1
|
Ciaglia T, Miranda MR, Di Micco S, Vietri M, Smaldone G, Musella S, Di Sarno V, Auriemma G, Sardo C, Moltedo O, Pepe G, Bifulco G, Ostacolo C, Campiglia P, Manfra M, Vestuto V, Bertamino A. Neuroprotective Potential of Indole-Based Compounds: A Biochemical Study on Antioxidant Properties and Amyloid Disaggregation in Neuroblastoma Cells. Antioxidants (Basel) 2024; 13:1585. [PMID: 39765912 PMCID: PMC11673510 DOI: 10.3390/antiox13121585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Based on the established neuroprotective properties of indole-based compounds and their significant potential as multi-targeted therapeutic agents, a series of synthetic indole-phenolic compounds was evaluated as multifunctional neuroprotectors. Each compound demonstrated metal-chelating properties, particularly in sequestering copper ions, with quantitative analysis revealing approximately 40% chelating activity across all the compounds. In cellular models, these hybrid compounds exhibited strong antioxidant and cytoprotective effects, countering reactive oxygen species (ROS) generated by the Aβ(25-35) peptide and its oxidative byproduct, hydrogen peroxide, as demonstrated by quantitative analysis showing on average a 25% increase in cell viability and a reduction in ROS levels to basal states. Further analysis using thioflavin T fluorescence assays, circular dichroism, and computational studies indicated that the synthesized derivatives effectively promoted the self-disaggregation of the Aβ(25-35) fragment. Taken together, these findings suggest a unique profile of neuroprotective actions for indole-phenolic derivatives, combining chelating, antioxidant, and anti-aggregation properties, which position them as promising compounds for the development of multifunctional agents in Alzheimer's disease therapy. The methods used provide reliable in vitro data, although further in vivo validation and assessment of blood-brain barrier penetration are needed to confirm therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS), Via Salvatore de Renzi 50, 84125 Salerno, Italy;
| | - Mariapia Vietri
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carla Sardo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| |
Collapse
|
2
|
Carrazoni GS, Garces NB, Cadore CR, Sosa PM, Cattaneo R, Mello-Carpes PB. Supplementation with Manihot esculenta Crantz (Cassava) leaves' extract prevents recognition memory deficits and hippocampal antioxidant dysfunction induced by Amyloid-β. Nutr Neurosci 2024; 27:942-950. [PMID: 37948133 DOI: 10.1080/1028415x.2023.2280815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The Manihot esculenta Crantz (Cassava) is a typical South American plant rich in nutrients and energetic compounds. Lately, our group has shown that non-pharmacological interventions with natural antioxidants present different neuroprotective effects on oxidative balance and memory deficits in AD-like animal models. Here, our objective was to evaluate the neuroprotective effects of Cassava leaves' extract (CAS) in an AD-like model induced by amyloid-beta (Aβ) 25-35 peptide. METHODS Male Wistar rats (n = 40; 60 days old) were subjected to 10 days of CAS supplementation; then, we injected 2 μL Aβ 25-35 in the hippocampus by stereotaxic surgery. Ten days later, we evaluated object recognition (OR) memory. Cassavas' total polyphenols, flavonoids, and condensed tannins content were measured, as well as hippocampal lipid peroxidation and total antioxidant capacity. RESULTS CAS protected against Aβ-induced OR memory deficits. In addition, Aβ promoted antioxidant capacity decrease, while CAS was able to prevent it, in addition to diminishing lipoperoxidation compared to Aβ. DISCUSSION We show that treatment with Cassava leaves' extract before AD induction prevents recognition memory deficits related to Aβ hippocampal injection. At least part of these effects can be related to the Cassava leaves' extract supplementation effects on diminishing lipid peroxidation and preventing a decrease in the hippocampal total antioxidant capacity in the hippocampus of AD-like animals without adverse effects. Once cassavais a plant of warm and dry ground that can adapt to growon various soil types and seems to resist several insects, our results enable Cassava to be considered asa potential preventive intervention to avoid or minimizeAD-induced memory deficits worldwide.
Collapse
Affiliation(s)
- Guilherme Salgado Carrazoni
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | | | - Caroline Ramires Cadore
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | - Priscila Marques Sosa
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | | | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| |
Collapse
|
3
|
Mapping secretome-mediated interaction between paired neuron–macrophage single cells. Proc Natl Acad Sci U S A 2022; 119:e2200944119. [PMID: 36288285 PMCID: PMC9636946 DOI: 10.1073/pnas.2200944119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neuron-immune interaction through secreted factors contributes significantly to the complex microenvironment in the central nervous system that could alter cell functionalities and fates in both physiological and pathological conditions, which remains poorly characterized at the single-cell level. Herein, using a spatially patterned antibody barcode microchip, we realized the mapping of 12 different secretomes, covering cytokines, neurotrophic factors (NFs), and neuron-derived exosomes (NDEs) from high-throughput, paired single cells (≥ 600) simultaneously under normal conditions and an Alzheimer’s disease (AD) model induced with amyloid beta protein 1-42 (Aβ
1–42
). We applied the platform to analyze the secretion profiles from paired neuron–macrophage and neuron–microglia single cells with human cell lines. We found that pairwise neuron–macrophage interaction would trigger immune responses and attenuate neuron cells’ secretion, while neuron–microglia interaction generally results in opposite outcomes in secretion. When neuron cells are induced with Aβ
1–42
protein into the AD model, both neuron–macrophage and neuron–microglia interactions lead to increased cytokines and NDEs and decreased NFs. Further analysis of AD patients’ serum showed that NDEs were significantly higher in patients’ samples than in the control group, validating our observation from the interaction assay. Furthermore, we resolved previously undifferentiated heterogeneity underlying the secretions from single-neuron cells. We found that the NDE and NF secretion was less dependent on the paracrine signaling between one another and that secretions from neuron cells would attenuate after differentiation with Aβ
1–42
. This study demonstrates the mapping of the different secretomes from paired neuron-immune single cells, providing avenues for understanding how neurons and immune cells interact through the complex secretome network.
Collapse
|
4
|
Rudajev V, Novotny J. Cholesterol as a key player in amyloid β-mediated toxicity in Alzheimer’s disease. Front Mol Neurosci 2022; 15:937056. [PMID: 36090253 PMCID: PMC9453481 DOI: 10.3389/fnmol.2022.937056] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that is one of the most devastating and widespread diseases worldwide, mainly affecting the aging population. One of the key factors contributing to AD-related neurotoxicity is the production and aggregation of amyloid β (Aβ). Many studies have shown the ability of Aβ to bind to the cell membrane and disrupt its structure, leading to cell death. Because amyloid damage affects different parts of the brain differently, it seems likely that not only Aβ but also the nature of the membrane interface with which the amyloid interacts, helps determine the final neurotoxic effect. Because cholesterol is the dominant component of the plasma membrane, it plays an important role in Aβ-induced toxicity. Elevated cholesterol levels and their regulation by statins have been shown to be important factors influencing the progression of neurodegeneration. However, data from many studies have shown that cholesterol has both neuroprotective and aggravating effects in relation to the development of AD. In this review, we attempt to summarize recent findings on the role of cholesterol in Aβ toxicity mediated by membrane binding in the pathogenesis of AD and to consider it in the broader context of the lipid composition of cell membranes.
Collapse
|
5
|
Petermann AB, Reyna-Jeldes M, Ortega L, Coddou C, Yévenes GE. Roles of the Unsaturated Fatty Acid Docosahexaenoic Acid in the Central Nervous System: Molecular and Cellular Insights. Int J Mol Sci 2022; 23:5390. [PMID: 35628201 PMCID: PMC9141004 DOI: 10.3390/ijms23105390] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
Fatty acids (FAs) are essential components of the central nervous system (CNS), where they exert multiple roles in health and disease. Among the FAs, docosahexaenoic acid (DHA) has been widely recognized as a key molecule for neuronal function and cell signaling. Despite its relevance, the molecular pathways underlying the beneficial effects of DHA on the cells of the CNS are still unclear. Here, we summarize and discuss the molecular mechanisms underlying the actions of DHA in neural cells with a special focus on processes of survival, morphological development, and synaptic maturation. In addition, we examine the evidence supporting a potential therapeutic role of DHA against CNS tumor diseases and tumorigenesis. The current results suggest that DHA exerts its actions on neural cells mainly through the modulation of signaling cascades involving the activation of diverse types of receptors. In addition, we found evidence connecting brain DHA and ω-3 PUFA levels with CNS diseases, such as depression, autism spectrum disorders, obesity, and neurodegenerative diseases. In the context of cancer, the existing data have shown that DHA exerts positive actions as a coadjuvant in antitumoral therapy. Although many questions in the field remain only partially resolved, we hope that future research may soon define specific pathways and receptor systems involved in the beneficial effects of DHA in cells of the CNS, opening new avenues for innovative therapeutic strategies for CNS diseases.
Collapse
Affiliation(s)
- Ana B. Petermann
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
| | - Mauricio Reyna-Jeldes
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Lorena Ortega
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Claudio Coddou
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Gonzalo E. Yévenes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
| |
Collapse
|
6
|
The Impact of Medium Chain and Polyunsaturated ω-3-Fatty Acids on Amyloid-β Deposition, Oxidative Stress and Metabolic Dysfunction Associated with Alzheimer's Disease. Antioxidants (Basel) 2021; 10:antiox10121991. [PMID: 34943094 PMCID: PMC8698946 DOI: 10.3390/antiox10121991] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 01/22/2023] Open
Abstract
Alzheimer’s disease (AD), the most common cause of dementia in the elderly population, is closely linked to a dysregulated cerebral lipid homeostasis and particular changes in brain fatty acid (FA) composition. The abnormal extracellular accumulation and deposition of the peptide amyloid-β (Aβ) is considered as an early toxic event in AD pathogenesis, which initiates a series of events leading to neuronal dysfunction and death. These include the induction of neuroinflammation and oxidative stress, the disruption of calcium homeostasis and membrane integrity, an impairment of cerebral energy metabolism, as well as synaptic and mitochondrial dysfunction. Dietary medium chain fatty acids (MCFAs) and polyunsaturated ω-3-fatty acids (ω-3-PUFAs) seem to be valuable for disease modification. Both classes of FAs have neuronal health-promoting and cognition-enhancing properties and might be of benefit for patients suffering from mild cognitive impairment (MCI) and AD. This review summarizes the current knowledge about the molecular mechanisms by which MCFAs and ω-3-PUFAs reduce the cerebral Aβ deposition, improve brain energy metabolism, and lessen oxidative stress levels.
Collapse
|
7
|
Matos J, Afonso C, Cardoso C, Serralheiro ML, Bandarra NM. Yogurt Enriched with Isochrysis galbana: An Innovative Functional Food. Foods 2021; 10:1458. [PMID: 34202539 PMCID: PMC8306745 DOI: 10.3390/foods10071458] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 01/19/2023] Open
Abstract
Microalgae are a valuable and innovative emerging source of natural nutrients and bioactive compounds that can be used as functional ingredients in order to increase the nutritional value of foods to improve human health and to prevent disease. The marine microalga Isochrysis galbana has great potential for the food industry as a functional ingredient, given its richness in ω3 long chain-polyunsaturated fatty acids (LC-PUFAs), with high contents of oleic, linoleic, alpha-linolenic acid (ALA), stearidonic, and docosahexaenoic (DHA) acids. This study focuses on the formulation of a functional food by the incorporation of 2% (w/w) of I. galbana freeze-dried biomass and 2% (w/w) of I. galbana ethyl acetate lipidic extract in solid natural yogurts preparation. In the functional yogurt enriched with microalgal biomass, the ω3 LC-PUFA's content increased (to 60 mg/100 g w/w), specifically the DHA content (9.6 mg/100 g ww), and the ω3/ω6 ratio (augmented to 0.8). The in vitro digestion study showed a poor bioaccessibility of essential ω3 LC-PUFAs, wherein linoleic acid (18:2 ω6) presented a bioaccessibility inferior to 10% and no DHA or eicosapentaenoic acid (EPA) was detected in the bioaccessible fraction of the functional yogurts, thus indicating a low accessibility of lipids during digestion. Notwithstanding, when compared to the original yogurt, an added value novel functional yogurt with DHA and a higher ω3 LC-PUFAs content was obtained. The functional yogurt enriched with I. galbana can be considered important from a nutritional point of view and a suitable source of essential FAs in the human diet. However, this needs further confirmation, entailing additional investigation into bioavailability through in vivo assays.
Collapse
Affiliation(s)
- Joana Matos
- Division of Aquaculture, Upgrading and Bioprospection (DivAV), Portuguese Institute for the Sea and Atmosphere (IPMA, I.P.), Avenida Alfredo Magalhães Ramalho, 6, 1495-165 Algés, Portugal; (C.A.); (C.C.); (N.M.B.)
- Faculty of Sciences, BioISI—Biosystems & Integrative Sciences Institute, University of Lisboa, Campo Grande 016, 1749-016 Lisboa, Portugal;
| | - Cláudia Afonso
- Division of Aquaculture, Upgrading and Bioprospection (DivAV), Portuguese Institute for the Sea and Atmosphere (IPMA, I.P.), Avenida Alfredo Magalhães Ramalho, 6, 1495-165 Algés, Portugal; (C.A.); (C.C.); (N.M.B.)
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua dos Bragas 289, 4050-123 Porto, Portugal
| | - Carlos Cardoso
- Division of Aquaculture, Upgrading and Bioprospection (DivAV), Portuguese Institute for the Sea and Atmosphere (IPMA, I.P.), Avenida Alfredo Magalhães Ramalho, 6, 1495-165 Algés, Portugal; (C.A.); (C.C.); (N.M.B.)
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua dos Bragas 289, 4050-123 Porto, Portugal
| | - Maria L. Serralheiro
- Faculty of Sciences, BioISI—Biosystems & Integrative Sciences Institute, University of Lisboa, Campo Grande 016, 1749-016 Lisboa, Portugal;
| | - Narcisa M. Bandarra
- Division of Aquaculture, Upgrading and Bioprospection (DivAV), Portuguese Institute for the Sea and Atmosphere (IPMA, I.P.), Avenida Alfredo Magalhães Ramalho, 6, 1495-165 Algés, Portugal; (C.A.); (C.C.); (N.M.B.)
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Rua dos Bragas 289, 4050-123 Porto, Portugal
| |
Collapse
|
8
|
Changhong K, Peng Y, Yuan Z, Cai J. Ginsenoside Rb1 protected PC12 cells from Aβ 25-35-induced cytotoxicity via PPARγ activation and cholesterol reduction. Eur J Pharmacol 2020; 893:173835. [PMID: 33359145 DOI: 10.1016/j.ejphar.2020.173835] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/09/2020] [Accepted: 12/18/2020] [Indexed: 12/27/2022]
Abstract
Accumulating evidences suggest that amyloid β (Aβ)-peptide plays a key role in pathogenesis of Alzheimer's disease (AD) through aggregation and deposition into plaques in neuronal cells. Membrane components such as cholesterol and gangliosides not only enhance the production of amyloidogenic Aβ fragments, but also appear to strengthen Aβ-membrane interaction. Ginsenoside Rb1 (GRb1) is a major active component of Panax, which is widely used to improve learning and memory. In the present study, whether ginsenoside Rb1 could protect pheochromocytoma cells (PC12 cells) from Aβ25-35-induced cytotoxicity including inhibiting cell growth, inducing apoptosis, producing reactive oxygen species (ROS), destroying the cytoskeleton and bringing about membrane toxicity was investigated. Our results indicated that ginsenoside Rb1 could serve as an agonist of peroxisom proliferator-activated receptor-γ (PPARγ) and reduce the level of cholesterol in AD model cells. Reduction of the Aβ25-35-induced cytotoxicity by lowering cholesterol was evidenced by reduction of ROS production, lipid peroxidation, and protection of cytoskeleton and membrane surface rigidity. Most importantly, the viability of PC12 cells increased from 50.42 ± 5.51% for the AD group to 102.72 ± 4.34% for the 50 μM ginsenoside Rb1 group with cholesterol reduction. Our results suggested that ginsenoside Rb1 might function as an effective candidate to promote reverse cholesterol transport and lower ROS production, therefore providing a new insight into prevention and treatment of AD.
Collapse
Affiliation(s)
- Ke Changhong
- Department of Chemistry, Jinan University, Guangzhou, 510632, China; YZ Health-tech Inc., Hengqin District, Zhuhai, 519000, China
| | - Yuan Peng
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Zhengqiang Yuan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 51006, China.
| | - Jiye Cai
- Department of Chemistry, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
9
|
Rossi Dare L, Garcia A, Alves N, Ventura Dias D, de Souza MA, Mello-Carpes PB. Physical and cognitive training are able to prevent recognition memory deficits related to amyloid beta neurotoxicity. Behav Brain Res 2019; 365:190-197. [DOI: 10.1016/j.bbr.2019.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/19/2019] [Accepted: 03/02/2019] [Indexed: 12/13/2022]
|
10
|
Ji S, Li S, Zhao X, Kang N, Cao K, Zhu Y, Peng P, Fan J, Xu Q, Yang S, Liu Y. Protective role of phenylethanoid glycosides, Torenoside B and Savatiside A, in Alzheimer's disease. Exp Ther Med 2019; 17:3755-3767. [PMID: 30988761 DOI: 10.3892/etm.2019.7355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 02/26/2019] [Indexed: 11/06/2022] Open
Abstract
The current study assessed the efficacy of two phenylethanoid glycosides (PhGs), Torenoside B (TB) and Savatiside A (SA), in the treatment of Alzheimer's disease (AD). The effects of TB and SA compounds were first assessed following amyloid beta (Aβ)25-35 induction in SH-SY5Y cells at a range of concentrations. Their effects on cell viability and reactive oxygen species (ROS) were determined by performing MTT and dichlorofluorescin diacetate assays, respectively. The concentration of intracellular Ca2+ was determined using Fluo-3AM to stain SH-SY5Y cells. SA and TB treatments were also assessed in Aβ25-35-induced mice. Y-maze and Morris water maze methods were utilized to assess murine learning and memory capability. The pathological changes of murine hippocampi was determined using H&E and Nissl staining. In addition, biochemical parameters associated with intracellular reactive oxygen pathways including Maleic dialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), acetylcholinesterase (AChE) and Calnexin were also assessed. TB and SA treatment in Aβ25-35-induced SH-SY5Y cells resulted in the restoration of cell morphology, an increase of SOD and GSH-Px activity, a decrease in ROS, Ca2+ and MDA content, and a decrease in Calnexin expression. Furthermore, SA or TB treatment administered to Aβ25-35-induced mice improved their spatial/non-spatial learning and memory capabilities. The efficacy of treatment was also supported by a marked change in the morphological structure of pyramidal neurons in the CA1 areas of murine hippocampi, as well as an increase of SOD and GSH-Px activity. Treatment also resulted in a decrease in MDA content, AchE activity and Calnexin expression in murine hippocampal tissue. As potential AD treatment drugs, SA and TB compounds have been demonstrated to alleviate the oxidative stress induced by Aβ25-35 via the regulation of intracellular calcium homeostasis and Calnexin, preventing AD development.
Collapse
Affiliation(s)
- Shiliang Ji
- Department of Pharmacy, Suzhou Science and Technology Town Hospital, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Shanshan Li
- Patent Examination Cooperation (Jiangsu) Center of the Patent Office, SIPO, Suzhou, Jiangsu 215000, P.R. China
| | - Xingxing Zhao
- Department of Neonatology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215000, P.R. China
| | - Naixin Kang
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Keke Cao
- Shenzhen Yuanxing Gene-Tech Co., Ltd., Shenzhen, Guangdong 518000, P.R. China
| | - Yingying Zhu
- Suzhou Yihua Biomedical Technology Co., Ltd., Suzhou, Jiangsu 215000, P.R. China
| | - Panpan Peng
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Jing Fan
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Qiongming Xu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Shilin Yang
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Yanli Liu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
11
|
Sereia AL, de Oliveira MT, Baranoski A, Marques LLM, Ribeiro FM, Isolani RG, de Medeiros DC, Chierrito D, Lazarin-Bidóia D, Zielinski AAF, Novello CR, Nakamura CV, Mantovani MS, de Mello JCP. In vitro evaluation of the protective effects of plant extracts against amyloid-beta peptide-induced toxicity in human neuroblastoma SH-SY5Y cells. PLoS One 2019; 14:e0212089. [PMID: 30763379 PMCID: PMC6375598 DOI: 10.1371/journal.pone.0212089] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and has no cure. Therapeutic strategies focusing on the reduction of oxidative stress, modulation of amyloid-beta (Aβ) toxicity and inhibition of tau protein hyperphosphorylation are warranted to avoid the development and progression of AD. The aim of this study was to screen the crude extracts (CEs) and ethyl-acetate fractions (EAFs) of Guazuma ulmifolia, Limonium brasiliense, Paullinia cupana, Poincianella pluviosa, Stryphnodendron adstringens and Trichilia catigua using preliminary in vitro bioassays (acetylcholinesterase inhibition, antioxidant activity and total polyphenol content) to select extracts/fractions and assess their protective effects against Aβ25-35 toxicity in SH-SY5Y cells. The effect of the EAF of S. adstringens on mitochondrial membrane potential, lipid peroxidation, superoxide production and mRNA expression of 10 genes related to AD was also evaluated and the electropherogram fingerprints of EAFs were established by capillary electrophoresis. Chemometric tools were used to correlate the in vitro activities of the samples with their potential to be evaluated against AD and to divide extracts/fractions into four clusters. Pretreatment with the EAFs grouped in cluster 1 (S. adstringens, P. pluviosa and L. brasiliense) protected SH-SY5Y cells from Aβ25-35-induced toxicity. The EAF of S. adstringens at 15.62 μg/mL was able completely to inhibit the mitochondrial depolarization (69%), superoxide production (49%) and Aβ25-35-induced lipid peroxidation (35%). With respect to mRNA expression, the EAF of S. adstringens also prevented the MAPT mRNA overexpression (expression ratio of 2.387x) induced by Aβ25-35, which may be related to tau protein hyperphosphorylation. This is the first time that the neuroprotective effects of these fractions have been demonstrated and that the electropherogram fingerprints for the EAFs of G. ulmifolia, L. brasiliense, P. cupana, P. pluviosa and S. adstringens have been established. The study expands knowledge of the in vitro protective effects and quality control of the evaluated fractions.
Collapse
Affiliation(s)
- Ana Luiza Sereia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Marcelo Tempesta de Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Adrivanio Baranoski
- Programa de Pós-Graduação em Genética e Biologia Molecular, Department of General Biology, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Leila Larisa Medeiros Marques
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Fabianne Martins Ribeiro
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Raquel Garcia Isolani
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Daniela Cristina de Medeiros
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Danielly Chierrito
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Danielle Lazarin-Bidóia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Acácio Antonio Ferreira Zielinski
- Department of Chemical Engineering and Food Engineering, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cláudio Roberto Novello
- Academic Department of Chemistry and Biology, Universidade Tecnológica Federal do Paraná, Francisco Beltrão, Paraná, Brazil
| | - Celso Vataru Nakamura
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Mário Sérgio Mantovani
- Programa de Pós-Graduação em Genética e Biologia Molecular, Department of General Biology, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - João Carlos Palazzo de Mello
- Programa de Pós-Graduação em Ciências Farmacêuticas, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
- * E-mail:
| |
Collapse
|
12
|
Characterization of phospholipids from Pacific saury ( Cololabis saira ) viscera and their neuroprotective activity. FOOD BIOSCI 2018. [DOI: 10.1016/j.fbio.2018.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
13
|
Zhang YP, Miao R, Li Q, Wu T, Ma F. Effects of DHA Supplementation on Hippocampal Volume and Cognitive Function in Older Adults with Mild Cognitive Impairment: A 12-Month Randomized, Double-Blind, Placebo-Controlled Trial. J Alzheimers Dis 2018; 55:497-507. [PMID: 27716665 DOI: 10.3233/jad-160439] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Docosahexaenoic acid (DHA) is important for brain function, and higher DHA intake is inversely correlated with relative risk of Alzheimer's disease. The potential benefits of DHA supplementation in people with mild cognitive impairment (MCI) have not been fully examined. Our study aimed to determine the effect of DHA supplementation on cognitive function and hippocampal atrophy in elderly subjects with MCI. This was a randomized, double-blind, placebo-controlled trial in Tianjin, China. 240 individuals with MCI aged 65 years and over were recruited and equalized randomly allocated to the DHA or the placebo group. Participants received 12-month DHA supplementation (2 g/day) or corn oil as placebo. Both global and specific subdomains of cognitive function and hippocampal volume were measured at baseline, 6 months, and 12 months. Both changes were analyzed by repeated-measure analysis of variance (ANOVA). This trial has been registered: ChiCTR-IOR-15006058. A total of 219 participants (DHA: 110, Placebo: 109) completed the trial. The change in mean serum DHA levels was greater in the intervention group (+3.85%) compared to the control group (+1.06%). Repeated-measures analyses of covariance showed that, over 12 months, there was a significant difference in the Full-Scale Intelligence Quotient (ηp2 = 0.084; p = 0.039), Information (ηp2 = 0.439; p = 0.000), and Digit Span (ηp2 = 0.375; p = 0.000) between DHA-treated versus the placebo group. In addition, there were significant differences in volumes of left hippocampus (ηp2 = 0.121, p = 0.016), right hippocampus (ηp2 = 0.757, p = 0.008), total hippocampus (ηp2 = 0.124, p = 0.023), and global cerebrum (ηp2 = 0.145, p = 0.032) between the two groups. These findings suggest that DHA supplementation (2 g/day) for 12 months in MCI subjects can significantly improve cognitive function and slow the progression of hippocampal atrophy. Larger, longer-term confirmatory studies are warranted.
Collapse
Affiliation(s)
- Yan-Ping Zhang
- Department of Infectious Disease, General Hospital, Tianjin Medical University, Tianjin, China
| | - Rujuan Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qing Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Tianfeng Wu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Fei Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Zárate R, el Jaber-Vazdekis N, Tejera N, Pérez JA, Rodríguez C. Significance of long chain polyunsaturated fatty acids in human health. Clin Transl Med 2017; 6:25. [PMID: 28752333 PMCID: PMC5532176 DOI: 10.1186/s40169-017-0153-6] [Citation(s) in RCA: 314] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
In the last decades, the development of new technologies applied to lipidomics has revitalized the analysis of lipid profile alterations and the understanding of the underlying molecular mechanisms of lipid metabolism, together with their involvement in the occurrence of human disease. Of particular interest is the study of omega-3 and omega-6 long chain polyunsaturated fatty acids (LC-PUFAs), notably EPA (eicosapentaenoic acid, 20:5n-3), DHA (docosahexaenoic acid, 22:6n-3), and ARA (arachidonic acid, 20:4n-6), and their transformation into bioactive lipid mediators. In this sense, new families of PUFA-derived lipid mediators, including resolvins derived from EPA and DHA, and protectins and maresins derived from DHA, are being increasingly investigated because of their active role in the "return to homeostasis" process and resolution of inflammation. Recent findings reviewed in the present study highlight that the omega-6 fatty acid ARA appears increased, and omega-3 EPA and DHA decreased in most cancer tissues compared to normal ones, and that increments in omega-3 LC-PUFAs consumption and an omega-6/omega-3 ratio of 2-4:1, are associated with a reduced risk of breast, prostate, colon and renal cancers. Along with their lipid-lowering properties, omega-3 LC-PUFAs also exert cardioprotective functions, such as reducing platelet aggregation and inflammation, and controlling the presence of DHA in our body, especially in our liver and brain, which is crucial for optimal brain functionality. Considering that DHA is the principal omega-3 FA in cortical gray matter, the importance of DHA intake and its derived lipid mediators have been recently reported in patients with major depressive and bipolar disorders, Alzheimer disease, Parkinson's disease, and amyotrophic lateral sclerosis. The present study reviews the relationships between major diseases occurring today in the Western world and LC-PUFAs. More specifically this review focuses on the dietary omega-3 LC-PUFAs and the omega-6/omega-3 balance, in a wide range of inflammation disorders, including autoimmune diseases. This review suggests that the current recommendations of consumption and/or supplementation of omega-3 FAs are specific to particular groups of age and physiological status, and still need more fine tuning for overall human health and well being.
Collapse
Affiliation(s)
- Rafael Zárate
- Canary Islands Cancer Research Institute (ICIC), Ave. La Trinidad 61, Torre A. Arévalo, 7th floor, 38204 La Laguna, Tenerife Spain
| | - Nabil el Jaber-Vazdekis
- Centre Algatech, Institute of Microbiology, Academy of Sciences of the Czech Republic, Třeboň, Czech Republic
| | - Noemi Tejera
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, NR4 7UQ UK
| | - José A. Pérez
- Department of Animal Biology, Soil Science and Geology (Animal Physiology Unit), Faculty of Sciences, Universidad de La Laguna, Ave. Astrofísico Francisco Sánchez s/n, 38206 La Laguna, Tenerife Spain
| | - Covadonga Rodríguez
- Department of Animal Biology, Soil Science and Geology (Animal Physiology Unit), Faculty of Sciences, Universidad de La Laguna, Ave. Astrofísico Francisco Sánchez s/n, 38206 La Laguna, Tenerife Spain
- Institute of Biomedical Technologies (ITB), Universidad de La Laguna, Campus de Ofra, 38071 La Laguna, Tenerife Spain
| |
Collapse
|
15
|
Zhuo Y, Guo H, Cheng Y, Wang C, Wang C, Wu J, Zou Z, Gan D, Li Y, Xu J. Inhibition of phosphodiesterase-4 reverses the cognitive dysfunction and oxidative stress induced by Aβ25-35 in rats. Metab Brain Dis 2016; 31:779-91. [PMID: 26920899 DOI: 10.1007/s11011-016-9814-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/23/2016] [Indexed: 02/05/2023]
Abstract
Phosphodiesterase-4 (PDE4) inhibitors prevent the breakdown of the second messenger cAMP and have been demonstrated to improve learning in several animal models of cognition. In this study, we explored the antioxidative effects of rolipram in Alzheimer's disease (AD) by using bilateral Aβ25-35 injection into the hippocampus of rats, which were used as an AD model. Rats received 3 intraperitoneal (i.p.) doses of rolipram (0.1, 0.5 and 1.25 mg/kg) daily after the injection of Aβ25-35 for 25 days. Chronic administration of rolipram prevented the memory impairments induced by Aβ25-35, as assessed using the passive avoidance test and the Morris water maze test. Furthermore, rolipram significantly reduced the oxidative stress induced by Aβ25-35, as evidenced by the decrease in the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and restored the reduced GSH levels and superoxide dismutase (SOD) activity. Moreover, western blotting and real-time reverse transcription polymerase chain reaction (RT-PCR) analysis showed that rolipram remarkably upregulated thioredoxin (Trx) and inhibited the inducible nitric oxide synthase/nitric oxide (iNOS/NO) pathway in the hippocampus. These results demonstrated that rolipram improved the learning and memory abilities in an Aβ25-35-induced AD rat model. The mechanism underlying these effects may be due to the noticeable antioxidative effects of rolipram.
Collapse
Affiliation(s)
- Yeye Zhuo
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
- The first affiliated hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Haibiao Guo
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yufang Cheng
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuang Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
| | - Canmao Wang
- Department of Pharmacy, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, 518000, China
| | - Jingang Wu
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhengqiang Zou
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Danna Gan
- Department of Pharmacy, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, 518000, China
| | - Yiwen Li
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiangping Xu
- Department of Pharmacology, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
16
|
Ghobeh M, Ahmadian S, Meratan AA, Ebrahim-Habibi A, Ghasemi A, Shafizadeh M, Nemat-Gorgani M. Interaction of Aβ(25-35) fibrillation products with mitochondria: Effect of small-molecule natural products. Biopolymers 2016; 102:473-86. [PMID: 25297917 DOI: 10.1002/bip.22572] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/18/2014] [Accepted: 10/06/2014] [Indexed: 12/25/2022]
Abstract
The 25-35 fragment of the amyloid β (Aβ) peptide is a naturally occurring proteolytic by-product that retains the pathophysiology of its larger parent molecule, whose deposition has been shown to involve mitochondrial dysfunction. Hence, disruption of Aβ(25-35) aggregates could afford an effective remedial strategy for Alzheimer's disease (AD). In the present study, the effect of a number of selected small-molecule natural products (polyphenols: resveratrol, quercetin, biochanin A, and indoles: indole-3-acetic acid, indole-3-carbinol (I3C)) on Aβ(25-35) fibrillogenesis was explored under physiological conditions, and interaction of the resulting structures with rat brain mitochondria was investigated. Several techniques, including fluorescence, circular dichroism, and transmission electron microscopy were utilized to characterize the aggregation products, and possible mitochondrial membrane permeabilization was determined following release of marker enzymes. Results demonstrate the capacity of Aβ(25-35) fibrils to damage mitochondria and suggest how small molecules may afford protection. While I3C appeared more effective in inhibiting the fibrillation process, all natural products behaved similarly in destabilizing preformed aggregates. It is concluded that elucidation of such protection may provide important insights into the development of preventive and therapeutic agents for AD.
Collapse
Affiliation(s)
- Maryam Ghobeh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
17
|
Grimm MOW, Mett J, Stahlmann CP, Haupenthal VJ, Blümel T, Stötzel H, Grimm HS, Hartmann T. Eicosapentaenoic acid and docosahexaenoic acid increase the degradation of amyloid-β by affecting insulin-degrading enzyme. Biochem Cell Biol 2016; 94:534-542. [PMID: 27813426 DOI: 10.1139/bcb-2015-0149] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) have been proposed to be highly beneficial in Alzheimer's disease (AD). AD pathology is closely linked to an overproduction and accumulation of amyloid-β (Aβ) peptides as extracellular senile plaques in the brain. Total Aβ levels are not only dependent on its production by proteolytic processing of the amyloid precursor protein (APP), but also on Aβ-clearance mechanisms, including Aβ-degrading enzymes. Here we show that the omega-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) increase Aβ-degradation by affecting insulin-degrading enzyme (IDE), the major Aβ-degrading enzyme secreted into the extracellular space of neuronal and microglial cells. The identification of the molecular mechanisms revealed that EPA directly increases IDE enzyme activity and elevates gene expression of IDE. DHA also directly stimulates IDE enzyme activity and affects IDE sorting by increasing exosome release of IDE, resulting in enhanced Aβ-degradation in the extracellular milieu. Apart from the known positive effect of DHA in reducing Aβ production, EPA and DHA might ameliorate AD pathology by increasing Aβ turnover.
Collapse
Affiliation(s)
- Marcus O W Grimm
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany.,b Neurodegeneration and Neurobiology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany.,c Deutsches Institut für DemenzPrävention (DIDP), Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Janine Mett
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Christoph P Stahlmann
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Viola J Haupenthal
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Tamara Blümel
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Hannah Stötzel
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Heike S Grimm
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| | - Tobias Hartmann
- a Experimental Neurology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany.,b Neurodegeneration and Neurobiology, Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany.,c Deutsches Institut für DemenzPrävention (DIDP), Saarland University, Kirrberger Str. 1, 66421 Homburg/Saar, Germany
| |
Collapse
|
18
|
Belkouch M, Hachem M, Elgot A, Lo Van A, Picq M, Guichardant M, Lagarde M, Bernoud-Hubac N. The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer's disease. J Nutr Biochem 2016; 38:1-11. [PMID: 27825512 DOI: 10.1016/j.jnutbio.2016.03.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/14/2015] [Accepted: 03/03/2016] [Indexed: 12/17/2022]
Abstract
Among omega-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA, 22:6n-3) is important for adequate brain development and cognition. DHA is highly concentrated in the brain and plays an essential role in brain functioning. DHA, one of the major constituents in fish fats, readily crosses the blood-brain barrier from blood to the brain. Its critical role was further supported by its reduced levels in the brain of Alzheimer's disease (AD) patients. This agrees with a potential role of DHA in memory, learning and cognitive processes. Since there is yet no cure for dementia such as AD, there is growing interest in the role of DHA-supplemented diet in the prevention of AD pathogenesis. Accordingly, animal, epidemiological, preclinical and clinical studies indicated that DHA has neuroprotective effects in a number of neurodegenerative conditions including AD. The beneficial effects of this key omega-3 fatty acid supplementation may depend on the stage of disease progression, other dietary mediators and the apolipoprotein ApoE genotype. Herein, our review investigates, from animal and cell culture studies, the molecular mechanisms involved in the neuroprotective potential of DHA with emphasis on AD.
Collapse
Affiliation(s)
- Mounir Belkouch
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France.
| | - Mayssa Hachem
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Abdeljalil Elgot
- Laboratoire des Sciences et Technologies de la Santé, Unité des Sciences Biomédicales, Institut Supérieur des Sciences de la Santé, Université Hassan 1er, Settat, Morocco
| | - Amanda Lo Van
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Madeleine Picq
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Guichardant
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Lagarde
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Nathalie Bernoud-Hubac
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| |
Collapse
|
19
|
Heras-Sandoval D, Pedraza-Chaverri J, Pérez-Rojas JM. Role of docosahexaenoic acid in the modulation of glial cells in Alzheimer's disease. J Neuroinflammation 2016; 13:61. [PMID: 26965310 PMCID: PMC4787218 DOI: 10.1186/s12974-016-0525-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 03/03/2016] [Indexed: 01/25/2023] Open
Abstract
Docosahexaenoic acid (DHA) is an omega-3 (ω-3) long-chain polyunsaturated fatty acid (LCPUFA) relevant for brain function. It has largely been explored as a potential candidate to treat Alzheimer’s disease (AD). Clinical evidence favors a role for DHA in the improvement of cognition in very early stages of the AD. In response to stress or damage, DHA generates oxygenated derivatives called docosanoids that can activate the peroxisome proliferator-activated receptor γ (PPARγ). In conjunction with activated retinoid X receptors (RXR), PPARγ modulates inflammation, cell survival, and lipid metabolism. As an early event in AD, inflammation is associated with an excess of amyloid β peptide (Aβ) that contributes to neural insult. Glial cells are recognized to be actively involved during AD, and their dysfunction is associated with the early appearance of this pathology. These cells give support to neurons, remove amyloid β peptides from the brain, and modulate inflammation. Since DHA can modulate glial cell activity, the present work reviews the evidence about this modulation as well as the effect of docosanoids on neuroinflammation and in some AD models. The evidence supports PPARγ as a preferred target for gene modulation. The effective use of DHA and/or its derivatives in a subgroup of people at risk of developing AD is discussed.
Collapse
Affiliation(s)
- David Heras-Sandoval
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, México, DF, México.,Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Av. San Fernando #22, Tlalpan 14080, Apartado Postal 22026, México, DF, México
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, México, DF, México
| | - Jazmin M Pérez-Rojas
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Av. San Fernando #22, Tlalpan 14080, Apartado Postal 22026, México, DF, México.
| |
Collapse
|
20
|
Teng E, Taylor K, Bilousova T, Weiland D, Pham T, Zuo X, Yang F, Chen PP, Glabe CG, Takacs A, Hoffman DR, Frautschy SA, Cole GM. Dietary DHA supplementation in an APP/PS1 transgenic rat model of AD reduces behavioral and Aβ pathology and modulates Aβ oligomerization. Neurobiol Dis 2015; 82:552-560. [PMID: 26369878 DOI: 10.1016/j.nbd.2015.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/22/2015] [Accepted: 09/07/2015] [Indexed: 12/18/2022] Open
Abstract
Increased dietary consumption of docosahexaenoic acid (DHA) is associated with decreased risk for Alzheimer's disease (AD). These effects have been postulated to arise from DHA's pleiotropic effects on AD pathophysiology, including its effects on β-amyloid (Aβ) production, aggregation, and toxicity. While in vitro studies suggest that DHA may inhibit and reverse the formation of toxic Aβ oligomers, it remains uncertain whether these mechanisms operate in vivo at the physiological concentrations of DHA attainable through dietary supplementation. We sought to clarify the effects of dietary DHA supplementation on Aβ indices in a transgenic APP/PS1 rat model of AD. Animals maintained on a DHA-supplemented diet exhibited reductions in hippocampal Aβ plaque density and modest improvements on behavioral testing relative to those maintained on a DHA-depleted diet. However, DHA supplementation also increased overall soluble Aβ oligomer levels in the hippocampus. Further quantification of specific conformational populations of Aβ oligomers indicated that DHA supplementation increased fibrillar (i.e. putatively less toxic) Aβ oligomers and decreased prefibrillar (i.e. putatively more toxic) Aβ oligomers. These results provide in vivo evidence suggesting that DHA can modulate Aβ aggregation by stabilizing soluble fibrillar Aβ oligomers and thus reduce the formation of both Aβ plaques and prefibrillar Aβ oligomers. However, since fibrillar Aβ oligomers still retain inherent neurotoxicity, DHA may need to be combined with other interventions that can additionally reduce fibrillar Aβ oligomer levels for more effective prevention of AD in clinical settings.
Collapse
Affiliation(s)
- Edmond Teng
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| | - Karen Taylor
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tina Bilousova
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - David Weiland
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Thaidan Pham
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Xiaohong Zuo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Department of Neurobiology and Neurology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fusheng Yang
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Ping-Ping Chen
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Charles G Glabe
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA; Biochemistry Department and Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Dennis R Hoffman
- Retina Foundation of the Southwest, Dallas, TX, USA; Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sally A Frautschy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Gregory M Cole
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
21
|
Yu H, Yao L, Zhou H, Qu S, Zeng X, Zhou D, Zhou Y, Li X, Liu Z. Neuroprotection against Aβ25–35-induced apoptosis by Salvia miltiorrhiza extract in SH-SY5Y cells. Neurochem Int 2014; 75:89-95. [DOI: 10.1016/j.neuint.2014.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/09/2014] [Accepted: 06/04/2014] [Indexed: 12/30/2022]
|
22
|
Casañas-Sánchez V, Pérez JA, Fabelo N, Herrera-Herrera AV, Fernández C, Marín R, González-Montelongo MC, Díaz M. Addition of docosahexaenoic acid, but not arachidonic acid, activates glutathione and thioredoxin antioxidant systems in murine hippocampal HT22 cells: potential implications in neuroprotection. J Neurochem 2014; 131:470-83. [DOI: 10.1111/jnc.12833] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 07/12/2014] [Accepted: 07/21/2014] [Indexed: 01/20/2023]
Affiliation(s)
- Verónica Casañas-Sánchez
- Department of Genetics; University Institute of Tropical Diseases and Public Health; University of La Laguna; Tenerife Spain
| | - José A. Pérez
- Department of Genetics; University Institute of Tropical Diseases and Public Health; University of La Laguna; Tenerife Spain
| | - Noemí Fabelo
- Laboratory of Membrane Physiology and Biophysics; Department of Animal Biology; University of La Laguna; Tenerife Spain
| | | | - Cecilia Fernández
- Laboratory of Cellular Neurobiology; Department of Physiology; University of La Laguna; Tenerife Spain
| | - Raquel Marín
- Laboratory of Cellular Neurobiology; Department of Physiology; University of La Laguna; Tenerife Spain
| | - María C. González-Montelongo
- Laboratory of Membrane Physiology and Biophysics; Department of Animal Biology; University of La Laguna; Tenerife Spain
| | - Mario Díaz
- Laboratory of Membrane Physiology and Biophysics; Department of Animal Biology; University of La Laguna; Tenerife Spain
| |
Collapse
|
23
|
Chua A, Thomas P, Wijesundera C, Clifton P, Fenech M. Effect of docosahexaenoic acid and furan fatty acids on cytokinesis block micronucleus cytome assay biomarkers in astrocytoma cell lines under conditions of oxidative stress. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:573-590. [PMID: 24828973 DOI: 10.1002/em.21873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/24/2014] [Accepted: 04/25/2014] [Indexed: 06/03/2023]
Abstract
Fatty acids from fish such as docosahexaenoic acid (DHA) are associated with improved brain function, whereas furan fatty acids (FFAs) also found in fish oil at low levels (1%) are thought to have antioxidant properties. Understanding their effects in astrocytes is important as these cells are responsible for maintaining healthy neurons via lipid homeostasis and distribution within the brain, and their decline with aging is a possible cause of dementia. We investigated the cytotoxic and genotoxic effects of DHA and FFA using the cytokinesis-block micronucleus cytome assay in in vitro cultures of U87MG (APOE ɛ3/ɛ3) and U118MG (APOE ɛ2/ɛ4) astrocytoma cell lines with and without a hydrogen peroxide (H2O2, 100 µM) challenge. U118MG was found to be more sensitive to the cytostatic, cytotoxic (i.e., apoptosis), and DNA damaging effects [micronuclei (MNi), nucleoplasmic bridges (NPBs), and nuclear buds (NBUDs)] of H2O2 (P < 0.01 and P < 0.001) when compared with U87MG. DHA at 100 µg/mL significantly affected cytostasis (P < 0.05) and increased DNA damage in the form of NPBs and MNi (P < 0.05) in both cell lines, whereas it decreased necrosis (P = 0.0251) in U87MG. Significant DHA-H2O2 interactions were observed for decreased necrosis (P = 0.0033) and DNA damage biomarkers (P < 0.0001) in the U87MG cell line and increased cytostasis (P < 0.0001) in the U118MG cell line. The effects of FFA also varied between the cell lines, with significant effects observed in decreased cytostasis (P = 0.0022) in the U87MG cell line, whereas increasing cytostasis (P = 0.0144) in the U118MG cell line. Overall, FFA exerted minimal effects on DNA damage biomarkers.
Collapse
Affiliation(s)
- Ann Chua
- Department of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, Australia; Nutrigenomics and Neurodegenerative Disease Prevention, Preventative Health Flagship, CSIRO, Animal, Food and Health Sciences, Adelaide, Australia
| | | | | | | | | |
Collapse
|
24
|
Miwa K, Hashimoto M, Hossain S, Katakura M, Shido O. Evaluation of the inhibitory effect of docosahexaenoic acid and arachidonic acid on the initial stage of amyloid β1-42 polymerization by fluorescence correlation spectroscopy. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/aad.2013.22009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Massaad CA. Neuronal and vascular oxidative stress in Alzheimer's disease. Curr Neuropharmacol 2011; 9:662-73. [PMID: 22654724 PMCID: PMC3263460 DOI: 10.2174/157015911798376244] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 10/06/2010] [Accepted: 10/11/2010] [Indexed: 01/22/2023] Open
Abstract
The brain is a highly metabolically active organ producing large amounts of reactive oxygen species (ROS). These ROS are kept in check by an elaborate network of antioxidants. Although ROS are necessary for signaling and synaptic plasticity, their uncontrolled levels cause oxidation of essential macromolecules such as membrane lipids, nucleic acids, enzymes and cytoskeletal proteins. Indeed, overproduction of ROS and/or failure of the antioxidant network lead to neuronal oxidative stress, a condition associated with not only aging but also Alzheimer's disease (AD). However, the specific source of excessive ROS production has not yet been identified. On one hand, amyloid beta (Aβ) has been extensively shown to act as an oxidant molecule. On the other hand, oxidative stress has been shown to precede and exacerbate Aβ pathology. This review will address the involvement of oxidative stress in the context of neuronal as well as vascular dysfunction associated with AD.
Collapse
Affiliation(s)
- Cynthia A Massaad
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
26
|
Giordano C, Sansone A, Masi A, Masci A, Mosca L, Chiaraluce R, Pasquo A, Consalvi V. Inhibition of Amyloid Peptide Fragment Aβ25-35 Fibrillogenesis and Toxicity by N-Terminal β-Amino Acid-Containing Esapeptides: Is Taurine Moiety Essential for In Vivo Effects? Chem Biol Drug Des 2011; 79:30-7. [DOI: 10.1111/j.1747-0285.2011.01259.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Hashimoto M, Hossain S. Neuroprotective and Ameliorative Actions of Polyunsaturated Fatty Acids Against Neuronal Diseases: Beneficial Effect of Docosahexaenoic Acid on Cognitive Decline in Alzheimer’s Disease. J Pharmacol Sci 2011; 116:150-62. [DOI: 10.1254/jphs.10r33fm] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|