moderate to good antioxidant activities. Specifically, compound 9e displayed high inhibitory potency toward AChE (IC50=0.59±0.02µM), MAO-A and MAO-B (IC50=1.01±0.02µM and 0.90±0.01µM respectively), excellent efficiency to block both self- and Cu2+-induced Aß1-42 aggregation (74.8±1.2% and 87.7±1.9% at 25µM, respectively), good metal-chelating property and a low toxicity in SH-SY5Y cells. Furthermore, kinetic and molecular modeling studies revealed that compound 9e binds simultaneously to the catalytic active site and peripheral anionic site of AChE, and could penetrate the BBB. Collectively, these results suggested that 9e might be a potential multifunctional agent for further development in the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/química , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Xantonas/farmacología , Acetilcolinesterasa/efectos de los fármacos , Enfermedad de Alzheimer/enzimología , Línea Celular , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/uso terapéutico , Humanos , Cinética , Modelos Moleculares , Inhibidores de la Monoaminooxidasa/química , Inhibidores de la Monoaminooxidasa/uso terapéutico , Tioxantenos/química , Tioxantenos/farmacología , Tioxantenos/uso terapéutico , Xantonas/química , Xantonas/uso terapéutico
13.
Chemistry ; 22(16): 5458-66, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-26945679

RESUMEN

Sequence-specific peptidomimetics are molecules that mimic the structure and function of peptides and proteins. With new backbones and molecular frameworks, peptidomimetics are of considerable interest in addressing challenges encountered in chemical biology and biomedical sciences. Based on the γ-PNA backbone, a new class of peptidomimetics - "γ-AApeptides" were recently developed. Both linear and cyclic γ-AApeptides can be synthesized with high efficiency. Compared with α-peptides, γ-AApeptides are resistant to enzymatic degradation, and amendable to diversification with a variety of chemical groups. Moreover, they could mimic primary and secondary structure, as well as the function of peptides, and show promise in biological applications, such as the development of new agents combating bacteria, cancer, and Alzheimer's disease. A few research outcomes of γ-AApeptides are highlighted in this Concept article, and a future perspective is also proposed.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Péptidos/química , Peptidomiméticos/química , Peptidomiméticos/síntesis química , Fenómenos Bioquímicos , Estructura Molecular
14.
Biomed Pharmacother ; 173: 116404, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38471275

RESUMEN

High-fat diet (HFD)-induced fatty liver disease is a deteriorating risk factor for Alzheimer's disease (AD). Mitigating fatty liver disease has been shown to attenuate AD-like pathology in animal models. However, it remains unclear whether enhancing Aß clearance through immunotherapy would in turn attenuate HFD-induced fatty liver or whether its efficacy would be compromised by long-term exposure to HFD. Here, the therapeutic potentials of an anti-Aß antibody, NP106, was investigated in APP/PS1 mice by HFD feeding for 44 weeks. The data demonstrate that NP106 treatment effectively reduced Aß burden and pro-inflammatory cytokines in HFD-fed APP/PS1 mice and ameliorated HFD-aggravated cognitive impairments during the final 18 weeks of the study. The rejuvenating characteristics of microglia were evident in APP/PS1 mice with NP106 treatment, namely enhanced microglial Aß phagocytosis and attenuated microglial lipid accumulation, which may explain the benefits of NP106. Surprisingly, NP106 also reduced HFD-induced hyperglycemia, fatty liver, liver fibrosis, and hepatic lipids, concomitant with modifications in the expressions of genes involved in hepatic lipogenesis and fatty acid oxidation. The data further reveal that brain Aß burden and behavioral deficits were positively correlated with the severity of fatty liver disease and fasting serum glucose levels. In conclusion, our study shows for the first time that anti-Aß immunotherapy using NP106, which alleviates AD-like disorders in APP/PS1 mice, ameliorates fatty liver disease. Minimizing AD-related pathology and symptoms may reduce the vicious interplay between central AD and peripheral fatty liver disease, thereby highlighting the importance of developing AD therapies from a systemic disease perspective.


Asunto(s)
Enfermedad de Alzheimer , Hígado Graso , Hepatopatías , Ratones , Animales , Precursor de Proteína beta-Amiloide/metabolismo , Ratones Transgénicos , Dieta Alta en Grasa/efectos adversos , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Hepatopatías/metabolismo , Hígado Graso/metabolismo , Modelos Animales de Enfermedad , Péptidos beta-Amiloides/metabolismo
15.
Methods Mol Biol ; 2761: 337-354, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38427249

RESUMEN

The gradual loss of neurons' structure and function in the central nervous system is known as neurodegeneration. It is a defining feature of several incapacitating illnesses, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. The buildup of amyloid beta (Aß) protein in the brain is one of the several variables linked to neurodegeneration. We shall delve into the fascinating realm of Aß in this chapter and examine its role in the etiology of neurodegenerative illnesses. Insights into the processes through which Aß exerts its toxicity are crucial for the creation of therapeutic approaches to treat these life-threatening diseases. Despite the presence of multiple obstacles, recent research shows promise for the development of some new anti-Aß therapies that will help millions of people suffering from neurodegeneration. In this chapter, we discuss the role of Aß in contributing to neurotoxicity and several anti-Aß therapies for neuroprotection.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Parkinson , Humanos , Péptidos beta-Amiloides/metabolismo , Neuroprotección/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Parkinson/terapia
16.
Biomed Pharmacother ; 175: 116784, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38781865

RESUMEN

1,8-Cineole is a bicyclic monoterpene widely distributed in the essential oils of various medicinal plants, and it exhibits significant anti-inflammatory and antioxidant activities. We aimed to investigate the therapeutic effect of 1,8-cineole on anti-Alzheimer's disease by using transgenic Caenorhabditis elegans models. Our studies demonstrated that 1,8-cineole significantly relieved Aß1-42-induced paralysis and exhibited remarkable antioxidant and anti-Aß1-42 aggregation activities in transgenic nematodes CL4176, CL2006 and CL2355. We developed a 1,8-cineole/cyclodextrin inclusion complex, displaying enhanced anti-paralysis, anti-Aß aggregation and antioxidant activities compared to 1,8-cineole. In addition, we found 1,8-cineole treatment activated the SKN-1/Nrf-2 pathway and induced autophagy in nematodes. Our results demonstrated the antioxidant and anti-Alzheimer's disease activities of 1,8-cineole, which provide a potential therapeutic approach for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Animales Modificados Genéticamente , Antioxidantes , Caenorhabditis elegans , Eucaliptol , Eucaliptol/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Caenorhabditis elegans/efectos de los fármacos , Antioxidantes/farmacología , Péptidos beta-Amiloides/metabolismo , Ciclodextrinas/farmacología , Ciclodextrinas/química , Fragmentos de Péptidos/farmacología , Autofagia/efectos de los fármacos , Modelos Animales de Enfermedad
17.
J Control Release ; 355: 604-621, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36738970

RESUMEN

Since the complex interactions of multiple mechanisms involved in Alzheimer's disease (AD) preclude the monotherapeutic approaches from clinical application, combination therapy has become an attractive strategy for AD treatment. However, to be emphasized, the realization of the edges of combination therapy greatly depends on the reasonable choice of targets and the rational design of combination scheme. Acknowledgedly, amyloid plaques and hyperphosphorylated tau (p-tau) are two main hallmarks in AD with close pathological correlations, implying the hopeful prospect of combined intervention in them for AD treatment. Herein, we developed the nano-combination system, neuron-targeting PEG-PLA nanoparticles (CT-NP) loading two peptide drugs H102, a ß-sheet breaker acting on Aß, and NAP, a microtubule stabilizer acting on p-tau. Compared with free peptide combination, nano-combination system partly aligned the in vivo behaviors of combined peptides and enhanced peptide accumulation in lesion neurons by the guidance of targeting peptide CGN and Tet1, facilitating the therapeutic performance of peptide combination. Further, to maximize the therapeutic potential of nano-combination system, the combination ratio and mode were screened by the quantitative evaluation with combination index and U test, respectively, in vitro and in vivo. The results showed that the separated-loading CT-NP at the combination molar ratio of 2:1 (H102:NAP), CT-NP/H102 + CT-NP/NAP(2:1), generated the strongest synergistic therapeutic effects on Aß, p-tau and their linkage, and effectually prevented neuroinflammation, reversed the neuronal damage and restored cognitive performance in 3 × Tg-AD transgenic mice. Our studies provide critical data on the effectiveness of nano-combination therapy simultaneously intervening in Aß and p-tau, confirming the promising application of nano-combination strategy in AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas , Ratones , Animales , Enfermedad de Alzheimer/terapia , Encéfalo/metabolismo , Péptidos/uso terapéutico , Péptidos/farmacología , Ratones Transgénicos , Neuronas/metabolismo , Péptidos beta-Amiloides/metabolismo , Modelos Animales de Enfermedad
18.
J Biomol Struct Dyn ; 40(6): 2498-2515, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33111617

RESUMEN

To confront a disease like Alzheimer's disease having complex pathogenesis, development of multitarget-directed ligands has emerged as a promising drug discovery approach. In our endeavor towards the development of multitarget-directed ligands for Alzheimer's disease, a series of indoloquinoxaline derivatives were designed and synthesized. In vitro cholinesterase inhibition studies revealed that all the synthesized compounds exhibited moderate to good cholinesterase inhibitory activity. 6-(6-(Piperidin-1-yl)hexyl)-6H-indolo[2,3-b]quinoxaline 9f was identified as the most potent and selective BuChE inhibitor (IC50 = 0.96 µM, selectivity index = 0.17) that possessed 2 fold higher BuChE inhibitory activity compared to the commercially approved reference drug donepezil (IC50 = 1.87 µM). Moreover, compound 9f is also endowed with self-induced Aß1-42 aggregation inhibitory activity (51.24% inhibition at 50 µM concentration). Some of the compounds of the series also displayed moderate anti-oxidant activity. To perceive a putative binding mode of the compound 9f, molecular docking studies were carried out, and the results pointed out significant interactions of compound 9f with the enzymes in the binding sites of cholinesterases as well as Aß1-42. Additionally, compound 9f exhibited favorable in silico ADMET properties. Put together these findings project compound 9f as a potential multitarget-directed ligand in the direction of developing novel anti-AD drugs.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Enfermedad de Alzheimer , Inhibidores de la Colinesterasa , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides , Inhibidores de la Colinesterasa/química , Colinesterasas/metabolismo , Diseño de Fármacos , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad
19.
J Alzheimers Dis ; 85(3): 1195-1204, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34924395

RESUMEN

BACKGROUND: Evidence summaries for efficacy and safety of frequently employed treatments of Alzheimer's disease (AD) are sparse. OBJECTIVE: We aimed to perform an updated umbrella review to identify an efficacious and safe treatment for AD patients. METHODS: We conducted a search for meta-analyses and systematic reviews on the Embase, PubMed, The Cochrane Library, and Web of Science to address this knowledge gap. We examined the cognitive functions, behavioral symptoms, global clinical assessment, and Activities of Daily Living as efficacy endpoints, and the incidence of adverse events as safety profiles. RESULTS: Sixteen eligible papers including 149 studies were included in the umbrella review. The results showed that AChE inhibitors (donepezil, galantamine, rivastigmine, Huperzine A), Ginkgo biloba, and cerebrolysin appear to be beneficial for cognitive, global performances, and activities of daily living in patients with AD. Furthermore, anti-Aß agents are unlikely to have an important effect on slowing cognitive or functional impairment in mild to moderate AD. CONCLUSION: Our study demonstrated that AChE inhibitors, Ginkgo biloba, and cerebrolysin are the optimum cognitive and activities of daily living medication for patients with AD.


Asunto(s)
Actividades Cotidianas/psicología , Enfermedad de Alzheimer/tratamiento farmacológico , Inhibidores de la Colinesterasa/uso terapéutico , Nootrópicos/uso terapéutico , Seguridad del Paciente , Aminoácidos/uso terapéutico , Cognición/efectos de los fármacos , Donepezilo/uso terapéutico , Galantamina/uso terapéutico , Ginkgo biloba , Humanos , Extractos Vegetales/uso terapéutico , Rivastigmina/uso terapéutico
20.
Clin Interv Aging ; 17: 797-810, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35611326

RESUMEN

Aducanumab is a monoclonal antibody selective for amyloid ß (Aß) aggregates. In June 2021, aducanumab became the first drug underlying the pathophysiology of Alzheimer's disease (AD) approved by the US Food and Drug Administration (FDA), under the accelerated approval pathway. The decision was based on the ability of aducanumab to remove Aß plaques, without any evidence that the Aß clearance is correlated with less cognitive or functional decline. This decision has generated a considerable debate in the scientific community, especially because the results from the two Phase 3 trials, EMERGE and ENGAGE, were divergent and, even after the post hoc analysis, the data were insufficient to prove aducanumab efficacy. Moreover, some researchers think that this approval will be an obstacle to the progress and also demonstrated concerns about aducanumab cost and its safety profile. The European Medicines Agency's rejection of aducanumab in December 2021 just brought more controversy over FDA's decision. Now, Biogen is designing the FDA's required confirmatory study, named ENVISION, which should be complete in 2026. Despite the controversy, the aducanumab showed to affect downstream tau pathology, which could open doors for a combination therapy approach for AD (anti-tau and anti-amyloid drug). This review summarizes the clinical development of aducanumab until regulatory agencies' decisions, the available trials data and the controversy over aducanumab approval for AD.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos Monoclonales Humanizados , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales Humanizados/uso terapéutico , Humanos , Estados Unidos , United States Food and Drug Administration
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda