1
|
Huang J, Wu F, Cao W, Chen Y, Yao Q, Cen P, Wang J, Hong L, Zhang X, Zhou R, Jin C, Tian M, Zhang H, Zhong Y. Ultrasmall iron-gallic acid coordination polymer nanoparticles for scavenging ROS and suppressing inflammation in tauopathy-induced Alzheimer's disease. Biomaterials 2025; 317:123042. [PMID: 39805185 DOI: 10.1016/j.biomaterials.2024.123042] [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] [Received: 07/08/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder globally, with no effective treatment available yet. A crucial pathological hallmark of AD is the accumulation of hyperphosphorylated tau protein, which is deteriorated by reactive oxygen species (ROS) and neuroinflammation in AD progression. Thus, alleviation of ROS and inflammation has become a potential therapeutic strategy in many studies. Herein, we reported ultrasmall coordination polymer nanoparticles formed by ferric ions and gallic acid (Fe-GA CPNs), which owned antioxidant and anti-inflammation properties for AD therapeutics. The facilely prepared Fe-GA CPNs exhibited remarkable superoxide dismutase-like, peroxidase-like enzyme activity, and ROS eliminating ability with great water solubility, compared with gallic acid. We demonstrated that Fe-GA CPNs effectively relieved oxidative stress, ameliorated inflammation by modulating microglial polarization towards anti-inflammation phenotype, and reduced hyperphosphorylated tau protein levels. Furthermore, Fe-GA CPNs treatment significantly improved cognitive function in tauopathy-induced AD rats, and achieved a neuroprotective effect against AD pathology. This study highlights the potential of coordination polymer nanoparticles as promising therapeutic candidates for AD and other tau-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiani Huang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Fei Wu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Wenzhao Cao
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Yuhan Chen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Qiong Yao
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Peili Cen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Lu Hong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Human Phenome Institute, Fudan University, Shanghai, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China; College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Abbas K, Mustafa M, Alam M, Habib S, Ahmad W, Adnan M, Hassan MI, Usmani N. Multi-target approach to Alzheimer's disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid- modulating mechanisms. Neurogenetics 2025; 26:39. [PMID: 40167826 DOI: 10.1007/s10048-025-00821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation, neurofibrillary tangles, neuroinflammation, and progressive cognitive decline, posing a significant global health challenge. Growing evidence suggests that dietary polyphenols may reduce the risk and progression of AD through multifaceted neuroprotective mechanisms. Polyphenols regulate amyloid proteostasis by inhibiting β/γ-secretase activity, preventing Aβ aggregation, and enhancing clearance pathways. Their strong antioxidant properties neutralize reactive oxygen species, chelate redox-active metals, and activate cytoprotective enzymes via Nrf2 signaling. This review examines the potential therapeutic targets, signaling pathways, and molecular mechanisms by which dietary polyphenols exert neuroprotective effects in AD, focusing on their roles in modulating amyloid proteostasis, oxidative stress, neuroinflammation, and cerebrovascular health. Polyphenols mitigate neuroinflammation by suppressing NF-κB signaling and upregulating brain-derived neurotrophic factor, supporting neuroplasticity and neurogenesis. They also enhance cerebrovascular health by improving cerebral blood flow, maintaining blood-brain barrier integrity, and modulating angiogenesis. This review examines the molecular and cellular pathways through which polyphenols exert neuroprotective effects, focusing on their antioxidant, anti-inflammatory, and amyloid-modulating roles. We also discuss their influence on key AD pathologies, including Aβ deposition, tau hyperphosphorylation, oxidative stress, and neuroinflammation. Insights from clinical and preclinical studies highlight the potential of polyphenols in preventing or slowing AD progression. Future research should explore personalized dietary strategies that integrate genetic and lifestyle factors to optimize the neuroprotective effects of polyphenols.
Collapse
Affiliation(s)
- Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'Il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
3
|
Regmi D, Haque S, Karim MRU, Stanic A, Du D. Inhibition of amyloid formation of prion fragment (106-128) by polyphenolic compounds. Biochim Biophys Acta Gen Subj 2025; 1869:130778. [PMID: 39988109 DOI: 10.1016/j.bbagen.2025.130778] [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] [Received: 07/12/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Prion diseases are characterized by the self-association and amyloid formation of misfolded prion proteins. Developing effective inhibitors of protein aggregation is critical for therapeutic intervention. In this study, we systematically evaluated a range of polyphenolic compounds as potential inhibitors of amyloid fibril formation of PrP(106-128), a prion fragment crucially involved in prion aggregation and propagation. Our findings demonstrate that the basic aromatic backbone structure of flavone alone is insufficient to inhibit PrP(106-128) amyloid formation. Remarkably, flavone molecules containing adjacent hydroxyl groups on the phenolic B or A ring efficiently inhibited PrP(106-128) fibrillization, whereas compounds lacking vicinal hydroxyl groups were less effective in inhibiting amyloid formation. Epigallocatechin-3-gallate (EGCG) was one of the most potent inhibitors found in this study, with the gallate moiety playing an active role in the inhibitory function. Our findings indicate a structure-dependent inhibition activity of the phenolic small molecules, where the number and positioning of hydroxyl groups on the phenyl ring play a pivotal role in inhibiting the aggregation of the peptide. The auto-oxidation of the catechol or pyrogallol moieties to form quinone structures, followed by their reaction with amino acid side chains of the peptide to form covalent adducts, likely account for the inhibitory activity of these phenolic compounds on PrP(106-128) amyloidogenesis. These results will help the design of novel polyphenolic molecules with optimized structural features as potent inhibitors of amyloid formation of both PrP(106-128) and the full-length prion proteins.
Collapse
Affiliation(s)
- Deepika Regmi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Seymour Haque
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Md Raza Ul Karim
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Aleksander Stanic
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Deguo Du
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA.
| |
Collapse
|
4
|
Kemeh MM, Furnelli AJ, Lazo ND. Differential Effects of Aβ Peptides on the Plasmin-Dependent Degradation of ApoE3 and ApoE4. ACS Chem Neurosci 2025; 16:1227-1237. [PMID: 40019771 DOI: 10.1021/acschemneuro.5c00065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025] Open
Abstract
The ApoE4 allele of apolipoprotein E (ApoE4) is the strongest hereditary predisposition to Alzheimer's disease, even though ApoE4 only differs from the more common ApoE3 by a single amino acid substitution. Previous studies have shown that ApoE4 is more susceptible to proteolytic degradation than ApoE3. This is an important finding because of ApoE's role in cholesterol homeostasis and lipid transport in the brain. The molecular determinants of the increased susceptibility of ApoE4 to proteolysis are unknown. Here, we apply a combination of spectrometric and spectroscopic methods to show that amyloid-β (Aβ) peptides, including Aβ(1-40) and Aβ(pyroE3-42), differentially modulate the plasmin-dependent degradation of ApoE3 and ApoE4. In particular, our data reveal that while the Aβ peptides do not affect the proteolysis of ApoE3, the peptides enhance the degradation of ApoE4 significantly. Overall, this work motivates therapeutic development that targets the Aβ-induced dysregulation of ApoE4 homeostasis in individuals carrying the ApoE4 allele.
Collapse
Affiliation(s)
- Merc M Kemeh
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Anthony J Furnelli
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Noel D Lazo
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| |
Collapse
|
5
|
Paul PS, Rathnam M, Khalili A, Cortez LM, Srinivasan M, Planel E, Cho JY, Wille H, Sim VL, Mok SA, Kar S. Temperature-Dependent Aggregation of Tau Protein Is Attenuated by Native PLGA Nanoparticles Under in vitro Conditions. Int J Nanomedicine 2025; 20:1999-2019. [PMID: 39968061 PMCID: PMC11834738 DOI: 10.2147/ijn.s494104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/01/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Hyperphosphorylation and aggregation of the microtubule-associated tau protein, which plays a critical role in many neurodegenerative diseases (ie, tauopathies) including Alzheimer's disease (AD), are known to be regulated by a variety of environmental factors including temperature. In this study we evaluated the effects of FDA-approved poly (D,L-lactide-co-glycolic) acid (PLGA) nanoparticles, which can inhibit amyloid-β aggregation/toxicity in cellular/animal models of AD, on temperature-dependent aggregation of 0N4R tau isoforms in vitro. Methods We have used a variety of biophysical (Thioflavin T kinetics, dynamic light scattering and asymmetric-flow field-flow fractionation), structural (fluorescence imaging and transmission electron microscopy) and biochemical (Filter-trap assay and detection of soluble protein) approaches, to evaluate the effects of native PLGA nanoparticles on the temperature-dependent tau aggregation. Results Our results show that the aggregation propensity of 0N4R tau increases significantly in a dose-dependent manner with a rise in temperature from 27°C to 40°C, as measured by lag time and aggregation rate. Additionally, the aggregation of 2N4R tau increases in a dose-dependent manner. Native PLGA significantly inhibits tau aggregation at all temperatures in a concentration-dependent manner, possibly by interacting with the aggregation-prone hydrophobic hexapeptide motifs of tau. Additionally, native PLGA is able to trigger disassembly of preformed 0N4R tau aggregates as a function of temperature from 27°C to 40°C. Conclusion These results, taken together, suggest that native PLGA nanoparticles can not only attenuate temperature-dependent tau aggregation but also promote disassembly of preformed aggregates, which increased with a rise of temperature. Given the evidence that temperature can influence tau pathology, we believe that native PLGA may have a unique potential to regulate tau abnormalities associated with AD-related pathology.
Collapse
Affiliation(s)
- Pallabi Sil Paul
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Mallesh Rathnam
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Aria Khalili
- Quantum and Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta, Canada
| | - Leonardo M Cortez
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Mahalashmi Srinivasan
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Emmanuel Planel
- Department of Psychiatry and Neurosciences, University of Laval, Quebec, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Holger Wille
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Valerie L Sim
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Sue-Ann Mok
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Satyabrata Kar
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| |
Collapse
|
6
|
Kola A, Vigni G, Lamponi S, Valensin D. Protective Contribution of Rosmarinic Acid in Rosemary Extract Against Copper-Induced Oxidative Stress. Antioxidants (Basel) 2024; 13:1419. [PMID: 39594560 PMCID: PMC11590892 DOI: 10.3390/antiox13111419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Rosemary extract (Rosmarinus officinalis) is a natural source of bioactive compounds with significant antioxidant properties. Among these, rosmarinic acid is celebrated for its potent antioxidant, anti-inflammatory, antimicrobial, and neuroprotective properties, making it a valuable component in both traditional medicine and modern therapeutic research. Neurodegenerative diseases like Alzheimer's and Parkinson's are closely linked to oxidative damage, and research indicates that rosmarinic acid may help protect neurons by mitigating this harmful process. Rosmarinic acid is able to bind cupric ions (Cu2+) and interfere with the production of reactive oxygen species (ROS) produced by copper through Fenton-like reactions. This study aims to further evaluate the contribution of rosmarinic acid within rosemary extract by comparing its activity to that of isolated rosmarinic acid. By using a detailed approach that includes chemical characterization, antioxidant capacity assessment, and neuroprotective activity testing, we have determined whether the combined components in rosemary extract enhance or differ from the effects of rosmarinic acid alone. This comparison is crucial for understanding whether the full extract offers added benefits beyond those of isolated rosmarinic acid in combating oxidative stress and Aβ-induced toxicity.
Collapse
Affiliation(s)
| | | | | | - Daniela Valensin
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.K.); (G.V.); (S.L.)
| |
Collapse
|
7
|
Middleton DA. NMR studies of amyloid interactions. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2024; 144-145:63-96. [PMID: 39645351 DOI: 10.1016/j.pnmrs.2024.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 12/09/2024]
Abstract
Amyloid fibrils are insoluble, fibrous nanostructures that accumulate extracellularly in biological tissue during the progression of several human disorders, including Alzheimer's disease (AD) and type 2 diabetes. Fibrils are assembled from protein monomers via the transient formation of soluble, cytotoxic oligomers, and have a common molecular architecture consisting of a spinal core of hydrogen-bonded protein β-strands. For the past 25 years, NMR spectroscopy has been at the forefront of research into the structure and assembly mechanisms of amyloid aggregates. Until the recent boom in fibril structure analysis by cryo-electron microscopy, solid-state NMR was unrivalled in its ability to provide atomic-level models of amyloid fibril architecture. Solution-state NMR has also provided complementary information on the early stages in the amyloid assembly mechanism. Now, both NMR modalities are proving to be valuable in unravelling the complex interactions between amyloid species and a diverse range of physiological metal ions, molecules and surfaces that influence the assembly pathway, kinetics, morphology and clearance in vivo. Here, an overview is presented of the main applications of solid-state and solution-state NMR for studying the interactions between amyloid proteins and biomembranes, glycosaminoglycan polysaccharides, metal ions, polyphenols, synthetic therapeutics and diagnostics. Key NMR methodology is reviewed along with examples of how to overcome the challenges of detecting interactions with aggregating proteins. The review heralds this new role for NMR in providing a comprehensive and pathologically-relevant view of the interactions between protein and non-protein components of amyloid. Coverage of both solid- and solution-state NMR methods and applications herein will be informative and valuable to the broad communities that are interested in amyloid proteins.
Collapse
Affiliation(s)
- David A Middleton
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom.
| |
Collapse
|
8
|
El Rayess Y, Nehme N, Azzi-Achkouty S, Julien SG. Wine Phenolic Compounds: Chemistry, Functionality and Health Benefits. Antioxidants (Basel) 2024; 13:1312. [PMID: 39594454 PMCID: PMC11591289 DOI: 10.3390/antiox13111312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Wine phenolic compounds, often known as polyphenols, are a diverse group of secondary bioactive compounds derived from grapes. They play a crucial role in defining the sensory characteristics, functionality, and health benefits of wine. This review explores the complex chemistry of these compounds, focusing on key classes such as flavonoids, which include flavanones, flavonols, anthocyanins, and flavan-3-ols, and non-flavonoids, such as hydroxycinnamic acids, hydroxybenzoic acids, and stilbenes. The health benefits of wine phenolics, particularly their antioxidant and anti-inflammatory properties, are also discussed in relation to preventing and reducing the risk of non-communicable diseases (NCDs) such as cardiovascular diseases, cancers, and neurodegenerative conditions. Furthermore, this review summarized the most current data from human population-based research that investigated the bioactivity of these red wine phytochemicals with relevant health benefits for NCDs. Finally, this review proposes some perspectives for future research to better understand the bioavailability, metabolism, and long-term health effects of these compounds.
Collapse
Affiliation(s)
- Youssef El Rayess
- Department of Agriculture and Food Engineering, School of Engineering, Holy Spirit University of Kaslik, Jounieh P.O. Box 446, Lebanon;
| | - Nancy Nehme
- Faculty of Agricultural Engineering and Veterinary Medicine, Lebanese University, Dekwaneh P.O. Box 446, Lebanon;
| | - Samar Azzi-Achkouty
- Department of Agriculture and Food Engineering, School of Engineering, Holy Spirit University of Kaslik, Jounieh P.O. Box 446, Lebanon;
| | - Sofi G. Julien
- Department of Nutrition and Food Sciences, Faculty of Art and Sciences, Holy Spirit University of Kaslik, Jounieh P.O. Box 446, Lebanon
| |
Collapse
|
9
|
Cerasuolo M, Di Meo I, Auriemma MC, Paolisso G, Papa M, Rizzo MR. Exploring the Dynamic Changes of Brain Lipids, Lipid Rafts, and Lipid Droplets in Aging and Alzheimer's Disease. Biomolecules 2024; 14:1362. [PMID: 39595539 PMCID: PMC11591903 DOI: 10.3390/biom14111362] [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] [Received: 09/06/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Aging induces complex changes in the lipid profiles across different areas of the brain. These changes can affect the function of brain cells and may contribute to neurodegenerative diseases such as Alzheimer's disease. Research shows that while the overall lipid profile in the human brain remains quite steady throughout adulthood, specific changes occur with age, especially after the age of 50. These changes include a slow decline in total lipid content and shifts in the composition of fatty acids, particularly in glycerophospholipids and cholesterol levels, which can vary depending on the brain region. Lipid rafts play a crucial role in maintaining membrane integrity and facilitating cellular signaling. In the context of Alzheimer's disease, changes in the composition of lipid rafts have been associated with the development of the disease. For example, alterations in lipid raft composition can lead to increased accumulation of amyloid β (Aβ) peptides, contributing to neurotoxic effects. Lipid droplets store neutral lipids and are key for cellular energy metabolism. As organisms age, the dynamics of lipid droplets in the brain change, with evidence suggesting a decline in metabolic activity over time. This reduced activity may lead to an imbalance in lipid synthesis and mobilization, contributing to neurodegenerative processes. In model organisms like Drosophila, studies have shown that lipid metabolism in the brain can be influenced by diet and insulin signaling pathways, crucial for maintaining metabolic balance. The interplay between lipid metabolism, oxidative stress, and inflammation is critical in the context of aging and Alzheimer's disease. Lipid peroxidation, a consequence of oxidative stress, can lead to the formation of reactive aldehydes that further damage neurons. Inflammatory processes can also disrupt lipid metabolism, contributing to the pathology of AD. Consequently, the accumulation of oxidized lipids can affect lipid raft integrity, influencing signaling pathways involved in neuronal survival and function.
Collapse
Affiliation(s)
- Michele Cerasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Maria Chiara Auriemma
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| |
Collapse
|
10
|
Pennisi M, Cantone M, Cappellani F, Concerto C, Ferri R, Godos J, Grosso G, Lanza G, Rodolico A, Torrisi G, Al-Qahtani WH, Fisicaro F, Bella R. Combined Effect of Red Wine and Mocha Pot Coffee in Mild Vascular Cognitive Impairment. Exp Gerontol 2024; 194:112498. [PMID: 38901216 DOI: 10.1016/j.exger.2024.112498] [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] [Received: 10/11/2023] [Revised: 03/27/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVES Moderate daily mocha pot coffee intake has been associated with better mood and cognition in patients with mild vascular cognitive impairment (VCI). Similarly, moderate red wine consumption has shown protective effects on cognitive disorders, including Alzheimer's disease and vascular dementia. The aim of this study was to explore the synergistic relation between red wine and coffee intake on mood and cognitive status in mild VCI patients at risk for dementia. METHODS A total of 300 non-demented older patients with mild VCI were asked for coffee and red wine consumption and administered with the 17-items Hamilton Depression Rating Scale (HDRS), the Mini Mental State Examination (MMSE), and the Stroop Color-Word Interference Test (Stroop T), as well as the Activities of Daily Living (ADL) and the Instrumental ADL to measure their mood status, cognitive performance, and functional independence. Linear regression models were used to test the association between variables. RESULTS Moderate wine drinkers tended to show the best Stroop T score at any level of coffee consumption; conversely, heavy wine consumers performed worse at the Stroop T, especially in patients reporting high coffee intake. Moderate drinkers of both coffee and wine showed the lowest HDRS scores. Finally, a progressive increase in MMSE score was evident with increasing coffee consumption, which peaks when combined with a moderate wine consumption. CONCLUSIONS Daily mocha pot coffee and red wine intake seem to be synergistically associated with global cognition, executive functioning, and mood status in patients with mild VCI; the association was not linear, resulting in a protective direction for moderate intake and detrimental for heavy consumption. Future studies are needed to further corroborate the present findings and the potential long-term protective effects of these dietary compounds over time.
Collapse
Affiliation(s)
- Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Mariagiovanna Cantone
- Neurology Unit, Policlinico University Hospital "G. Rodolico-San Marco", Catania, Italy
| | - Francesco Cappellani
- Ophthalmology Unit, Policlinico University Hospital "G. Rodolico-San Marco", Catania, Italy
| | - Carmen Concerto
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Raffaele Ferri
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Troina, Italy
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Giuseppe Lanza
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Troina, Italy; Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy.
| | - Alessandro Rodolico
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giulia Torrisi
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Wahidah H Al-Qahtani
- Department of Food Sciences & Nutrition, College of Food & Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies "G. F. Ingrassia", University of Catania, Catania, Italy
| |
Collapse
|
11
|
Kemeh MM, Lazo ND. Highly toxic Aβ begets more Aβ. Neural Regen Res 2024; 19:1871-1872. [PMID: 38227503 DOI: 10.4103/1673-5374.390983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/08/2023] [Indexed: 01/17/2024] Open
Affiliation(s)
- Merc M Kemeh
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, MA, USA
| | | |
Collapse
|
12
|
Xie S, Liang Y, Song Y, Li T, Jia J. Repurposing Anidulafungin for Alzheimer's Disease via Fragment-Based Drug Discovery. ACS Chem Neurosci 2024; 15:2995-3008. [PMID: 39096284 PMCID: PMC11342299 DOI: 10.1021/acschemneuro.4c00150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/19/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
The misfolding and aggregation of beta-amyloid (Aβ) peptides have been implicated as key pathogenic events in the early stages of Alzheimer's disease (AD). Inhibiting Aβ aggregation represents a potential disease-modifying therapeutic approach to AD treatment. Previous studies have identified various molecules that inhibit Aβ aggregation, some of which share common chemical substructures (fragments) that may be key to their inhibitory activity. Employing fragment-based drug discovery (FBDD) methods may facilitate the identification of these fragments, which can subsequently be used to screen new inhibitors and provide leads for further drug development. In this study, we used an in silico FBDD approach to identify 17 fragment clusters that are significantly enriched among Aβ aggregation inhibitors. These fragments were then used to screen anti-infective agents, a promising drug class for repurposing against amyloid aggregation. This screening process identified 16 anti-infective drugs, 5 of which were chosen for further investigation. Among the 5 candidates, anidulafungin, an antifungal compound, showed high efficacy in inhibiting Aβ aggregation in vitro. Kinetic analysis revealed that anidulafungin selectively blocks the primary nucleation step of Aβ aggregation, substantially delaying Aβ fibril formation. Cell viability assays demonstrated that anidulafungin can reduce the toxicity of oligomeric Aβ on BV2 microglia cells. Molecular docking simulations predicted that anidulafungin interacted with various Aβ species, including monomers, oligomers, and fibrils, potentially explaining its activity against Aβ aggregation and toxicity. This study suggests that anidulafungin is a potential drug to be repurposed for AD, and FBDD is a promising approach for discovering drugs to combat Aβ aggregation.
Collapse
Affiliation(s)
- Siqi Xie
- Innovation
Center for Neurological Disorders and Department of Neurology, Xuanwu
Hospital, Capital Medical University, National
Clinical Research Center for Geriatric Diseases, Beijing 100053, P. R. China
| | - Yumei Liang
- Innovation
Center for Neurological Disorders and Department of Neurology, Xuanwu
Hospital, Capital Medical University, National
Clinical Research Center for Geriatric Diseases, Beijing 100053, P. R. China
| | - Yang Song
- Innovation
Center for Neurological Disorders and Department of Neurology, Xuanwu
Hospital, Capital Medical University, National
Clinical Research Center for Geriatric Diseases, Beijing 100053, P. R. China
| | - Tingting Li
- Innovation
Center for Neurological Disorders and Department of Neurology, Xuanwu
Hospital, Capital Medical University, National
Clinical Research Center for Geriatric Diseases, Beijing 100053, P. R. China
| | - Jianping Jia
- Innovation
Center for Neurological Disorders and Department of Neurology, Xuanwu
Hospital, Capital Medical University, National
Clinical Research Center for Geriatric Diseases, Beijing 100053, P. R. China
- Beijing
Key Laboratory of Geriatric Cognitive Disorders, Beijing 100053, P. R. China
- Clinical
Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing 100053, P. R. China
- Center
of Alzheimer’s Disease, Beijing Institute of Brain Disorders,
Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100053, P. R. China
- Key
Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, P. R. China
| |
Collapse
|
13
|
Waugh ML, Wolf LM, Moore KA, Servoss SL, Moss MA. Rationally Designed Peptoid Inhibitors of Amyloid-β Oligomerization. Chembiochem 2024; 25:e202400060. [PMID: 38715149 PMCID: PMC11219258 DOI: 10.1002/cbic.202400060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
While plaques comprised of fibrillar Aβ aggregates are hallmarks of Alzheimer's disease, soluble Aβ oligomers present higher neurotoxicity. Thus, one therapeutic approach is to prevent the formation of Aβ oligomers and reduce their associated harmful effects. We have proposed a peptoid mimic of the Aβ hydrophobic KLVFF core as an ideal candidate aggregation inhibitor due to its ability to evade proteolytic degradation via repositioning of the side chain from the α-carbon to the amide nitrogen. This peptoid, JPT1, utilizes chiral sidechains to achieve a helical structure, while C-terminal addition of two phenylalanine residues places aromatic groups on two sides of the helix with spacing designed to facilitate interaction with amyloid β-sheet structure. We have previously shown that JPT1 modulates Aβ fibril formation. Here, we demonstrate that JPT1 also modulates Aβ oligomerization, and we explore the role of the charge on the linker between the KLVFF mimic and the extended aromatic residues. Additionally, we demonstrate that peptoid-induced changes in Aβ oligomerization correlate with attenuation of oligomer-induced nuclear factor-κB activation in SH-SY5Y human neuroblastoma cells. These findings support the therapeutic potential of peptoids to target early stages of Aβ aggregation and impact the associated Aβ-induced cellular response.
Collapse
Affiliation(s)
- Mihyun Lim Waugh
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
| | - Lauren M Wolf
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
| | - Kelly A Moore
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
| | - Shannon L Servoss
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR 72701, USA
| | - Melissa A Moss
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
- Department of Chemical Engineering, University of South Carolina, 2C02 Swearingen Engineering Center, Columbia, SC 29208, USA
| |
Collapse
|
14
|
Lu X, Lu J, Li S, Feng S, Wang Y, Cui L. The Role of Liquid-Liquid Phase Separation in the Accumulation of Pathological Proteins: New Perspectives on the Mechanism of Neurodegenerative Diseases. Aging Dis 2024; 16:769-786. [PMID: 38739933 PMCID: PMC11964424 DOI: 10.14336/ad.2024.0209] [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] [Received: 02/22/2024] [Accepted: 04/20/2024] [Indexed: 05/16/2024] Open
Abstract
It is widely accepted that living organisms form highly dynamic membrane-less organelles (MLOS) with various functions through phase separation, and the indispensable role that phase separation plays in the mechanisms of normal physiological functions and pathogenesis is gradually becoming clearer. Pathological aggregates, regarded as hallmarks of neurodegenerative diseases, have been revealed to be closely related to aberrant phase separation. Specific proteins are assembled into condensates and transform into insoluble inclusions through aberrant phase separation, contributing to the development of diseases. In this review, we present an overview of the progress of phase separation research, involving its biological mechanisms and the status of research in neurodegenerative diseases, focusing on five main disease-specific proteins, tau, TDP-43, FUS, α-Syn and HTT, and how exactly these proteins reside within dynamic liquid-like compartments and thus turn into solid deposits. Further studies will yield new perspectives for understanding the aggregation mechanisms and potential therapeutic strategies, and future research directions are anticipated.
Collapse
Affiliation(s)
- Xingyu Lu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Jiongtong Lu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Shengnan Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Sifan Feng
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- The Marine Biomedical Research Institute of Guangdong, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
15
|
Moorthy H, Kamala N, Ramesh M, Govindaraju T. Biphasic modulation of tau liquid-liquid phase separation by polyphenols. Chem Commun (Camb) 2024; 60:4334-4337. [PMID: 38545836 DOI: 10.1039/d4cc00473f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Molecular tools that modulate tau liquid-liquid phase separation (LLPS) promise to treat tauopathies. We screened a set of polyphenols and demonstrated concentration-dependent biphasic modulation of tau LLPS by gallic acid (GA), showcasing its ability to expedite the liquid-to-gel transition in tau condensates and effectively impede the formation of deleterious fibrillar aggregates.
Collapse
Affiliation(s)
- Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru 560064, Karnataka, India.
| | - Nimsha Kamala
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru 560064, Karnataka, India.
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru 560064, Karnataka, India.
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru 560064, Karnataka, India.
| |
Collapse
|
16
|
Clemente-Suárez VJ, Redondo-Flórez L, Beltrán-Velasco AI, Belinchón-deMiguel P, Ramos-Campo DJ, Curiel-Regueros A, Martín-Rodríguez A, Tornero-Aguilera JF. The Interplay of Sports and Nutrition in Neurological Health and Recovery. J Clin Med 2024; 13:2065. [PMID: 38610829 PMCID: PMC11012304 DOI: 10.3390/jcm13072065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
This comprehensive review explores the dynamic relationship between sports, nutrition, and neurological health. Focusing on recent clinical advancements, it examines how physical activity and dietary practices influence the prevention, treatment, and rehabilitation of various neurological conditions. The review highlights the role of neuroimaging in understanding these interactions, discusses emerging technologies in neurotherapeutic interventions, and evaluates the efficacy of sports and nutritional strategies in enhancing neurological recovery. This synthesis of current knowledge aims to provide a deeper understanding of how lifestyle factors can be integrated into clinical practices to improve neurological outcomes.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (A.C.-R.); (J.F.T.-A.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, Villaviciosa de Odón, 28670 Madrid, Spain;
| | | | - Pedro Belinchón-deMiguel
- Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain;
| | - Domingo Jesús Ramos-Campo
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Science-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
| | - Agustín Curiel-Regueros
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (A.C.-R.); (J.F.T.-A.)
| | - Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (A.C.-R.); (J.F.T.-A.)
| | - José Francisco Tornero-Aguilera
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (A.C.-R.); (J.F.T.-A.)
| |
Collapse
|
17
|
Rani A, Zia-Ul-Sabah, Tabassum F, Sharma AK. Molecular interplay between phytoconstituents of Ficus Racemosa and neurodegenerative diseases. Eur J Neurosci 2024; 59:1833-1847. [PMID: 38217338 DOI: 10.1111/ejn.16250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/15/2024]
Abstract
Neurodegenerative diseases (NDs) are a significant global health concern, primarily affecting middle and older populations. Recently, there has been growing interest in herbal therapeutics as a potential approach to address diverse neuropathological conditions. Despite the widespread prevalence of NDs, limited phytochemical has been reported for their promising therapeutic potential with distinct underlying mechanisms. Additionally, the intricate molecular pathways influenced by herbal phytoconstituents, particularly in neurodegenerative disorders, are also not well documented. This report explores the phytoconstituents of Ficus racemosa (F. racemosa), an unfamiliar plant of the Moraceae family, for their potential interactions with pathological pathways of NDs. The influential phytoconstituents of F. racemosa, including polyphenols, glycosides, terpenoids, and furocoumarin, have been reported for targeting diverse pathological states. We proposed the most convincing molecular interplay between leading phytoconstituents and detrimental signalling cascades. However, extensive research is required to thoroughly understand the phytochemical persuaded intricate molecular pathway. The comprehensive evidence strongly suggests that F. racemosa and its natural compounds could be valuable in treating NDs. This points towards an exciting path for future research and the development of potential treatments based on a molecular level.
Collapse
Affiliation(s)
- Anu Rani
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, India
| | - Zia-Ul-Sabah
- Department of Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Fauzia Tabassum
- Department of Pharmacology, Vision College, Riyadh, Saudi Arabia
| | - Arun K Sharma
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, India
| |
Collapse
|
18
|
Tang J, Sun R, Wan J, Xu Z, Zou Y, Zhang Q. Atomic insights into the inhibition of R3 domain of tau protein by epigallocatechin gallate, quercetin and gallic acid. Biophys Chem 2024; 305:107142. [PMID: 38088006 DOI: 10.1016/j.bpc.2023.107142] [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] [Received: 08/25/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024]
Abstract
Inhibiting tau protein aggregation has become a prospective avenue for the therapeutic development of tauopathies. The third microtubule-binding repeat (R3) domain of tau is confirmed as the most aggregation-favorable fragment of the whole protein. As dimerization is the first step of the aggregation of tau into amyloid fibrils, impeding the dimerization of the R3 domain is critical to prevent the full-length tau aggregation. Natural polyphenol small molecules epigallocatechin gallate (EGCG), quercetin (QE) and gallic acid (GA) are proven to inhibit the aggregation of the full-length recombinant tau (For EGCG and QE) or the R3 domain (For GA) of tau in vitro. However, the underlying molecular mechanisms of the inhibitive effects on the R3 domain of tau remain largely unknown. In this study, we conducted numerous all-atom molecular dynamics simulations on R3 dimers with and without EGCG, QE or GA, respectively. The results reveal that all three molecules can effectively decrease the β structure composition of the R3 dimer, induce the dimer to adopt loosely-packed conformations, and weaken interchain interactions, thus impeding the dimerization of the R3 peptide chains. The specific preferentially binding sites for the three molecules exhibit similarities and differences. Hydrophobic, π-π stacking and hydrogen-bonding interactions collectively drive EGCG, QE and GA respectively binding on the R3 dimer, while QE also binds with the dimer through cation-π interaction. Given the incurable nature of tauopathies hitherto, our research provides helpful knowledge for the development of drugs to treat tauopathies.
Collapse
Affiliation(s)
- Jiaxing Tang
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Ruiqing Sun
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Jiaqian Wan
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Zhengdong Xu
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Yu Zou
- Department of Sport and Exercise Science, College of Education, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China.
| | - Qingwen Zhang
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China.
| |
Collapse
|
19
|
Shah MA, Faheem HI, Hamid A, Yousaf R, Haris M, Saleem U, Shah GM, Alhasani RH, Althobaiti NA, Alsharif I, Silva AS. The entrancing role of dietary polyphenols against the most frequent aging-associated diseases. Med Res Rev 2024; 44:235-274. [PMID: 37486109 DOI: 10.1002/med.21985] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 01/27/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
Aging, a fundamental physiological process influenced by innumerable biological and genetic pathways, is an important driving factor for several aging-associated disorders like diabetes mellitus, osteoporosis, cancer, and neurodegenerative diseases including Alzheimer's and Parkinson's diseases. In the modern era, the several mechanisms associated with aging have been deeply studied. Treatment and therapeutics for age-related diseases have also made considerable advances; however, for the effective and long-lasting treatment, nutritional therapy particularly including dietary polyphenols from the natural origin are endorsed. These dietary polyphenols (e.g., apigenin, baicalin, curcumin, epigallocatechin gallate, kaempferol, quercetin, resveratrol, and theaflavin), and many other phytochemicals target certain molecular, genetic mechanisms. The most common pathways of age-associated diseases are mitogen-activated protein kinase, reactive oxygen species production, nuclear factor kappa light chain enhancer of activated B cells signaling pathways, metal chelation, c-Jun N-terminal kinase, and inflammation. Polyphenols slow down the course of aging and help in combatting age-linked disorders. This exemplified in the form of clinical trials on specific dietary polyphenols in various aging-associated diseases. With this context in mind, this review reveals the new insights to slow down the aging process, and consequently reduce some classic diseases associated with age such as aforementioned, and targeting age-associated diseases by the activities of dietary polyphenols of natural origin.
Collapse
Affiliation(s)
| | - Hafiza Ishmal Faheem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Ayesha Hamid
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Rimsha Yousaf
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Muhammad Haris
- Faculty of Pharmaceutical Sciences, Universiteit Gent, Ghent, Belgium
| | - Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Ghulam Mujtaba Shah
- Department of Botany, Faculty of Health and Biological Sciences, Hazara University, Mansehra, Pakistan
| | - Reem H Alhasani
- Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Norah A Althobaiti
- Department of Biology, College of Science and Humanities, Shaqra University, Al-Quwaiiyah, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ana Sanches Silva
- National Institute for Agrarian and Veterinary Research (INIAV), I.P., Rua dos Lágidos, Lugar da Madalena, Vairão, Vila do Conde, Portugal
- University of Coimbra, Faculty of Pharmacy, Polo III, Azinhaga de St Comba, Coimbra, Portugal
- Centre for Animal Science Studies (CECA), ICETA, University of Porto, Porto, Portugal
| |
Collapse
|
20
|
Dassamiour S, Bensaad MS, Ghebache W. Utility of phenolic acids in neurological disorders. ADVANCEMENT OF PHENOLIC ACIDS IN DRUG DISCOVERY 2024:295-344. [DOI: 10.1016/b978-0-443-18538-0.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Pluta R, Miziak B, Czuczwar SJ. Apitherapy in Post-Ischemic Brain Neurodegeneration of Alzheimer's Disease Proteinopathy: Focus on Honey and Its Flavonoids and Phenolic Acids. Molecules 2023; 28:5624. [PMID: 37570596 PMCID: PMC10420307 DOI: 10.3390/molecules28155624] [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] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Neurodegeneration of the brain after ischemia is a major cause of severe, long-term disability, dementia, and mortality, which is a global problem. These phenomena are attributed to excitotoxicity, changes in the blood-brain barrier, neuroinflammation, oxidative stress, vasoconstriction, cerebral amyloid angiopathy, amyloid plaques, neurofibrillary tangles, and ultimately neuronal death. In addition, genetic factors such as post-ischemic changes in genetic programming in the expression of amyloid protein precursor, β-secretase, presenilin-1 and -2, and tau protein play an important role in the irreversible progression of post-ischemic neurodegeneration. Since current treatment is aimed at preventing symptoms such as dementia and disability, the search for causative therapy that would be helpful in preventing and treating post-ischemic neurodegeneration of Alzheimer's disease proteinopathy is ongoing. Numerous studies have shown that the high contents of flavonoids and phenolic acids in honey have antioxidant, anti-inflammatory, anti-apoptotic, anti-amyloid, anti-tau protein, anticholinesterase, serotonergic, and AMPAK activities, influencing signal transmission and neuroprotective effects. Notably, in many preclinical studies, flavonoids and phenolic acids, the main components of honey, were also effective when administered after ischemia, suggesting their possible use in promoting recovery in stroke patients. This review provides new insight into honey's potential to prevent brain ischemia as well as to ameliorate damage in advanced post-ischemic brain neurodegeneration.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (S.J.C.)
| | | | | |
Collapse
|
22
|
Zheng Y, Zheng C, Tu W, Jiang Y, Lin H, Chen W, Lee Q, Zheng W. Danshensu inhibits Aβ aggregation and neurotoxicity as one of the main prominent features of Alzheimer's disease. Int J Biol Macromol 2023:125294. [PMID: 37315666 DOI: 10.1016/j.ijbiomac.2023.125294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
It has been found that the main cause of neurodegenerative proteinopathies, especially Alzheimer's disease (AD) is the formation of Aβ amyloid plaques, which can be regulated by application of potential small molecules. In the present study, we aimed to investigate the inhibitory effect of danshensu on Aβ(1-42) aggregation and relevant apoptotic pathway in neurons. A broad range of spectroscopic, theoretical, and cellular assays were done to investigate the anti-amyloidogenic characteristics of danshensu. It was found that danshensu triggers its inhibitory effect against Aβ(1-42) aggregation through modulation of hydrophobic patches as well as structural and morphological changes through a stacking interaction. Furthermore, it was observed that incubation of Aβ(1-42) samples with danshensu during aggregation process recovered the cell viability and mitigated the expression of caspase-3 mRNA and protein as well caspase-3 activity deregulated by Aβ(1-42) amyloid fibrils alone. In general, obtained data showed that danshensu potentially inhibits Aβ(1-42) aggregation and associated proteinopathies through regulation of apoptotic pathway in a concentration-dependent manner. Therefore, danshensu may be used as a promising biomolecule against the Aβ aggregation and associated proteinopathies, which can be further analyzed in the future studies for the treatment of AD.
Collapse
Affiliation(s)
- Yuyin Zheng
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Cheng Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Wenzhan Tu
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yiwei Jiang
- Alberta Institute, Wenzhou Medical University, Wenzhou 325000, China
| | - Haiyan Lin
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wangchao Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qian Lee
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wu Zheng
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
23
|
Chu Z, Han S, Luo Y, Zhou Y, Zhu L, Luo F. Targeting gut-brain axis by dietary flavonoids ameliorate aging-related cognition decline: Evidences and mechanisms. Crit Rev Food Sci Nutr 2023; 64:10281-10302. [PMID: 37300491 DOI: 10.1080/10408398.2023.2222404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Aging-related cognitive impairment, mainly Alzheimer's disease (AD), has been widely studied. However, effective prevention and treatment methods are still lacking. In recent years, researchers have observed beneficial effects of plant-based supplements, such as flavonoids, on cognitive protection. This provides a new clue for the prevention of cognitive dysfunction. Studies have shown that dietary flavonoids have neuroprotective effects, but the mechanism is not clear. In this review, we systematically reviewed the research progress on the effects of dietary flavonoids on gut microbes and their metabolites, and concluded that flavonoids could improve cognitive function through the gut-brain axis. Flavonoids can be absorbed through the intestine, cross the blood-brain barrier, and enter the brain tissue. Flavonoids can inhibit the expression and secretion of inflammatory factors in brain tissue, reduce the damage caused by oxidative stress, clear neural damage proteins and inhibit neuronal apoptosis, thereby ameliorating age-related cognitive disorders. Future work will continue to explore the gut-brain axis and target genes regulated by flavonoids. In addition, clinical research and its mechanisms need to be further explored to provide solutions or advise for patients with cognitive impairment.
Collapse
Affiliation(s)
- Zhongxing Chu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Shuai Han
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Yi Luo
- Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Lingfeng Zhu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
24
|
Firouzi R, Sowlati-Hashjin S, Chávez-García C, Ashouri M, Karimi-Jafari MH, Karttunen M. Identification of Catechins' Binding Sites in Monomeric A β42 through Ensemble Docking and MD Simulations. Int J Mol Sci 2023; 24:ijms24098161. [PMID: 37175868 PMCID: PMC10179585 DOI: 10.3390/ijms24098161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/09/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
The assembly of the amyloid-β peptide (Aβ) into toxic oligomers and fibrils is associated with Alzheimer's disease and dementia. Therefore, disrupting amyloid assembly by direct targeting of the Aβ monomeric form with small molecules or antibodies is a promising therapeutic strategy. However, given the dynamic nature of Aβ, standard computational tools cannot be easily applied for high-throughput structure-based virtual screening in drug discovery projects. In the current study, we propose a computational pipeline-in the framework of the ensemble docking strategy-to identify catechins' binding sites in monomeric Aβ42. It is shown that both hydrophobic aromatic interactions and hydrogen bonding are crucial for the binding of catechins to Aβ42. Additionally, it has been found that all the studied ligands, especially EGCG, can act as potent inhibitors against amyloid aggregation by blocking the central hydrophobic region of Aβ. Our findings are evaluated and confirmed with multi-microsecond MD simulations. Finally, it is suggested that our proposed pipeline, with low computational cost in comparison with MD simulations, is a suitable approach for the virtual screening of ligand libraries against Aβ.
Collapse
Affiliation(s)
- Rohoullah Firouzi
- Department of Physical Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Tehran 1496813151, Iran
| | | | - Cecilia Chávez-García
- Department of Chemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
- The Centre of Advanced Materials and Biomaterials Research, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
| | - Mitra Ashouri
- Department of Physical Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6619, Iran
| | - Mohammad Hossein Karimi-Jafari
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran P.O. Box 14155-6619, Iran
| | - Mikko Karttunen
- Department of Chemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
- The Centre of Advanced Materials and Biomaterials Research, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
- Department of Physics and Astronomy, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
| |
Collapse
|
25
|
Tang J, Zou Y, Gong Y, Xu Z, Wan J, Wei G, Zhang Q. Molecular Mechanism in the Disruption of Chronic Traumatic Encephalopathy-Related R3-R4 Tau Protofibril by Quercetin and Gallic Acid: Similarities and Differences. ACS Chem Neurosci 2023; 14:897-908. [PMID: 36749931 DOI: 10.1021/acschemneuro.2c00688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a unique progressive neurodegenerative tauopathy pathologically related to the aggregation of the tau protein to neurofibrillary tangles. Disrupting tau oligomers (protofibril) is a promising strategy to prevent CTE. Quercetin (QE) and gallic acid (GA), two polyphenol small molecules abundant in natural crops, were proved to inhibit recombinant tau and the R3 fragment of human full-length tau in vitro. However, their disruptive effect on CTE-related protofibril and the underlying molecular mechanism remain elusive. Cryo-electron microscopy resolution reveals that the R3-R4 fragment of tau forms the core of the CTE-related tau protofibril. In this study, we conducted extensive all-atom molecular dynamics simulations on CTE-related R3-R4 tau protofibril with and without QE/GA molecules. The results disclose that both QE and GA can disrupt the global structure of the protofibril, while GA shows a relatively strong effect. The binding sites, exact binding patterns, and disruptive modes for the two molecules show similarities and differences. Strikingly, both QE and GA can insert into the hydrophobic cavity of the protofibril, indicating they have the potential to compete for the space in the cavity with aggregation cofactors unique to CTE-related protofibril and thus impede the further aggregation of the tau protein. Due to relatively short time scale, our study captures the early disruptive mechanism of CTE-related R3-R4 tau protofibril by QE/GA. However, our research does provide valuable knowledge for the design of supplements or drugs to prevent or delay the development of CTE.
Collapse
Affiliation(s)
- Jiaxing Tang
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Yu Zou
- Department of Sport and Exercise Science, College of Education, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yehong Gong
- School of Sports Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Zhengdong Xu
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Jiaqian Wan
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People's Republic of China
| | - Qingwen Zhang
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| |
Collapse
|
26
|
Andreo-López MC, Contreras-Bolívar V, Muñoz-Torres M, García-Fontana B, García-Fontana C. Influence of the Mediterranean Diet on Healthy Aging. Int J Mol Sci 2023; 24:4491. [PMID: 36901921 PMCID: PMC10003249 DOI: 10.3390/ijms24054491] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
The life expectancy of the global population has increased. Aging is a natural physiological process that poses major challenges in an increasingly long-lived and frail population. Several molecular mechanisms are involved in aging. Likewise, the gut microbiota, which is influenced by environmental factors such as diet, plays a crucial role in the modulation of these mechanisms. The Mediterranean diet, as well as the components present in it, offer some proof of this. Achieving healthy aging should be focused on the promotion of healthy lifestyle habits that reduce the development of pathologies that are associated with aging, in order to increase the quality of life of the aging population. In this review we analyze the influence of the Mediterranean diet on the molecular pathways and the microbiota associated with more favorable aging patterns, as well as its possible role as an anti-aging treatment.
Collapse
Affiliation(s)
| | - Victoria Contreras-Bolívar
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), 18014 Granada, Spain
| | - Manuel Muñoz-Torres
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), 18014 Granada, Spain
- CIBER on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 18012 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), 18014 Granada, Spain
- CIBER on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 18012 Granada, Spain
- Department of Cell Biology, University of Granada, 18016 Granada, Spain
| | - Cristina García-Fontana
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), 18014 Granada, Spain
- CIBER on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 18012 Granada, Spain
| |
Collapse
|
27
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Honey and Alzheimer's Disease-Current Understanding and Future Prospects. Antioxidants (Basel) 2023; 12:427. [PMID: 36829985 PMCID: PMC9952506 DOI: 10.3390/antiox12020427] [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: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, has been a global concern. AD is associated with the involvement of the central nervous system that causes the characteristic impaired memory, cognitive deficits, and behavioral abnormalities. These abnormalities caused by AD is known to be attributed by extracellular aggregates of amyloid beta plaques and intracellular neurofibrillary tangles. Additionally, genetic factors such as abnormality in the expression of APOE, APP, BACE1, PSEN-1, and PSEN-2 play a role in the disease. As the current treatment aims to treat the symptoms and to slow the disease progression, there has been a continuous search for new nutraceutical agent or medicine to help prevent and cure AD pathology. In this quest, honey has emerged as a powerful nootropic agent. Numerous studies have demonstrated that the high flavonoids and phenolic acids content in honey exerts its antioxidant, anti-inflammatory, and neuroprotective properties. This review summarizes the effect of main flavonoid compounds found in honey on the physiological functioning of the central nervous system, and the effect of honey intake on memory and cognition in various animal model. This review provides a new insight on the potential of honey to prevent AD pathology, as well as to ameliorate the damage in the developed AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
28
|
Zhang Q, Li J, Sun Y, Song S, Li X, Chen G. Neoagarohexaose Protects against Amyloid β-Induced Oxidative Stress and Aggregation. APPL BIOCHEM MICRO+ 2022. [DOI: 10.1134/s0003683822100179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
|
29
|
Recent Progress in Research on Mechanisms of Action of Natural Products against Alzheimer's Disease: Dietary Plant Polyphenols. Int J Mol Sci 2022; 23:ijms232213886. [PMID: 36430365 PMCID: PMC9695301 DOI: 10.3390/ijms232213886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable degenerative disease of the central nervous system and the most common type of dementia in the elderly. Despite years of extensive research efforts, our understanding of the etiology and pathogenesis of AD is still highly limited. Nevertheless, several hypotheses related to risk factors for AD have been proposed. Moreover, plant-derived dietary polyphenols were also shown to exert protective effects against neurodegenerative diseases such as AD. In this review, we summarize the regulatory effects of the most well-known plant-derived dietary polyphenols on several AD-related molecular mechanisms, such as amelioration of oxidative stress injury, inhibition of aberrant glial cell activation to alleviate neuroinflammation, inhibition of the generation and promotion of the clearance of toxic amyloid-β (Aβ) plaques, inhibition of cholinesterase enzyme activity, and increase in acetylcholine levels in the brain. We also discuss the issue of bioavailability and the potential for improvement in this regard. This review is expected to encourage further research on the role of natural dietary plant polyphenols in the treatment of AD.
Collapse
|
30
|
Dong Y, Li T, Ma Z, Zhou C, Wang X, Li J. HSPA1A, HSPA2, and HSPA8 Are Potential Molecular Biomarkers for Prognosis among HSP70 Family in Alzheimer's Disease. DISEASE MARKERS 2022; 2022:9480398. [PMID: 36246562 PMCID: PMC9553556 DOI: 10.1155/2022/9480398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/24/2022] [Accepted: 09/07/2022] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease, which leads to impairment of cognition and memory. The heat shock protein 70 (HSP70) family plays an important role in the pathogenesis of AD. It is known to regulate protein misfolding in a variety of diseases, including inhibition of Aβ aggregation and NFT formation in AD. As yet, the diagnostic molecular markers of AD remain unclear. Herein, we sought to investigate molecular markers of HSP70 family that can affect diagnosis and treatment in AD through computational analysis. In this study, the intersection between HSP70 family members and immune molecules was taken to screen immune-related HSP70 family genes. Based on the datasets from the NCBI-Gene Expression Omnibus (GEO) database, we found that the expression levels of HSPA1A and HSPA2 were significantly increased in AD samples, while HSPA8 significantly decreased. Surprisingly, the combination of the 3 hub genes had a good diagnosis of AD via receiver operating characteristic curve (ROC). Moreover, the clinical value of the 3 hub genes was further assessed by the Spearman correlation analysis with AD-related genes, β-secretase activity, and γ-secretase activity. In terms of immune cell infiltration, we showed that the distribution of seven immune cell types (macrophages M2, neutrophils, T cells CD4 memory activated, macrophages M0, NK cells activated, plasma cells, and T cells follicular helper) was associated with the occurrence of AD by CIBERSORT. Furthermore, our data suggested that EP300, MYC, TP53, JUN, CREBBP, and ESR1 might be key transcription factors (TFs) for the 3 hub genes. In general, these findings suggest that HSPA1A, HSPA2, and HSPA8 are potential molecular biomarkers for prognosis among HSP70 family in AD, and it provides a new perspective on diagnostic and therapeutic targets for AD.
Collapse
Affiliation(s)
- Yeqing Dong
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Tongxin Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Zhonghui Ma
- Department of Laboratory Medicine, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Chi Zhou
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Xinxu Wang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| |
Collapse
|
31
|
Dehabadi MH, Caflisch A, Ilie IM, Firouzi R. Interactions of Curcumin's Degradation Products with the Aβ 42 Dimer: A Computational Study. J Phys Chem B 2022; 126:7627-7637. [PMID: 36148988 DOI: 10.1021/acs.jpcb.2c05846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Amyloid-β (Aβ) dimers are the smallest toxic species along the amyloid-aggregation pathway and among the most populated oligomeric accumulations present in the brain affected by Alzheimer's disease (AD). A proposed therapeutic strategy to avoid the aggregation of Aβ into higher-order structures is to develop molecules that inhibit the early stages of aggregation, i.e., dimerization. Under physiological conditions, the Aβ dimer is highly dynamic and does not attain a single well-defined structure but is rather characterized by an ensemble of conformations. In a recent study, a highly heterogeneous library of conformers of the Aβ dimer was generated by an efficient sampling method with constraints based on ion mobility mass spectrometry data. Here, we make use of the Aβ dimer library to study the interaction with two curcumin degradation products, ferulic aldehyde and vanillin, by molecular dynamics (MD) simulations. Ensemble docking and MD simulations are used to provide atomistic detail of the interactions between the curcumin degradation products and the Aβ dimer. The simulations show that the aromatic residues of Aβ, and in particular 19FF20, interact with ferulic aldehyde and vanillin through π-π stacking. The binding of these small molecules induces significant changes on the 16KLVFF20 region.
Collapse
Affiliation(s)
- Maryam Haji Dehabadi
- Department of Physical Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Pajohesh Boulevard, 1496813151 Tehran, Iran
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Ioana M Ilie
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Rohoullah Firouzi
- Department of Physical Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Pajohesh Boulevard, 1496813151 Tehran, Iran
| |
Collapse
|
32
|
Campane LZ, Nucci MP, Nishiyama M, Von Zuben M, Amaro E, da Luz PL. Long term effects of red wine consumption in brain: an MRI, fMRI and neuropsychological evaluation study. Nutr Neurosci 2022:1-12. [PMID: 35943074 DOI: 10.1080/1028415x.2022.2108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Red wine (RW) consumption has been proposed to have a potential health benefit. However, the effect of RW consumption on the brain is not entirely known, mainly when associated with aging. Regular red wine consumers (n = 30) and abstainers (ABST; n = 27) without cognitive impairment were evaluated for brain structural characteristics (Fazekas score and voxel-based morphometry) and for functional adaptations assessed by fMRI (using the Word Tasks Color Stroop (WCST) and Two-Back (TBT)), as well as by neuropsychological tests in different domains. There were no significant differences regarding brain morphological features. RW consumers showed greater activation in the thalamus during WCST and in paracingulate/anterior cingulate cortices, left superior frontal gyrus and frontal pole during TBT. ABST required higher activation of different cortical areas in the left parietal lobe during WCST. Age and intelligence quotient influenced those activations. In Stroop and trail-making neuropsychological tests, RW consumers performed slightly better than ABST. This study should be viewed as hypothesis-generating rather than conclusive.
Collapse
Affiliation(s)
- Lucas Zoppi Campane
- LIM-44 (NIF - Neuroimagem Funcional), Departamento de Radiologia, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Mariana Penteado Nucci
- LIM-44 (NIF - Neuroimagem Funcional), Departamento de Radiologia, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Marcelo Nishiyama
- Instituto de Cardiologia (InCor), Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Marina Von Zuben
- Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Edson Amaro
- LIM-44 (NIF - Neuroimagem Funcional), Departamento de Radiologia, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Protasio Lemos da Luz
- Instituto de Cardiologia (InCor), Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
33
|
Nguyen PH, Derreumaux P. Molecular Dynamics Simulations of the Tau Amyloid Fibril Core Dimer at the Surface of a Lipid Bilayer Model: I. In Alzheimer's Disease. J Phys Chem B 2022; 126:4849-4856. [PMID: 35759677 DOI: 10.1021/acs.jpcb.2c02836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A tau R3-R4 domain spanning residues 306-378 was shown to form an amyloid fibril core of a full-length tau in the brain of patients with Alzheimer's disease. Recently, we studied the dynamics of a tau R3-R4 monomer at the surface of a lipid bilayer model and revealed deep insertion of the amino acids spanning the PHF6 motif (residues 306-311) and its flanking residues. Here, we explore the membrane-associated conformational ensemble of a tau R3-R4 dimer by means of atomistic molecular dynamics. Similar to the monomer simulation, the R3-R4 dimer has the propensity to form β-hairpin-like conformation. Unlike the monomer, the dimer shows insertion of the C-terminal R4 region and transient adsorption of the PHF6 motif. Taken together, these results reveal the multiplicity of adsorption and insertion modes of tau into membranes depending on its oligomer size.
Collapse
Affiliation(s)
- Phuong H Nguyen
- CNRS, Université Paris Cité, UPR 9080, Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - Philippe Derreumaux
- CNRS, Université Paris Cité, UPR 9080, Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, 13 rue Pierre et Marie Curie, 75005 Paris, France.,Institut Universitaire de France (IUF), 75005 Paris, France
| |
Collapse
|
34
|
Ferdosh S, Banerjee S, Singh J, Barat C. Amyloid protein-induced sequestration of the eukaryotic ribosome: effect of stoichiometry and polyphenolic inhibitors. FEBS Lett 2022; 596:1190-1202. [PMID: 35114013 DOI: 10.1002/1873-3468.14308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/12/2022] [Accepted: 01/22/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease (AD) is characterized by the appearance of neurofibrillary tangles comprising of the Tau protein and aggregation of amyloid-β peptides (Aβ 1-40 and Aβ 1-42). A concomitant loss of the ribosomal population is also observed in AD-affected neurons. Our studies demonstrate that, similarly to Tau protein aggregation, in vitro aggregation of Aβ peptides in the vicinity of the yeast 80S ribosome can induce co-aggregation of ribosomal components. The RNA-stimulated aggregation of Aβ peptides and the Tau-K18 variant is dependent on the RNA:protein stoichiometric ratio. A similar effect of stoichiometry is also observed on the ribosome-protein co-aggregation process. Polyphenolic inhibitors of amyloid aggregation, such as rosmarinic acid and myricetin, inhibit RNA-stimulated Aβ and Tau-K18 aggregation and can mitigate the co-aggregation of ribosomal components.
Collapse
Affiliation(s)
- Sehnaz Ferdosh
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Senjuti Banerjee
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Jayshree Singh
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Chandana Barat
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| |
Collapse
|
35
|
Bie N, Li J, Li C, Lian R, Qin L, Wang C. Protective effect and mechanism of docosahexaenoic acid on the cognitive function in female APP/PS1 mice. Food Funct 2021; 12:11435-11448. [PMID: 34676845 DOI: 10.1039/d1fo01922h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Docosahexaenoic acid (DHA) has been studied for many years owing to its protective effect on the decline in brain function. DHA intake reduces the risk of Alzheimer's disease (AD) and decreases amyloid deposition; however, the underlying molecular mechanism has not been completed elucidated. In this study, the effect of DHA on the cognitive function of amyloid precursor protein (APP)/PS1 in wild-type mice and its related mechanism were investigated. Results from the Morris water maze test showed that DHA improved learning and memory function in mice. Moreover, DHA reduced neuronal damage in mice brains, as determined using Nissl staining. Unsaturated fatty acid levels in the brain of mice increased (p < 0.01) after DHA administration and saturated fatty acid levels decreased (p < 0.01). The deposition of amyloid-beta (Aβ) plaques and tau protein neurofibrillary tangles was significantly inhibited. The mechanism of action of DHA was attributed to the upregulation of the expression of β-secretase (BACE)2, which competed with BACE1 to cleave APP, thus decreasing the production of extracellular Aβ fragments (p < 0.01). The expression level of insulin-degrading enzyme was not significantly different. The expression of N-methyl-D-aspartate receptors was further downregulated and the phosphorylation of glycogen synthase kinase-3β and tau protein was inhibited (p < 0.01). These data indicated that DHA could protect cognitive function in mice by reducing Aβ plaque formation and decreasing tau phosphorylation levels.
Collapse
Affiliation(s)
- Nana Bie
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin Economy Technological Development Area, No. 29, 13th Avenue, Tianjin, 300457, People Republic of China.
| | - Jingyao Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin Economy Technological Development Area, No. 29, 13th Avenue, Tianjin, 300457, People Republic of China.
| | - Chenjing Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin Economy Technological Development Area, No. 29, 13th Avenue, Tianjin, 300457, People Republic of China.
| | - Rui Lian
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin Economy Technological Development Area, No. 29, 13th Avenue, Tianjin, 300457, People Republic of China.
| | - Liehao Qin
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin Economy Technological Development Area, No. 29, 13th Avenue, Tianjin, 300457, People Republic of China.
| | - Chunling Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin Economy Technological Development Area, No. 29, 13th Avenue, Tianjin, 300457, People Republic of China.
| |
Collapse
|
36
|
Wan J, Feng M, Pan W, Zheng X, Xie X, Hu B, Teng C, Wang Y, Liu Z, Wu J, Cai S. Inhibitory Effects of Six Types of Tea on Aging and High-Fat Diet-Related Amyloid Formation Activities. Antioxidants (Basel) 2021; 10:1513. [PMID: 34679648 PMCID: PMC8533055 DOI: 10.3390/antiox10101513] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 01/02/2023] Open
Abstract
Aging and lipid metabolism disorders promote the formation and accumulation of amyloid with β-sheet structure, closely related to cardiovascular disease, senile dementia, type 2 diabetes, and other senile degenerative diseases. In this study, five representative teas were selected from each of the six types of tea, and a total of 30 teas were selected to evaluate the inhibitory activities on the formation of aging-related amyloid in vitro. The results showed that the 30 teas had a significant inhibitory effect on the formation activity on aging-related amyloid at the protein level in vitro. Although the content of catechins is relatively low, black tea and dark tea still have significant antioxidant activity and inhibit the formation of amyloid. A high-fat diet established the model of lipid metabolism disorder in premature aging SAMP8 mice, and these mice were gavaged different tea water extracts. The results showed that different tea types have a significant inhibitory effect on the formation of β-amyloid and Aβ42 mediated by age-related lipid metabolism disorders, and the in vivo activity of fully fermented teas was better than that of green tea. The action mechanism was related to antioxidation, anti-inflammatory, and improving lipid metabolism.
Collapse
Affiliation(s)
- Juan Wan
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Meiyan Feng
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Wenjing Pan
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Xin Zheng
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Xinya Xie
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Baozhu Hu
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Cuiqin Teng
- Wuzhou Institute of Agricultural, Wuzhou 543003, China; (C.T.); (J.W.)
| | - Yingzi Wang
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Zhonghua Liu
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Jianhua Wu
- Wuzhou Institute of Agricultural, Wuzhou 543003, China; (C.T.); (J.W.)
| | - Shuxian Cai
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; (J.W.); (M.F.); (W.P.); (X.Z.); (X.X.); (B.H.); (Y.W.)
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
37
|
Zhu L, Gong Y, Lju H, Sun G, Zhang Q, Qian Z. Mechanisms of melatonin binding and destabilizing the protofilament and filament of tau R3-R4 domains revealed by molecular dynamics simulation. Phys Chem Chem Phys 2021; 23:20615-20626. [PMID: 34514491 DOI: 10.1039/d1cp03142b] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The accumulation of β-amyloid (Aβ) and tau protein is considered to be an important pathological characteristic of Alzheimer's disease (AD). Failure of medicine targeting Aβ has drawn more attention to the influence of tau protein and its fibrillization on neurodegeneration. Increasing evidence shows that melatonin (Mel) can effectively inhibit the formation of tau fibrils and disassemble preformed tau fibrils. However, the underlying mechanism is poorly understood. In this work, we investigated the kinetics of melatonin binding and destabilizing the tetrameric protofilament and octameric filament of tau R3-R4 domains by performing microsecond all-atom molecular dynamics simulations. Our results show that Mel is able to disrupt the C-shaped structure of the tau protofilament and filament, and destabilizes the association between N- and C-termini. Mel predominantly binds to β1 and β6-β8 regions and favors contact with the elongation surface, which is dominantly driven by hydrogen bonding interactions and facilitated by other interactions. The strong π-π stacking interaction of Mel with Y310 impedes the intramolecular CH-π interaction between I308 and Y310, and the cation-π interaction of Mel with R379 interferes with the formation of the D348-R379 salt bridge. Moreover, Mel occupies the protofilament surface in the tetrameric protofilament and prevents the formation of intermolecular hydrogen bonds between residues K331 and Q336 in the octameric filament. Our work provides molecular insights into Mel hindering tau fibrillization or destabilizing the protofilament and filament, and the revealed inhibitory mechanisms provide useful clues for the design of efficient anti-amyloid agents.
Collapse
Affiliation(s)
- Lili Zhu
- Key Laboratory of Exercise and Health Sciences (Ministry of Education) and School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China.
| | - Yehong Gong
- College of Physical Education and Training, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China
| | - Hao Lju
- Key Laboratory of Exercise and Health Sciences (Ministry of Education) and School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China.
| | - Gongwu Sun
- Key Laboratory of Exercise and Health Sciences (Ministry of Education) and School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China.
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China
| | - Zhenyu Qian
- Key Laboratory of Exercise and Health Sciences (Ministry of Education) and School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China.
| |
Collapse
|
38
|
Gour N, Gazit E. Metabolite assemblies: A surprising extension to the amyloid hypothesis. Curr Opin Chem Biol 2021; 64:154-164. [PMID: 34482124 DOI: 10.1016/j.cbpa.2021.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/10/2021] [Accepted: 07/25/2021] [Indexed: 12/15/2022]
Abstract
The realization of the ability of metabolites to form self-assembled amyloid-like nanostructures was a surprising phenomenon. This discovery paved the way towards understanding the pathophysiology of the inborn error of metabolism disorders from a new perspective, relating them to amyloid-associated diseases that are characterized by the aggregation of proteins and polypeptides. Hence, a 'generic amyloid hypothesis' can be proposed. This theory implies that the formation of amyloid-like structures is a general phenomenon not limited to proteins and reflects a common etiology for both age-related amyloid-associated diseases and inborn error of metabolism disorders. Here, we present a comprehensive survey of the recent research related to metabolite amyloids including their structure formation through self-association, propagation, interactions, transmission, and their role in metabolic disorders and neurodegenerative diseases and their applications for the fabrication of novel materials which implicate metabolite assemblies as a surprising extension to the amyloid scheme.
Collapse
Affiliation(s)
- Nidhi Gour
- School of Science, Department of Chemistry, Indrashil University, Mehsana, Gujarat, 382740 India
| | - Ehud Gazit
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, 6997801, Israel; BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
39
|
Zheng Q, Kebede MT, Lee B, Krasinski CA, Islam S, Wurfl LA, Kemeh MM, Ivancic VA, Jakobsche CE, Spratt DE, Lazo ND. Differential Effects of Polyphenols on Insulin Proteolysis by the Insulin-Degrading Enzyme. Antioxidants (Basel) 2021; 10:1342. [PMID: 34572974 PMCID: PMC8467823 DOI: 10.3390/antiox10091342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022] Open
Abstract
The insulin-degrading enzyme (IDE) possesses a strong ability to degrade insulin and Aβ42 that has been linked to the neurodegeneration in Alzheimer's disease (AD). Given this, an attractive IDE-centric strategy for the development of therapeutics for AD is to boost IDE's activity for the clearance of Aβ42 without offsetting insulin proteostasis. Recently, we showed that resveratrol enhances IDE's activity toward Aβ42. In this work, we used a combination of chromatographic and spectroscopic techniques to investigate the effects of resveratrol on IDE's activity toward insulin. For comparison, we also studied epigallocatechin-3-gallate (EGCG). Our results show that the two polyphenols affect the IDE-dependent degradation of insulin in different ways: EGCG inhibits IDE while resveratrol has no effect. These findings suggest that polyphenols provide a path for developing therapeutic strategies that can selectively target IDE substrate specificity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Noel D. Lazo
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, MA 01610, USA; (Q.Z.); (M.T.K.); (B.L.); (C.A.K.); (S.I.); (L.A.W.); (M.M.K.); (V.A.I.); (C.E.J.); (D.E.S.)
| |
Collapse
|
40
|
Zhao H, Huang X, Tong Z. Formaldehyde-Crosslinked Nontoxic Aβ Monomers to Form Toxic Aβ Dimers and Aggregates: Pathogenicity and Therapeutic Perspectives. ChemMedChem 2021; 16:3376-3390. [PMID: 34396700 DOI: 10.1002/cmdc.202100428] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/14/2021] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is characterized by the presence of senile plaques in the brain. However, medicines targeting amyloid-beta (Aβ) have not achieved the expected clinical effects. This review focuses on the formation mechanism of the Aβ dimer (the basic unit of oligomers and fibrils) and its tremendous potential as a drug target. Recently, age-associated formaldehyde and Aβ-derived formaldehyde have been found to crosslink the nontoxic Aβ monomer to form the toxic dimers, oligomers and fibrils. Particularly, Aβ-induced formaldehyde accumulation and formaldehyde-promoted Aβ aggregation form a vicious cycle. Subsequently, formaldehyde initiates Aβ toxicity in both the early-and late-onset AD. These facts also explain why AD drugs targeting only Aβ do not have the desired therapeutic effects. Development of the nanoparticle-based medicines targeting both formaldehyde and Aβ dimer is a promising strategy for improving the drug efficacy by penetrating blood-brain barrier and extracellular space into the cortical neurons in AD patients.
Collapse
Affiliation(s)
- Hang Zhao
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xuerong Huang
- Wenzhou Medical University Affiliated Hospital 3, Department of Neurology, Wenzhou, 325200, China
| | - Zhiqian Tong
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
41
|
Ochiishi T, Kaku M, Kajsongkram T, Thisayakorn K. Mulberry fruit extract alleviates the intracellular amyloid-β oligomer-induced cognitive disturbance and oxidative stress in Alzheimer's disease model mice. Genes Cells 2021; 26:861-873. [PMID: 34387016 DOI: 10.1111/gtc.12889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/30/2022]
Abstract
Intracellular amyloid-β (Aβ) oligomers are key therapeutic targets because they are strongly cytotoxic and play crucial roles in the cognitive function in Alzheimer's disease (AD). Anthocyanins, polyphenolic flavonoids with antioxidant and neuroprotective properties, are potential therapeutic candidates for AD. Here, we investigated the effects of anthocyanin-enriched extracts from fruits of mulberry (Morus alba Linn.) in Thailand against the neurotoxicity of Aβ oligomers. Using the monitoring system for Aβ aggregation, we showed that the extract induced the dissociation of Aβ in cultured HEK293T cells. To investigate the effects on cognitive function, we orally administered the extract to Aβ-GFP transgenic mice (Aβ-GFP Tg), a mouse model that expresses Aβ oligomers inside neurons, and performed the novel object recognition test and passive avoidance test. Aβ-GFP Tg usually showed deficits in novel object recognition memory and reference memory compared with non-Tg, but administration of the extract improved both compared with vehicle-treated Aβ-GFP Tg. Aβ-GFP Tg exhibited lower superoxide dismutase (SOD) activity than non-Tg. However, after the administration of the extract, the SOD activity was restored. These results suggest that Thai mulberry fruit extract ameliorates cytotoxicity induced by the intracellular Aβ oligomers and may be an effective therapeutic or preventive candidate for AD.
Collapse
Affiliation(s)
- Tomoyo Ochiishi
- Molecular Neurobiology Research Group, Biomedical Research Institute (BMRI), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Masami Kaku
- Faculty of Health and Science, Uekusa Gakuen University, Chiba, Japan
| | - Tanwarat Kajsongkram
- Expert Centre of Innovative Herbal Products (InnoHerb), Thailand Institute of Scientific and Technological Research (TISTR), Khlong Luang, Thailand
| | - Krittiya Thisayakorn
- Expert Centre of Innovative Herbal Products (InnoHerb), Thailand Institute of Scientific and Technological Research (TISTR), Khlong Luang, Thailand
| |
Collapse
|
42
|
Diabetes and Alzheimer's Disease: Might Mitochondrial Dysfunction Help Deciphering the Common Path? Antioxidants (Basel) 2021; 10:antiox10081257. [PMID: 34439505 PMCID: PMC8389322 DOI: 10.3390/antiox10081257] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
A growing number of clinical and epidemiological studies support the hypothesis of a tight correlation between type 2 diabetes mellitus (T2DM) and the development risk of Alzheimer's disease (AD). Indeed, the proposed definition of Alzheimer's disease as type 3 diabetes (T3D) underlines the key role played by deranged insulin signaling to accumulation of aggregated amyloid beta (Aβ) peptides in the senile plaques of the brain. Metabolic disturbances such as hyperglycemia, peripheral hyperinsulinemia, dysregulated lipid metabolism, and chronic inflammation associated with T2DM are responsible for an inefficient transport of insulin to the brain, producing a neuronal insulin resistance that triggers an enhanced production and deposition of Aβ and concomitantly contributes to impairment in the micro-tubule-associated protein Tau, leading to neural degeneration and cognitive decline. Furthermore, the reduced antioxidant capacity observed in T2DM patients, together with the impairment of cerebral glucose metabolism and the decreased performance of mitochondrial activity, suggests the existence of a relationship between oxidative damage, mitochondrial impairment, and cognitive dysfunction that could further reinforce the common pathophysiology of T2DM and AD. In this review, we discuss the molecular mechanisms by which insulin-signaling dysregulation in T2DM can contribute to the pathogenesis and progression of AD, deepening the analysis of complex mechanisms involved in reactive oxygen species (ROS) production under oxidative stress and their possible influence in AD and T2DM. In addition, the role of current therapies as tools for prevention or treatment of damage induced by oxidative stress in T2DM and AD will be debated.
Collapse
|
43
|
Salahuddin P, Khan RH, Furkan M, Uversky VN, Islam Z, Fatima MT. Mechanisms of amyloid proteins aggregation and their inhibition by antibodies, small molecule inhibitors, nano-particles and nano-bodies. Int J Biol Macromol 2021; 186:580-590. [PMID: 34271045 DOI: 10.1016/j.ijbiomac.2021.07.056] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
Protein misfolding and aggregation can be induced by a wide variety of factors, such as dominant disease-associated mutations, changes in the environmental conditions (pH, temperature, ionic strength, protein concentration, exposure to transition metal ions, exposure to toxins, posttranslational modifications including glycation, phosphorylation, and sulfation). Misfolded intermediates interact with similar intermediates and progressively form dimers, oligomers, protofibrils, and fibrils. In amyloidoses, fibrillar aggregates are deposited in the tissues either as intracellular inclusion or extracellular plaques (amyloid). When such proteinaceous deposit occurs in the neuronal cells, it initiates degeneration of neurons and consequently resulting in the manifestation of various neurodegenerative diseases. Several different types of molecules have been designed and tested both in vitro and in vivo to evaluate their anti-amyloidogenic efficacies. For instance, the native structure of a protein associated with amyloidosis could be stabilized by ligands, antibodies could be used to remove plaques, oligomer-specific antibody A11 could be used to remove oligomers, or prefibrillar aggregates could be removed by affibodies. Keeping the above views in mind, in this review we have discussed protein misfolding and aggregation, mechanisms of protein aggregation, factors responsible for aggregations, and strategies for aggregation inhibition.
Collapse
Affiliation(s)
- Parveen Salahuddin
- DISC, Interdisciplinary Biotechnology Unit, A.M.U., Aligarh 202002, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, A.M.U., Aligarh 202002, India.
| | - Mohammad Furkan
- Interdisciplinary Biotechnology Unit, A.M.U., Aligarh 202002, India
| | - Vladimir N Uversky
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, Moscow region 142290, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zeyaul Islam
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, P.O Box 5825, Doha, Qatar
| | - Munazza Tamkeen Fatima
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
44
|
Nan S, Wang P, Zhang Y, Fan J. Epigallocatechin-3-Gallate Provides Protection Against Alzheimer's Disease-Induced Learning and Memory Impairments in Rats. Drug Des Devel Ther 2021; 15:2013-2024. [PMID: 34012254 PMCID: PMC8128347 DOI: 10.2147/dddt.s289473] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
Purpose Recent evidence has highlighted the anti-inflammatory properties of the constituent of Green Tea Polyphenols (GTP), epigallocatechin-3-gallate (EGCG) which has been suggested to exert a neuroprotective effect on Alzheimer’s disease (AD). The current study aimed to elucidate the effect of EGCG on memory function in rats with AD. Methods AD rat models were initially established through an injection with Aβ 25–35 solution, followed by gavage with EGCG at varying doses to determine the effect of EGCG on learning and cognitive deficits in AD. Morris water maze test was conducted to evaluate the spatial memory function of the rats. Immunohistochemistry and Western blot analysis were performed to identify Tau phosphorylation. The expression of β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) mRNA and protein in rat hippocampus was measured by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Acetylcholinesterase (AchE) activity, Aβ1-42 expression and Ach content were all detected using enzyme-linked immunosorbent assay (ELISA). Results EGCG intervention brought about a decrease in the escape latency period while increasing the time at the target quadrant among the AD rats. EGCG decreased the hyperphosphorylation of Tau in hippocampus. BACE1 expression and activity as well as the expression of Aβ1-42 were suppressed by EGCG. Moreover, EGCG promoted Ach content by diminishing the activity of AchE. Conclusion The current study demonstrates that EGCG may diminish the hyperphosphorylation of the Tau protein, downregulate BACE1 and Aβ1-42 expression to improve the antioxidant system and learning and memory function of rats with AD.
Collapse
Affiliation(s)
- Shanji Nan
- Department of Neurology, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Peng Wang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Yizhi Zhang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Jia Fan
- Department of Neurology, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| |
Collapse
|
45
|
Exploratory dietary patterns and cognitive function in the "Seguimiento Universidad de Navarra" (SUN) Prospective Cohort. Eur J Clin Nutr 2021; 76:48-55. [PMID: 33986490 DOI: 10.1038/s41430-021-00922-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Dementia is projected to affect 135 million by 2050. Diet is a pertinent target for primary prevention, but firm recommendations for dementia prevention are not available yet. Our aim was to address the association between exploratory (empirically derived) dietary patterns (DP) and changes in the Spanish Telephone Interview for Cognitive Status (STICS-m, maximum score = 41 points) over 6 years. METHOD Information on diet was collected with a validated 136-item food-frequency questionnaire from 803 participants in the Mediterranean cohort "Seguimiento Universidad de Navarra." We used principal component analysis to derive exploratory DP. The derived DP were associated with change in STICS-m scores over 6 years, through adjusted multiple linear regression models. RESULTS Two main DP were identified. The first DP resembled a Western dietary pattern (WDP)-high in sugar, fat, processed foods, and red meat-and the second DP resembled a Mediterranean dietary pattern (MDP)-high in vegetables, fruits, nuts, fish, and olive oil. Adherence to the WDP (tertile 3 vs tertile 1) was significantly associated with negative STICS-m changes after 6 years (between-tertile difference in changes: -0.80 points; 95% confidence interval [CI] -1.51, -0.08, p value = 0.03). Meanwhile, the MDP showed a positive +0.71 point (95% CI 0.15, 1.26, p value = 0.01) between-tertile difference in changes in the STICS-m score. CONCLUSIONS A healthy, prudent, MDP was associated with less decline in cognitive function and, thus, could help to lower dementia incidence. Western-type diets were associated with a greater decline in cognitive performance and could increase dementia incidence.
Collapse
|
46
|
Yuan X, Wang Z, Zhang L, Sui R, Khan S. Exploring the inhibitory effects of liquiritigenin against tau fibrillation and related neurotoxicity as a model of preventive care in Alzheimer's disease. Int J Biol Macromol 2021; 183:1184-1190. [PMID: 33965487 DOI: 10.1016/j.ijbiomac.2021.05.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/18/2021] [Accepted: 05/03/2021] [Indexed: 01/12/2023]
Abstract
Aggregation of tau protein into the form of insoluble amyloid fibrils is linked with Alzheimer's disease. The identification of potential small molecules that can inhibit tau protein from undergoing aggregation has received a great deal of interest, recently. In the present study, the possible inhibitory effects of liquiritigenin as a member of chiral flavanone family on tau amyloid fibrils formation and their resulting neurotoxicity were assessed by different biophysical and cellular assays. The inhibitory effect of the liquiritigenin against tau amyloid formation was investigated using thioflavin T (ThT) and 1-Anilino-8-naphthalene sulfonate (ANS) fluorescence spectroscopy, Congo red (CR) binding assays, transmission electron microscopy (TEM) analysis, and circular dichroism (CD) spectroscopy. Neurotoxicity assays were also performed against neuron-like cells (SH-SY5Y) using 3-(4,5-Dimethylthiazol)-2,5-diphenyltetrazolium bromide (MTT) reduction, reactive oxygen species (ROS), catalase (CAT) and caspase-3 activity measurements. We found that liquiritigenin served as an efficient inhibitor of tau amyloid fibrils formation through prevention of structural transition in tau structure, exposure of hydrophobic patches and their associated neurotoxicity mediated by decrease in the production of ROS and caspase-3 activity and elevation of CAT activity. These data may finally find applications in the development of promising inhibitors against amyloid fibril formation and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Xueling Yuan
- Department of Neurology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121099, China
| | - Zhuo Wang
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Lei Zhang
- School of Nursing, Jinzhou Medical University, Jinzhou 121099, China
| | - Rubo Sui
- Department of Neurology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121099, China.
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
47
|
Gaudreault R, Hervé V, van de Ven TGM, Mousseau N, Ramassamy C. Polyphenol-Peptide Interactions in Mitigation of Alzheimer's Disease: Role of Biosurface-Induced Aggregation. J Alzheimers Dis 2021; 81:33-55. [PMID: 33749653 DOI: 10.3233/jad-201549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder, responsible for nearly two-thirds of all dementia cases. In this review, we report the potential AD treatment strategies focusing on natural polyphenol molecules (green chemistry) and more specifically on the inhibition of polyphenol-induced amyloid aggregation/disaggregation pathways: in bulk and on biosurfaces. We discuss how these pathways can potentially alter the structure at the early stages of AD, hence delaying the aggregation of amyloid-β (Aβ) and tau. We also discuss multidisciplinary approaches, combining experimental and modelling methods, that can better characterize the biochemical and biophysical interactions between proteins and phenolic ligands. In addition to the surface-induced aggregation, which can occur on surfaces where protein can interact with other proteins and polyphenols, we suggest a new concept referred as "confinement stability". Here, on the contrary, the adsorption of Aβ and tau on biosurfaces other than Aβ- and tau-fibrils, e.g., red blood cells, can lead to confinement stability that minimizes the aggregation of Aβ and tau. Overall, these mechanisms may participate directly or indirectly in mitigating neurodegenerative diseases, by preventing protein self-association, slowing down the aggregation processes, and delaying the progression of AD.
Collapse
Affiliation(s)
- Roger Gaudreault
- Department of Physics, Université de Montréal, Montreal, QC, Canada
| | - Vincent Hervé
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | | | - Normand Mousseau
- Department of Physics, Université de Montréal, Montreal, QC, Canada
| | | |
Collapse
|
48
|
Caruana M, Camilleri A, Farrugia MY, Ghio S, Jakubíčková M, Cauchi RJ, Vassallo N. Extract from the Marine Seaweed Padina pavonica Protects Mitochondrial Biomembranes from Damage by Amyloidogenic Peptides. Molecules 2021; 26:1444. [PMID: 33799979 PMCID: PMC7962105 DOI: 10.3390/molecules26051444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
The identification of compounds which protect the double-membrane of mitochondrial organelles from disruption by toxic confomers of amyloid proteins may offer a therapeutic strategy to combat human neurodegenerative diseases. Here, we exploited an extract from the marine brown seaweed Padina pavonica (PPE) as a vital source of natural bioactive compounds to protect mitochondrial membranes against insult by oligomeric aggregates of the amyloidogenic proteins amyloid-β (Aβ), α-synuclein (α-syn) and tau, which are currently considered to be major targets for drug discovery in Alzheimer's disease (AD) and Parkinson's disease (PD). We show that PPE manifested a significant inhibitory effect against swelling of isolated mitochondria exposed to the amyloid oligomers, and attenuated the release of cytochrome c from the mitochondria. Using cardiolipin-enriched synthetic lipid membranes, we also show that dye leakage from fluorophore-loaded vesicles and formation of channel-like pores in planar bilayer membranes are largely prevented by incubating the oligomeric aggregates with PPE. Lastly, we demonstrate that PPE curtails the ability of Aβ42 and α-syn monomers to self-assemble into larger β-aggregate structures, as well as potently disrupts their respective amyloid fibrils. In conclusion, the mito-protective and anti-aggregator biological activities of Padina pavonica extract may be of therapeutic value in neurodegenerative proteinopathies, such as AD and PD.
Collapse
Affiliation(s)
- Mario Caruana
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, 2023 Msida, Malta
| | - Angelique Camilleri
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, 2023 Msida, Malta
| | - Maria Ylenia Farrugia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, 2023 Msida, Malta
| | - Stephanie Ghio
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, 2023 Msida, Malta
| | - Michaela Jakubíčková
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Department of Experimental Biology, Faculty of Science, Masaryk University, 60300 Brno, Czech Republic
| | - Ruben J. Cauchi
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, 2023 Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta; (M.C.); (A.C.); (M.Y.F.); (S.G.); (M.J.); (R.J.C.)
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, 2023 Msida, Malta
| |
Collapse
|
49
|
Paul A, Viswanathan GK, Huber A, Arad E, Engel H, Jelinek R, Gazit E, Segal D. Inhibition of tau amyloid formation and disruption of its preformed fibrils by Naphthoquinone-Dopamine hybrid. FEBS J 2021; 288:4267-4290. [PMID: 33523571 DOI: 10.1111/febs.15741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/02/2021] [Accepted: 01/28/2021] [Indexed: 01/10/2023]
Abstract
Misfolding and aggregation of tau protein, into pathological amyloids, are hallmarks of a group of neurodegenerative diseases collectively termed tauopathies and their modulation may be therapeutically valuable. Herein, we describe the synthesis and characterization of a dopamine-based hybrid molecule, naphthoquinone-dopamine (NQDA). Using thioflavin S assay, CD, transmission electron microscopy, dynamic light scattering, Congo Red birefringence, and large unilamellar vesicle leakage assays, we demonstrated its efficacy in inhibiting the in vitro aggregation of key tau-derived amyloidogenic fragments, PHF6 (VQIVYK) and PHF6* (VQIINK), prime drivers of aggregation of full-length tau in disease pathology. Isothermal titration calorimetry analysis revealed that the interaction between NQDA and PHF6 is spontaneous and has significant binding efficiency driven by both entropic and enthalpic processes. Furthermore, NQDA efficiently disassembled preformed fibrils of PHF6 and PHF6* into nontoxic species. Molecular dynamic simulations supported the in vitro results and provided a plausible mode of binding of NQDA with PHF6 fibril. NQDA was also capable of inhibiting the aggregation of full-length tau protein and disrupting its preformed fibrils in vitro in a dose-dependent manner. In a comparative study, the IC50 value (50% inhibition of fibril formation) of NQDA in inhibiting the aggregation of PHF6 (25 µm) was ~ 17 µm, which is lower than for other bona fide amyloid inhibitors, naphthoquinone-tryptophan, rosmarinic acid, epigallocatechin gallate, ~ 21, ~ 77, or ~ 19 µm, respectively. Comparable superiority of NQDA was observed for inhibition of PHF6*. These findings suggest that NQDA can be a useful scaffold for designing new therapeutics for Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Ashim Paul
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel
| | - Guru KrishnaKumar Viswanathan
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel
| | - Adi Huber
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel
| | - Elad Arad
- Ilse Katz Institute for Nanoscale Science and Technology & Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Hamutal Engel
- Blavatnik Center for Drug Discovery, Tel Aviv University, Israel
| | - Raz Jelinek
- Ilse Katz Institute for Nanoscale Science and Technology & Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel.,Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Israel
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel.,Sagol Interdisciplinary School of Neuroscience, Tel Aviv University, Israel
| |
Collapse
|
50
|
Abstract
Herbal Teas prepared from leaves, roots, fruits, and flowers of different herbs contain
many useful nutrients that may be a good replacement for medicating certain diseases. These herbal
teas are very rich in poly-phenols, therefore are significant for their antioxidant, anti-inflammation,
anticancer, anticardiovascular, antimicrobial, antihyperglycemic, and antiobesity properties. Medical
chronic conditions, such as cardiovascular diseases, cancer, Alzheimer’s disease, Parkinson’s disease,
constipation, diabetes, and bed wetting in children can be easily cured by the use of these herbal
teas in regular and moderate amounts. This review focuses on the diverse constituents of herbal teas
due to which these can be an attractive alternative towards promoting human health.
Collapse
Affiliation(s)
- Tabinda Sattar
- Department of Chemistry, ICS, Bahauddin Zakariya University, Multan, Pakistan
| |
Collapse
|