1
|
Pyka P, Garbo S, Fioravanti R, Jacob C, Hittinger M, Handzlik J, Zwergel C, Battistelli C. Selenium-containing compounds: a new hope for innovative treatments in Alzheimer's disease and Parkinson's disease. Drug Discov Today 2024; 29:104062. [PMID: 38871111 DOI: 10.1016/j.drudis.2024.104062] [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: 01/31/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Neurodegenerative diseases are challenging to cure. To date, no cure has been found for Alzheimer's disease or Parkinson's disease, and current treatments are able only to slow the progression of the diseases and manage their symptoms. After an introduction to the complex biology of these diseases, we discuss the beneficial effect of selenium-containing agents, which show neuroprotective effects in vitro or in vivo. Indeed, selenium is an essential trace element that is being incorporated into innovative organoselenium compounds, which can improve outcomes in rodent or even primate models with neurological deficits. Herein, we critically discuss recent findings in the field of selenium-based applications in neurological disorders.
Collapse
Affiliation(s)
- Patryk Pyka
- Department of Technology and Biotechnology of Drugs, Jagiellonian University, Medical College, Medyczna 9, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, św. Łazarza 15, 31-530 Krakow, Poland; Istituto Pasteur Italia, Fondazione Cenci-Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Sabrina Garbo
- Istituto Pasteur Italia, Fondazione Cenci-Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Rossella Fioravanti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Claus Jacob
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, Campus B 2.1, D-66123 Saarbrücken, Germany
| | - Marius Hittinger
- Pharmbiotec gGmbH, Department of Drug Discovery, Nußkopf 39, 66578 Schiffweiler, Germany
| | - Jadwiga Handzlik
- Department of Technology and Biotechnology of Drugs, Jagiellonian University, Medical College, Medyczna 9, 30-688 Krakow, Poland.
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, Campus B 2.1, D-66123 Saarbrücken, Germany; Pharmbiotec gGmbH, Department of Drug Discovery, Nußkopf 39, 66578 Schiffweiler, Germany.
| | - Cecilia Battistelli
- Istituto Pasteur Italia, Fondazione Cenci-Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
2
|
Hou J, Wang X, Zhang J, Shen Z, Li X, Yang Y. Chuanxiong Renshen Decoction Inhibits Alzheimer's Disease Neuroinflammation by Regulating PPARγ/NF-κB Pathway. Drug Des Devel Ther 2024; 18:3209-3232. [PMID: 39071817 PMCID: PMC11283787 DOI: 10.2147/dddt.s462266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
Background and Aim Previous studies of our research group have shown that Chuanxiong Renshen Decoction (CRD) has the effect of treating AD, but the exact mechanism of its effect is still not clarified. The aim of this study was to investigate the effect and mechanism of CRD on AD neuroinflammation. Materials and Methods Morris Water Maze (MWM) tests were employed to assess the memory and learning capacity of AD mice. HE and Nissl staining were used to observe the neural cells of mice. The expression of Iba-1 and CD86 were detected by immunohistochemical staining. Utilize UHPLC-MS/MS metabolomics techniques and the KEGG to analyze the metabolic pathways of CRD against AD. Lipopolysaccharide (LPS) induced BV2 microglia cells to construct a neuroinflammatory model. The expression of Iba-1 and CD86 were detected by immunofluorescence and flow cytometry. The contents of TNF-α and IL-1β were detected by ELISA. Western blot assay was used to detect the expression of PPARγ, p-NF-κB p65, NF-κB p65 proteins and inflammatory cytokines iNOS and COX-2 in PPARγ/NF-κB pathway with and without PPARγ inhibitor GW9662. Results CRD ameliorated the learning and memory ability of 3×Tg-AD mice, repaired the damaged nerve cells in the hippocampus, reduced the area of Iba-1 and CD86 positive areas in both the hippocampus and cortex regions, as well as attenuated serum levels of IL-1β and TNF-α in mice. CRD-containing serum significantly decreased the expression level of Iba-1, significantly reduced the levels of TNF-α and IL-1β, significantly increased the protein expression of PPARγ, and significantly decreased the proteins expression of iNOS, COX-2 and p-NF-κB p65 in BV2 microglia cells. After addition of PPARγ inhibitor GW9662, the inhibitory effect of CRD-containing serum on NF-κB activation was significantly weakened. Conclusion CRD can activate PPARγ, regulating PPARγ/NF-κB signaling pathway, inhibiting microglia over-activation and reducing AD neuroinflammation.
Collapse
Affiliation(s)
- Jinling Hou
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Xiaoyan Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Jian Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Zhuojun Shen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Xiang Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Yuanxiao Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, People’s Republic of China
| |
Collapse
|
3
|
Skalny AV, Aschner M, Santamaria A, Filippini T, Gritsenko VA, Tizabi Y, Zhang F, Guo X, Rocha JBT, Tinkov AA. The Role of Gut Microbiota in the Neuroprotective Effects of Selenium in Alzheimer's Disease. Mol Neurobiol 2024:10.1007/s12035-024-04343-w. [PMID: 39012446 DOI: 10.1007/s12035-024-04343-w] [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: 03/06/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
The objective of the present review was to provide a timely update on the molecular mechanisms underlying the beneficial role of Se in Alzheimer's disease pathogenesis, and discuss the potential role of gut microbiota modulation in this neuroprotective effect. The existing data demonstrate that selenoproteins P, M, S, R, as well as glutathione peroxidases and thioredoxin reductases are involved in regulation of Aβ formation and aggregation, tau phosphorylation and neurofibrillary tangles formation, as well as mitigate the neurotoxic effects of Aβ and phospho-tau. Correspondingly, supplementation with various forms of Se in cellular and animal models of AD was shown to reduce Aβ formation, tau phosphorylation, reverse the decline in brain antioxidant levels, inhibit neuronal oxidative stress and proinflammatory cytokine production, improve synaptic plasticity and neurogenesis, altogether resulting in improved cognitive functions. In addition, most recent findings demonstrate that these neuroprotective effects are associated with Se-induced modulation of gut microbiota. In animal models of AD, Se supplementation was shown to improve gut microbiota biodiversity with a trend to increased relative abundance of Lactobacillus, Bifidobacterium, and Desulfivibrio, while reducing that of Lachnospiracea_NK4A136, Rikenella, and Helicobacter. Moreover, the relative abundance of Se-affected taxa was significantly associated with Aβ accumulation, tau phosphorylation, neuronal oxidative stress, and neuroinflammation, indicative of the potential role of gut microbiota to mediate the neuroprotective effects of Se in AD. Hypothetically, modulation of gut microbiota along with Se supplementation may improve the efficiency of the latter in AD, although further detailed laboratory and clinical studies are required.
Collapse
Affiliation(s)
- Anatoly V Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl, 150000, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Tommaso Filippini
- Environmental, Genetic and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Medical School, University of Modena and Reggio Emilia, Modena, Italy
- School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Viktor A Gritsenko
- Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, 460000, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Health Science Center, School of Public Health, National Health and Family Planning Commission, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiong Guo
- Key Laboratory of Trace Elements and Endemic Diseases, Health Science Center, School of Public Health, National Health and Family Planning Commission, Xi'an Jiaotong University, Xi'an 710061, China
| | - Joao B T Rocha
- Departamento de Bioquímica E Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Alexey A Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl, 150000, Russia.
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia.
| |
Collapse
|
4
|
Morán-Serradilla C, Plano D, Sanmartín C, Sharma AK. Selenization of Small Molecule Drugs: A New Player on the Board. J Med Chem 2024; 67:7759-7787. [PMID: 38716896 DOI: 10.1021/acs.jmedchem.3c02426] [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: 05/24/2024]
Abstract
There is an urgent need to develop safer and more effective modalities for the treatment of a wide range of pathologies due to the increasing rates of drug resistance, undesired side effects, poor clinical outcomes, etc. Throughout the years, selenium (Se) has attracted a great deal of attention due to its important role in human health. Besides, a growing body of work has unveiled that the inclusion of Se motifs into a great number of molecules is a promising strategy for obtaining novel therapeutic agents. In the current Perspective, we have gathered the most recent literature related to the incorporation of different Se moieties into the scaffolds of a wide range of known drugs and their feasible pharmaceutical applications. In addition, we highlight different representative examples as well as provide our perspective on Se drugs and the possible future directions, promises, opportunities, and challenges of this ground-breaking area of research.
Collapse
Affiliation(s)
| | - Daniel Plano
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
- Penn State Cancer Institute, 400 University Drive,Hershey, Pennsylvania 17033, United States
| |
Collapse
|
5
|
Guo M, Xu S, He X, He J, Yang H, Zhang L. Decoding emotional resilience in aging: unveiling the interplay between daily functioning and emotional health. Front Public Health 2024; 12:1391033. [PMID: 38694972 PMCID: PMC11061423 DOI: 10.3389/fpubh.2024.1391033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
Background EPs pose significant challenges to individual health and quality of life, attracting attention in public health as a risk factor for diminished quality of life and healthy life expectancy in middle-aged and older adult populations. Therefore, in the context of global aging, meticulous exploration of the factors behind emotional issues becomes paramount. Whether ADL can serve as a potential marker for EPs remains unclear. This study aims to provide new evidence for ADL as an early predictor of EPs through statistical analysis and validation using machine learning algorithms. Methods Data from the 2018 China Health and Retirement Longitudinal Study (CHARLS) national baseline survey, comprising 9,766 samples aged 45 and above, were utilized. ADL was assessed using the BI, while the presence of EPs was evaluated based on the record of "Diagnosed with Emotional Problems by a Doctor" in CHARLS data. Statistical analyses including independent samples t-test, chi-square test, Pearson correlation analysis, and multiple linear regression were conducted using SPSS 25.0. Machine learning algorithms, including Support Vector Machine (SVM), Decision Tree (DT), and Logistic Regression (LR), were implemented using Python 3.10.2. Results Population demographic analysis revealed a significantly lower average BI score of 65.044 in the "Diagnosed with Emotional Problems by a Doctor" group compared to 85.128 in the "Not diagnosed with Emotional Problems by a Doctor" group. Pearson correlation analysis indicated a significant negative correlation between ADL and EPs (r = -0.165, p < 0.001). Iterative analysis using stratified multiple linear regression across three different models demonstrated the persistent statistical significance of the negative correlation between ADL and EPs (B = -0.002, β = -0.186, t = -16.476, 95% CI = -0.002, -0.001, p = 0.000), confirming its stability. Machine learning algorithms validated our findings from statistical analysis, confirming the predictive accuracy of ADL for EPs. The area under the curve (AUC) for the three models were SVM-AUC = 0.700, DT-AUC = 0.742, and LR-AUC = 0.711. In experiments using other covariates and other covariates + BI, the overall prediction level of machine learning algorithms improved after adding BI, emphasizing the positive effect of ADL on EPs prediction. Conclusion This study, employing various statistical methods, identified a negative correlation between ADL and EPs, with machine learning algorithms confirming this finding. Impaired ADL increases susceptibility to EPs.
Collapse
Affiliation(s)
- Minhua Guo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Songyang Xu
- School of Mechatronics and Energy Engineering, NingboTech University, Ningbo, China
| | - Xiaofang He
- Nursing Department, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Yang
- Department of Neurology, The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, Guizhou, China
| | - Lin Zhang
- Department of Neurology, The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
6
|
Savall ASP, de Mello JD, Fidelis EM, Comis-Neto AA, Nepomuceno MR, Pacheco CDO, Haas SE, Pinton S. Nanoencapsulated Curcumin: Enhanced Efficacy in Reversing Memory Loss in An Alzheimer Disease Model. Brain Sci 2024; 14:130. [PMID: 38391705 PMCID: PMC10886961 DOI: 10.3390/brainsci14020130] [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: 12/20/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Investigating new drugs or formulations that target Alzheimer disease (AD) is critical for advancing therapeutic interventions. Therefore, this study aimed to assess the effectiveness of nanoencapsulated curcumin (NC Curc) in alleviating memory impairment, oxidative stress, and neuroinflammation in a validated AD model. Male Wistar rats were given bilateral intracerebroventricular injections of either saline or streptozotocin (STZ) (3 mg/3 µL/site) to establish the AD model (day 0). On day 22, daily oral administrations of curcumin (6 mg/kg), NC Curc (6 mg/kg), or a vehicle (unloaded NC) were initiated and continued for 14 days. NC Curc significantly reversed memory deficits in object recognition and inhibitory avoidance tests induced by STZ. Both formulations of curcumin attenuated elevated acetylcholinesterase activity caused by STZ. Importantly, NC Curc alone effectively mitigated STZ-induced oxidative stress. Additionally, NC Curc treatment normalized GFAP levels, suggesting a potential reduction in neuroinflammation in STZ-treated rats. Our findings indicate that NC Curc improves memory in an AD rat model, highlighting its enhanced therapeutic effects compared to unencapsulated curcumin. This research significantly contributes to understanding the therapeutic and neurorestorative potential of NC Curc in AD, particularly in reversing pathophysiological changes.
Collapse
Affiliation(s)
- Anne Suély Pinto Savall
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Jhuly Dorneles de Mello
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Eduarda Monteiro Fidelis
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Antonio Alvenir Comis-Neto
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Maria Regina Nepomuceno
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Camila de Oliveira Pacheco
- Laboratory of Pharmacology and Pharmacometrics, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Sandra Elisa Haas
- Laboratory of Pharmacology and Pharmacometrics, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Simone Pinton
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| |
Collapse
|
7
|
Elganzoury SS, Abdelfattah MS, Habotta OA, El-Khadragy M, Abdel Moneim AE, Abdalla MS. Neuro-amelioration of Ficus lyrata (fiddle-leaf fig) extract conjugated with selenium nanoparticles against aluminium toxicity in rat brain: relevance to neurotransmitters, oxidative, inflammatory, and apoptotic events. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:65822-65834. [PMID: 37093386 DOI: 10.1007/s11356-023-26935-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
Aluminium is a non-essential metal, and its accumulation in the brain is linked with potent neurotoxic action and the development of many neurological diseases. This investigation, therefore, intended to examine the antagonistic efficacy of Ficus lyrata (fiddle-leaf fig) extract (FLE) conjugated with selenium nanoparticles (FLE-SeNPs) against aluminium chloride (AlCl3)-induced hippocampal injury in rats. Rats were allocated to five groups: control, FLE, AlCl3 (100 mg/kg), AlCl3 + FLE (100 mg/kg), and AlCl3 + FLE-SeNPs (0.5 mg/kg). All agents were administered orally every day for 42 days. The result revealed that pre-treated rats with FLE-SeNPs showed markedly lower acetylcholinesterase and Na+/K+-ATPase activities in the hippocampus than those in AlCl3 group. Additionally, FLE-SeNPs counteracted the oxidant stress-mediated by AlCl3 by increasing superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, and glutathione contents in rat hippocampus. Besides, the formulated nanoparticles decreased the hippocampal malondialdehyde, carbonyl protein, and nitric oxide levels of AlCl3-exposed animals. Furthermore, FLE-SeNPs attenuated neural tissue inflammation, as demonstrated by decreased interleukin-1 beta, interleukin-6, nuclear factor kappa B, and glial fibrillary acidic protein. Remarkable anti-apoptotic action was exerted by FLE-SeNPs by increasing B cell lymphoma 2 and decreasing caspase-3 and Bcl-2-associated-X protein in AlCl3-exposed rats. The abovementioned results correlated well with the hippocampal histopathological findings. Given these results, SeNPs synthesized with FLE imparted a remarkable neuroprotective action against AlCl3-induced neurotoxicity by reversing oxidative damage, neuronal inflammation, and apoptosis in exposed rats.
Collapse
Affiliation(s)
- Sara S Elganzoury
- Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | | | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Manal El-Khadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt.
| | - Mohga S Abdalla
- Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
8
|
Dong X, Zhou S, Nao J. Kaempferol as a therapeutic agent in Alzheimer's disease: Evidence from preclinical studies. Ageing Res Rev 2023; 87:101910. [PMID: 36924572 DOI: 10.1016/j.arr.2023.101910] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common type of dementia and seriously affects human life and health. Kaempferol (KMP) is a common flavonoid, that is mainly derived from the rhizomes of Kaempferol galanga L. and is widely found in various fruits and vegetables. Previous studies have suggested that KMP has multiple pharmacological activities. However, the anti-AD mechanism of KMP has not been elucidated. METHODS This systematic review aims to summarize the existing preclinical experiments on KMP, further confirm the therapeutic effect of KMP in an AD model, and summarize the possible mechanism by which KMP exerts anti-AD effects. Electronic databases, including PubMed, China National Knowledge Infrastructure (CNKI), Baidu Academic, and Wanfang, were searched using the keywords of 'Kaempferol,' 'KMP,' 'pharmacology,' and 'Alzheimer's disease'. RESULTS We evaluated the reliability of the 12 included studies, and the results showed that the anti-AD mechanism of KMP was reliable and that the prospect of KMP in the treatment of cognitive impairment was promising. We comprehensively assessed the neuroprotective effects of KMP in in vivo and in vitro models of AD. These studies shown that KMP ameliorated AD through several mechanisms, including its antioxidant, anti-inflammatory, anti-apoptotic, and anti-acetylcholinesterase effects. CONCLUSION KMP may exert anti-AD effects through various mechanisms and is a potential drug with broad prospects for the treatment of AD.
Collapse
Affiliation(s)
- Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Siyu Zhou
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
9
|
A Purine Derivative Containing an Organoselenium Group Protects Against Memory Impairment, Sensitivity to Nociception, Oxidative Damage, and Neuroinflammation in a Mouse Model of Alzheimer's Disease. Mol Neurobiol 2023; 60:1214-1231. [PMID: 36427137 DOI: 10.1007/s12035-022-03110-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
In the present study, the effect of 6-((4-fluorophenyl) selanyl)-9H-purine (FSP) was tested against memory impairment and sensitivity to nociception induced by intracerebroventricular injection of amyloid-beta peptide (Aβ) (25-35 fragment), 3 nmol/3 μl/per site in mice. Memory impairment was determined by the object recognition task (ORT) and nociception by the Von-Frey test (VFT). Aβ caused neuroinflammation with upregulation of glial fibrillary acidic protein (GFAP) (in hippocampus), nuclear factor-κB (NF-κB), and the proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) in cerebral cortex and hippocampus. Additionally, Aβ increased oxidant levels and lipid peroxidation in cerebral cortex and hippocampus, but decreased heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prdx1) expression in the hippocampus. Anti-neuroinflammatory effects of FSP were demonstrated by a decrease in the expression of GFAP and NF-κB in the hippocampus, as well as a decrease in proinflammatory cytokines in both the hippocampus and cerebral cortex FSP protected against oxidative stress by decreasing oxidant levels and lipid peroxidation and by increasing HO-1 and Prdx1 expressions in the hippocampus of mice. Moreover, FSP prevented the activation of nuclear factor erythroid 2-related factor 2 (Nrf-2) in the hippocampus of mice induced by Aβ. In conclusion, treatment with FSP attenuated memory impairment, nociception sensitivity by decreasing oxidative stress, and neuroinflammation in a mouse model of Alzheimer's disease.
Collapse
|
10
|
Yang L, Nao J. Ferroptosis: a potential therapeutic target for Alzheimer's disease. Rev Neurosci 2022:revneuro-2022-0121. [PMID: 36514247 DOI: 10.1515/revneuro-2022-0121] [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: 10/03/2022] [Accepted: 10/30/2022] [Indexed: 12/15/2022]
Abstract
The most prevalent dementia-causing neurodegenerative condition is Alzheimer's disease (AD). The aberrant buildup of amyloid β and tau hyperphosphorylation are the two most well-known theories about the mechanisms underlying AD development. However, a significant number of pharmacological clinical studies conducted around the world based on the two aforementioned theories have not shown promising outcomes, and AD is still not effectively treated. Ferroptosis, a non-apoptotic programmed cell death defined by the buildup of deadly amounts of iron-dependent lipid peroxides, has received more attention in recent years. A wealth of data is emerging to support the role of iron in the pathophysiology of AD. Cell line and animal studies applying ferroptosis modulators to the treatment of AD have shown encouraging results. Based on these studies, we describe in this review the underlying mechanisms of ferroptosis; the role that ferroptosis plays in AD pathology; and summarise some of the research advances in the treatment of AD with ferroptosis modulators. We hope to contribute to the clinical management of AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
11
|
Ferreira Schopf P, Peglow Pinz M, Pereira da Motta K, Klein VP, Kolinski Machado A, Rhoden CRB, Wilhelm EA, Luchese C, Zanella I, Sagrillo M. SAFETY PROFILE AND PREVENTION OF COGNITIVE DEFICIT IN ALZHEIMER’S DISEASE MODEL OF GRAPHENE FAMILY NANOMATERIALS, TUCUMA OIL (Astrocaryum vulgare) AND ITS SYNERGISMS. INTERNATIONAL JOURNAL FOR INNOVATION EDUCATION AND RESEARCH 2022; 10:267-303. [DOI: 10.31686/ijier.vol10.iss3.3694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Alzheimer's disease is a worldwide health issue, and there are currently no treatments that can stop this disease. Oxidized graphene derivatives have gained prominence in use in biological systems due to their excellent physical-chemical characteristics, biocompatibility and ability to overcome the blood-brain barrier. Other substances highlighted are those of natural origin from the Amazon biome, such as tucuma, a fruit whose oil has been widely studied in therapeutic applications. Thus, the aim of this study was to investigate the action of graphene oxide, reduced graphene oxide and tucuma oil, isolated and combined, as an alternative for treatment of Alzheimer's disease through studies in silico, in vitro, in vivo and ex vivo. Computational simulation via docking was used to verify the affinity of the substances with the proteins β-amyloid and acetylcholinesterase, in which the reduced graphene oxide was the one that showed the most favorable interaction. The results of the ab initio simulation showed that the synergism between the nanostructures and the oil occurs through physical adsorption. The experimental results revealed that the substances and their combinations were nontoxic, both at the cellular and systemic level. In general, all treatments had positive results against induced memory deficit, but reduced graphene oxide was the most prominent, as it was able to protect against memory damage in all behavioral tests performed, with anticholinesterase activity and antioxidant effect. In conclusion, the reduced graphene oxide is, among the treatments studied, the one with great therapeutic potential to be investigated in the treatment of this disease.
Collapse
|
12
|
da Costa Rodrigues K, Leivas de Oliveira R, da Silva Chaves J, Esteves da Rocha VM, Fuzinato Dos Santos B, Fronza MG, Luís de Campos Domingues N, Savegnago L, Wilhelm EA, Luchese C. A new arylsulfanyl-benzo-2,1,3-thiadiazoles derivative produces an anti-amnesic effect in mice by modulating acetylcholinesterase activity. Chem Biol Interact 2021; 351:109736. [PMID: 34740600 DOI: 10.1016/j.cbi.2021.109736] [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: 06/25/2021] [Revised: 10/20/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022]
Abstract
The aim of the present study was investigate the binding affinity of 5-((4-methoxyphenyl)thio)benzo[c][1,2,5]thiadiazole (MTDZ) with acetylcholinesterase (AChE). We also evaluated the effect of MTDZ against scopolamine (SCO)-induced amnesia in mice and we looked at the toxicological potential of this compound in mice. The binding affinity of MTDZ with AChE was investigated by molecular docking analyses. For an experimental model, male Swiss mice were treated daily with MTDZ (10 mg/kg, intragastrically (i.g.)) or canola oil (10 ml/kg, i.g.), and induced, 30 min later, with injection of SCO (0.4 mg/kg, intraperitoneally (i.p.)) or saline (0.9%, 5 ml/kg, i.p.) daily. From day 1 to day 10, mice were submitted to the behavioral tasks (Barnes maze, open-field, object recognition and location, Y-maze and step-down inhibitory avoidance tasks), 30 min after induction with SCO. On the tenth day, the animals were euthanized and blood was collected for the analysis of biochemical markers (creatinine, aspartate (AST), and alanine (ALT) aminotransferase). MTDZ interacts with residues of the AChE active site. SCO caused amnesia in mice by changing behavioral tasks. MTDZ treatment attenuated the behavioral changes caused by SCO. In ex vivo assay, MTDZ also protected against the alteration of AChE activity, reactive species (RS) levels, thiobarbituric acid reative species (TBARS) levels, catalase (CAT) activity in tissues, as well as in transaminase activities of plasma caused by SCO in mice. In conclusion, MTDZ presented anti-amnesic action through modulation of the cholinergic system and provided protection from kidney and liver damage caused by SCO.
Collapse
Affiliation(s)
- Karline da Costa Rodrigues
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Research Group in Neurobiotechnology (GPN), Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), CEP 96010- 900, Pelotas, RS, Brazil
| | - Renata Leivas de Oliveira
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Research Group in Neurobiotechnology (GPN), Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), CEP 96010- 900, Pelotas, RS, Brazil
| | - Julia da Silva Chaves
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Research Group in Neurobiotechnology (GPN), Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), CEP 96010- 900, Pelotas, RS, Brazil
| | - Vanessa Macedo Esteves da Rocha
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Research Group in Neurobiotechnology (GPN), Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), CEP 96010- 900, Pelotas, RS, Brazil
| | - Beatriz Fuzinato Dos Santos
- Laboratory of Organic Catalysis and Biocatalysis, Federal University of Grande Dourados (UFGD), 79825-070, Dourados, MS, Brazil
| | - Mariana Gallio Fronza
- Postgraduate Program in Biotechnology, GPN, Technological Development Center, UFPel, CEP, 96010-900, Pelotas, RS, Brazil
| | - Nelson Luís de Campos Domingues
- Laboratory of Organic Catalysis and Biocatalysis, Federal University of Grande Dourados (UFGD), 79825-070, Dourados, MS, Brazil
| | - Lucielli Savegnago
- Postgraduate Program in Biotechnology, GPN, Technological Development Center, UFPel, CEP, 96010-900, Pelotas, RS, Brazil
| | - Ethel Antunes Wilhelm
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Research Group in Neurobiotechnology (GPN), Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), CEP 96010- 900, Pelotas, RS, Brazil.
| | - Cristiane Luchese
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Research Group in Neurobiotechnology (GPN), Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), CEP 96010- 900, Pelotas, RS, Brazil.
| |
Collapse
|