1
|
Yu SP, Choi E, Jiang MQ, Wei L. Acute and chronic excitotoxicity in ischemic stroke and late-onset Alzheimer's disease. Neural Regen Res 2025; 20:1981-1988. [PMID: 39101641 PMCID: PMC11691467 DOI: 10.4103/nrr.nrr-d-24-00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/13/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
Stroke and Alzheimer's disease are common neurological disorders and often occur in the same individuals. The comorbidity of the two neurological disorders represents a grave health threat to older populations. This review presents a brief background of the development of novel concepts and their clinical potentials. The activity of glutamatergic N-methyl-D-aspartate receptors and N-methyl-D-aspartate receptor-mediated Ca 2+ influx is critical for neuronal function. An ischemic insult induces prompt and excessive glutamate release and drastic increases of intracellular Ca 2+ mainly via N-methyl-D-aspartate receptors, particularly of those at the extrasynaptic site. This Ca 2+ -evoked neuronal cell death in the ischemic core is dominated by necrosis within a few hours and days known as acute excitotoxicity. Furthermore, mild but sustained Ca 2+ increases under neurodegenerative conditions such as in the distant penumbra of the ischemic brain and early stages of Alzheimer's disease are not immediately toxic, but gradually set off deteriorating Ca 2+ -dependent signals and neuronal cell loss mostly because of activation of programmed cell death pathways. Based on the Ca 2+ hypothesis of Alzheimer's disease and recent advances, this Ca 2+ -activated "silent" degenerative excitotoxicity evolves from years to decades and is recognized as a unique slow and chronic neuropathogenesis. The N-methyl-D-aspartate receptor subunit GluN3A, primarily at the extrasynaptic site, serves as a gatekeeper for the N-methyl-D-aspartate receptor activity and is neuroprotective against both acute and chronic excitotoxicity. Ischemic stroke and Alzheimer's disease, therefore, share an N-methyl-D-aspartate receptor- and Ca 2+ -mediated mechanism, although with much different time courses. It is thus proposed that early interventions to control Ca 2+ homeostasis at the preclinical stage are pivotal for individuals who are susceptible to sporadic late-onset Alzheimer's disease and Alzheimer's disease-related dementia. This early treatment simultaneously serves as a preconditioning therapy against ischemic stroke that often attacks the same individuals during abnormal aging.
Collapse
Affiliation(s)
- Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Emily Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Q. Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Kalra P, Grewal AK, Khan H, Singh TG. Unscrambling the cellular and molecular threads of Neuroplasticity: Insights into Alzheimer's disease pathogenesis. Neuroscience 2025; 571:74-88. [PMID: 39970983 DOI: 10.1016/j.neuroscience.2025.02.037] [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: 12/02/2024] [Revised: 01/14/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is predominantly the most recurring and devastating neurological condition among the elderly population, characterized by the accumulation of amyloid-β (Aβ) and phosphorylated tau proteins, and is accompanied by progressive decline of learning and memory. Due to its complex and multifactorial etiology, a wide variety of therapeutic interventions have been developed. Despite constant advancements in the field, effective treatments that ameliorate the severity of Alzheimer's symptoms or cease their progression are still insufficient. Mounting evidence suggests that synaptic dysfunction could be an essential component of AD pathogenesis as synapse signaling is impaired in the aging brain, which contributes to synaptic decline. Therefore, improving neuroplasticity such as synaptic plasticity or neurogenesis could be a promising therapeutic approach for alleviating the effects of AD. This article reviews the cellular and molecular threads of neuroplasticity as well as targets that restore neuronal survival and plasticity to provide functional recoveries, including receptors, downstream signaling pathways, ion channels, transporters, enzymes, and neurotrophic factors.
Collapse
Affiliation(s)
- Palak Kalra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; University School of Pharmaceutical Sciences, Rayat Bahra University, Mohali, Punjab 140103, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
3
|
Huang Z, Chen G, Ren Z, Xiao L, Chen Z, Xie Y, Wang G, Zhou B. Urolithin A ameliorates schizophrenia-like behaviors and cognitive impairments in female rats by modulating NLRP3 signaling. Int Immunopharmacol 2025; 151:114336. [PMID: 39987632 DOI: 10.1016/j.intimp.2025.114336] [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: 09/19/2024] [Revised: 02/16/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
The management of cognitive impairments in schizophrenia presents a considerable challenge, with a strong association between neuroinflammation and its progression. Urolithin A (UA) demonstrates important anti-inflammatory properties in multiple neurological disease models, contributing to the enhancement of cognitive deficits. However, it remains uncertain if UA can produce comparable neuroregulatory effects in female rat models of schizophrenia. Eight-week-old female Sprague Dawley rats received either 0.1 mg/kg of MK801 or volume-matched saline via intraperitoneal injection for 5 consecutive days. Furthermore, they were administered 150 mg/kg of UA through oral gavage for 4 weeks. Behavioral assessments were performed to evaluate cognitive function and behavior after UA treatment. Immunofluorescence staining was employed to assess microglial activity in the hippocampus, while Western blot analysis was conducted to investigate the expression of neuroinflammation-associated proteins. Prolonged exposure to MK801 induces schizophrenia-like behaviors and cognitive deficits in female rats. It also elevates the expression of NLRP3, Caspase-1, IL-1β, and IL-18 proteins in the hippocampus, accompanied by the activation of microglial cells. However, UA treatment can reverse the expression of these inflammatory proteins and the activation of microglial cells induced by MK801. This is the first study to evaluate the effects of UA on behavior and cognition in a female rat model of schizophrenia. The findings indicate that UA mitigates MK-801-induced cognitive deficits in female rats by inhibiting neuroinflammation and microglial activation via modulation of the NLRP3 signaling pathway. These findings offer preclinical data endorsing the possible application of UA as a dietary supplement to prevent cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Zhengyuan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guanghui Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhongyu Ren
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ziyue Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yinping Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Taikang center for life and medical sciences, Wuhan University, Wuhan, 430071, China.
| | - Benhong Zhou
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
4
|
Nabizadeh F. Connectomics and neurotransmitter receptor profile explain regional tau pathology in Alzheimer's disease. Cereb Cortex 2025; 35:bhaf053. [PMID: 40083151 DOI: 10.1093/cercor/bhaf053] [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: 12/27/2024] [Accepted: 02/03/2025] [Indexed: 03/16/2025] Open
Abstract
Alzheimer's disease tau pathology spreads through neuronal pathways and synaptic connections. Alteration in synaptic activity facilitates tau spreading. Multiple neurotransmitter systems are shown to be implicated in Alzheimer's disease, but their influence on the trans-synaptic spread of tau is not well understood. I aimed to combine resting-state functional magnetic resonance imaging connectomics, neurotransmitter receptor profiles, and tau-PET data to explain the regional susceptibility to tau accumulation. The tau-PET imaging data of 161 amyloid-beta-negative cognitively unimpaired participants as control and 259 amyloid-beta-positive subjects were recruited from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Linear regression analysis revealed that a higher tau-PET z-score is associated with a lower density of nine receptors in the serotonin, dopamine, gamma-aminobutyric acid (GABA), acetylcholine, and glutamate systems. Furthermore, adding four neurotransmitter receptor density z-scores significantly increased the proportion of explained variance by 3% to 7% compared to the epicenter-connectivity distance model in the group-level analysis. Also, adding nine neurotransmitter receptor density z-scores to the epicenter-connectivity distance model increased the explanatory power of variability in individual levels of tau-PET z-score by 3% to 8%. The current study demonstrated the additive value of atlas-based neurotransmitter receptor mapping and individual-level amyloid-beta-PET scans to enhance the connectivity-based explanation of tau accumulation.
Collapse
|
5
|
Vishnumurthy RH, Priya MGR, Tiwari P, Solomon VR. In-silico Studies and Antioxidant and Neuroprotective Assessment of Microencapsulated Celecoxib against Scopolamine-induced Alzheimer's Disease. Curr Pharm Des 2025; 31:320-329. [PMID: 39206485 DOI: 10.2174/0113816128298289240723103828] [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: 12/13/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Alzheimer's disease (AD) is an enervating and chronic progressive neurodegenerative disorder. Celecoxib (CXB) possesses efficacious antioxidants and has neuroprotective, anti-inflammatory, and immunomodulatory properties. However, the poor bioavailability of CXB limits its therapeutic utility. Thus, this study aimed to evaluate the microencapsulated celecoxib MCXB for neuroprotection. METHODOLOGY CXB was screened by molecular docking study using AutoDock (version 5.2), and the following proteins, such as 4EY7, 2HM1, 2Z5X, and 1PBQ were selected for predicting its neuroprotective effect. Scopolamine 20 mg/kg/day for approximately 7 days was administered to albino rats. Pure CXB 100 mg/kg/- day and 200 mg/kg/day, and MCXB 100 mg/kg/day and 200 mg/kg/day were administered, respectively. Further, to assess the oxidative stress, the nitric oxide (NO), superoxide dismutase (SOD), catalase, and lipid peroxidation (LPO) were evaluated using chemical methods. The neurochemical biomarkers like AChE, glutamate, and dopamine were evaluated using the ELISA method. Further, the histopathology of brain cells was carried out to assess the neuro-regeneration and neurodegeneration of the neurons. RESULTS There was a significant binding interaction of CXB (score -6.3, -6.5, -5.1, -9.1) and donepezil (score- 5.5, -7.6, -7.0, and -8.6) with AchE (4EY7), β-secretase (2HM1, monoamine oxidase (2Z5X), and glutamate (1PBQ), respectively. MCXB-treated rats (100 mg/kg/day, 200 mg/kg/day) showed increased SOD levels (p < 0.001), whereas NO, catalase, and LPO levels were significantly (p < 0.001) decreased as compared to scopolamine-treated rats. Further, MCXB-treated rats showed a modulatory effect in the level of dopamine and AchE. However, the glutamate level was significantly (p < 0.001) decreased. CONCLUSION In addition to that, histopathological examination of the hippocampus part showed remarkable improvement in brain cells. So, the findings of the results revealed that MCXB, in a dose-dependent manner, showed a neuroprotective effect against scopolamine-induced AD. This effect may be attributed to the activation of cholinergic pathways.
Collapse
Affiliation(s)
| | - M Gnana Ruba Priya
- College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka 560111, India
| | - Prashant Tiwari
- College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka 560111, India
| | - Viswas Raja Solomon
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy 502 294, India
| |
Collapse
|
6
|
Joodi SA, Ibrahim WW, Khattab MM. Drugs repurposing in the experimental models of Alzheimer's disease. Inflammopharmacology 2025; 33:195-214. [PMID: 39752040 PMCID: PMC11799062 DOI: 10.1007/s10787-024-01608-7] [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: 10/27/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
The currently approved drugs for Alzheimer's disease (AD) are only for symptomatic treatment in the early stages of the disease but they could not halt the neurodegeneration, additionally, the safety profile of the recently developed immunotherapy is a big issue. This review aims to explain the importance of the drugs repurposing technique and strategy to develop therapy for AD. We illustrated the biological alterations in the pathophysiology of AD including the amyloid pathology, the Tau pathology, oxidative stress, mitochondrial dysfunction, neuroinflammation, glutamate-mediated excitotoxicity, insulin signaling impairment, wingless-related integration site/β-catenin signaling, and autophagy. Additionally, we demonstrated the different repurposed drugs in the experimental models of AD including the anti-inflammatory, anti-hypertensive, anti-diabetic, antiepileptic, antidepressant and anticancer drugs. Further, we showed the pipeline and FDA approved drugs for AD. The repurposed drugs have a promising therapeutic activity against AD, confirming the value of the drugs repurposing technique to elucidate curative therapy for AD.
Collapse
Affiliation(s)
- Sheer A Joodi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt.
| | - Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt
| |
Collapse
|
7
|
Flieger J, Forma A, Flieger W, Flieger M, Gawlik PJ, Dzierżyński E, Maciejewski R, Teresiński G, Baj J. Carotenoid Supplementation for Alleviating the Symptoms of Alzheimer's Disease. Int J Mol Sci 2024; 25:8982. [PMID: 39201668 PMCID: PMC11354426 DOI: 10.3390/ijms25168982] [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: 07/26/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by, among other things, dementia and a decline in cognitive performance. In AD, dementia has neurodegenerative features and starts with mild cognitive impairment (MCI). Research indicates that apoptosis and neuronal loss occur in AD, in which oxidative stress plays an important role. Therefore, reducing oxidative stress with antioxidants is a natural strategy to prevent and slow down the progression of AD. Carotenoids are natural pigments commonly found in fruits and vegetables. They include lipophilic carotenes, such as lycopene, α- and β-carotenes, and more polar xanthophylls, for example, lutein, zeaxanthin, canthaxanthin, and β-cryptoxanthin. Carotenoids can cross the blood-brain barrier (BBB) and scavenge free radicals, especially singlet oxygen, which helps prevent the peroxidation of lipids abundant in the brain. As a result, carotenoids have neuroprotective potential. Numerous in vivo and in vitro studies, as well as randomized controlled trials, have mostly confirmed that carotenoids can help prevent neurodegeneration and alleviate cognitive impairment in AD. While carotenoids have not been officially approved as an AD therapy, they are indicated in the diet recommended for AD, including the consumption of products rich in carotenoids. This review summarizes the latest research findings supporting the potential use of carotenoids in preventing and alleviating AD symptoms. A literature review suggests that a diet rich in carotenoids should be promoted to avoid cognitive decline in AD. One of the goals of the food industry should be to encourage the enrichment of food products with functional substances, such as carotenoids, which may reduce the risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (A.F.); (M.F.); (G.T.)
| | - Wojciech Flieger
- Department of Plastic Surgery, St. John’s Cancer Center, ul. Jaczewskiego 7, 20-090 Lublin, Poland; (W.F.)
| | - Michał Flieger
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (A.F.); (M.F.); (G.T.)
| | - Piotr J. Gawlik
- Department of Plastic Surgery, St. John’s Cancer Center, ul. Jaczewskiego 7, 20-090 Lublin, Poland; (W.F.)
| | - Eliasz Dzierżyński
- Department of Plastic Surgery, St. John’s Cancer Center, ul. Jaczewskiego 7, 20-090 Lublin, Poland; (W.F.)
| | - Ryszard Maciejewski
- Institute of Health Sciences, John Paul II Catholic University of Lublin, Konstantynów 1 H, 20-708 Lublin, Poland;
| | - Grzegorz Teresiński
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (A.F.); (M.F.); (G.T.)
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland;
| |
Collapse
|
8
|
Balakrishnan R, Jannat K, Choi DK. Development of dietary small molecules as multi-targeting treatment strategies for Alzheimer's disease. Redox Biol 2024; 71:103105. [PMID: 38471283 PMCID: PMC10945280 DOI: 10.1016/j.redox.2024.103105] [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: 01/15/2024] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Cognitive dysfunction can occur both in normal aging and age-related neurological disorders, such as mild cognitive impairment and Alzheimer's disease (AD). These disorders have few treatment options due to side effects and limited efficacy. New approaches to slow cognitive decline are urgently needed. Dietary interventions (nutraceuticals) have received considerable attention because they exhibit strong neuroprotective properties and may help prevent or minimize AD symptoms. Biological aging is driven by a series of interrelated mechanisms, including oxidative stress, neuroinflammation, neuronal apoptosis, and autophagy, which function through various signaling pathways. Recent clinical and preclinical studies have shown that dietary small molecules derived from natural sources, including flavonoids, carotenoids, and polyphenolic acids, can modulate oxidative damage, cognitive impairments, mitochondrial dysfunction, neuroinflammation, neuronal apoptosis, autophagy dysregulation, and gut microbiota dysbiosis. This paper reviews research on different dietary small molecules and their bioactive constituents in the treatment of AD. Additionally, the chemical structure, effective dose, and specific molecular mechanisms of action are comprehensively explored. This paper also discusses the advantages of using nanotechnology-based drug delivery, which significantly enhances oral bioavailability, safety, and therapeutic effect, and lowers the risk of adverse effects. These agents have considerable potential as novel and safe therapeutic agents that can prevent and combat age-related AD.
Collapse
Affiliation(s)
- Rengasamy Balakrishnan
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju, 27478, South Korea; Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju, 27478, South Korea
| | - Khoshnur Jannat
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju, 27478, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju, 27478, South Korea; Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju, 27478, South Korea.
| |
Collapse
|
9
|
Alsharief M. How Do Retinoids Affect Alzheimer's Disease and Can They Be Novel Drug Candidates? Cureus 2024; 16:e57548. [PMID: 38572181 PMCID: PMC10990449 DOI: 10.7759/cureus.57548] [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] [Accepted: 04/03/2024] [Indexed: 04/05/2024] Open
Abstract
Alzheimer's disease is a chronic, neurological condition that faces many challenges in its management and therapy nowadays highlighting the importance and urgent need of researching new ways of approaching this disease. Retinoic acid and its derivatives, collectively known as the retinoids, are considered promising agents that have disease-modifying properties in affecting Alzheimer's disease. This thesis aims to address the research questions of what the role of retinoids is in Alzheimer's disease, and whether they can be used as a novel drug candidate for treating this condition. Retinoids' properties and agonistic actions on the nuclear receptors retinoic acid receptor (RAR) and retinoic X receptor (RXR) affect various pathways as well as their underlying genetic factors that compose important pathophysiological hallmarks causing the progression of Alzheimer's disease as amyloid β (Aβ) production and deposition, neurofibrillary tangle (NFT) formation and phosphorylation, and inflammatory and autoimmune responses. Retinoic acid inhibits the amplification of these pathways and modifies the disease progression in animal models, proposing a solid basis for human trials. Hence, investigating retinoids as pharmacological agents in human trials has been conducted, and several synthetic analogues have been developed to address issues concerning retinoic acid's instability and short half-life, as well as adverse drug reactions. The most prominent of these analogues is tamibarotene, a stable retinoic acid derivative with a higher half-life, higher specificity to target receptors, and fewer adverse reactions. A number of criteria that explain what a novel drug candidate should have when managing Alzheimer's disease have been formulated, and which also explain why most novel drug candidates other than retinoic acid have failed in achieving clinical results. Most of these candidates share one common trait which is a single-target approach in targeting disease pathways. This means that when administering these agents, their actions are to target a single disease-causing pathway at a time but do not affect other pathways. On the other hand, tamibarotene is a novel drug candidate that targets a range of pathways at once and provides a more comprehensive approach in its pharmacological actions.
Collapse
Affiliation(s)
- Mazen Alsharief
- International Postgraduate Medical Training Scheme (IPGMTS), University Hospitals Birmingham, Birmingham, GBR
| |
Collapse
|
10
|
Long Y, Zhao Z, Xie W, Shi J, Yang F, Zhu D, Jiang P, Tang Q, Ti Z, Jiang B, Yang X, Gao G, Qi W. Kallistatin leads to cognition impairment via downregulating glutamine synthetase. Pharmacol Res 2024; 202:107145. [PMID: 38492829 DOI: 10.1016/j.phrs.2024.107145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
In many neurodegenerative disorders, such as Alzheimer's disease (AD), glutamate-mediated neuronal excitotoxicity is considered the basis for cognitive impairment. The mRNA and protein expression of SERPINA4(Kallistatin) are higher in patients with AD. However, whether Kallistatin plays a regulatory role in glutamate-glutamine cycle homeostasis remains unclear. In this study, we identified impaired cognitive function in Kallistatin transgenic (KAL-TG) mice. Baseline glutamate levels were elevated and miniature excitatory postsynaptic current (mEPSC) frequency was increased in the hippocampus, suggesting the impairment of glutamate homeostasis in KAL-TG mice. Mechanistically, we demonstrated that Kallistatin promoted lysine acetylation and ubiquitination of glutamine synthetase (GS) and facilitated its degradation via the proteasome pathway, thereby downregulating GS. Fenofibrate improved cognitive memory in KAL-TG mice by downregulating serum Kallistatin. Collectively, our study findings provide insights the mechanism by which Kallistatin regulates cognitive impairment, and suggest the potential of fenofibrate to prevente and treat of AD patients with high levels of Kallistatin.
Collapse
Affiliation(s)
- Yanlan Long
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhen Zhao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wanting Xie
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinhui Shi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fengyu Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dan Zhu
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Jiang
- Department of Clinical Medical Laboratory, Guangzhou First People Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qilong Tang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhou Ti
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bin Jiang
- Guangdong Province Key Laboratory of Brain Function and Disease, School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong, China.
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products (Sun Yat-sen University), Guangzhou, China; Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
11
|
Velu L, Pellerin L, Julian A, Paccalin M, Giraud C, Fayolle P, Guillevin R, Guillevin C. Early rise of glutamate-glutamine levels in mild cognitive impairment: Evidence for emerging excitotoxicity. J Neuroradiol 2024; 51:168-175. [PMID: 37777087 DOI: 10.1016/j.neurad.2023.09.003] [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: 03/30/2023] [Revised: 09/16/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
BACKGROUND Use proton magnetic resonance spectroscopy (1H-MRS) non invasive technique to assess the modifications of glutamate-glutamine (Glx) and gammaaminobutyric acid (GABA) brain levels in patients reporting a cognitive complain METHODS: Posterior cingular cortex 1H-MRS spectra of 46 patients (19 male, 27 female) aged 57 to 87 years (mean : 73.32 ± 7.33 years) with a cognitive complaint were examined with a MEGA PRESS sequence at 3T, and compounds Glutamateglutamine (Glx), GABA, Creatine (Cr) and NAA were measured. From this data the metabolite ratios Glx/Cr, GABA/Cr and NAA/Cr were calculated. In addition, all patient performed the Mini Mental State Evaluation (MMSE) and 2 groups were realized with the clinical threshold of 24. RESULTS 16 patients with MMSE 〈 24 and 30 patients with MMSE 〉 24. Significant increase of Glx/Cr in PCC of patients with MMSE 〈 24 compared to patients with MMSE 〉 24. Moreover, GABA/Cr ratio exhibited a trend for a decrease in PCC between the two groups, while they showed a significant decrease NAA/Cr ratio. CONCLUSION Our results concerning Glx are in agreement with a physiopathological hypothesis involving a biphasic variation of glutamate levels associated with excitotoxicity, correlated with the clinical evolution of the disease. These observations suggest that MRS assessment of glutamate levels could be helpful for both diagnosis and classification of cognitive impairment in stage.
Collapse
Affiliation(s)
- Laura Velu
- University Hospital center of Poitiers, Department of Imaging, France
| | - Luc Pellerin
- University of Poitiers and University Hospital center of Poitiers, France
| | - Adrien Julian
- University Hospital Center of Poitiers, Department of neurology, France
| | - Marc Paccalin
- University Hospital Center of Poitiers, Department of neurology, France
| | - Clément Giraud
- University Hospital center of Poitiers, Department of Imaging, France
| | - Pierre Fayolle
- University Hospital center of Poitiers, Department of Imaging, France
| | - Rémy Guillevin
- University Hospital center of Poitiers, Department of Imaging, France
| | - Carole Guillevin
- University Hospital center of Poitiers, Department of Imaging, France.
| |
Collapse
|
12
|
Barnacle JR, Davis AG, Wilkinson RJ. Recent advances in understanding the human host immune response in tuberculous meningitis. Front Immunol 2024; 14:1326651. [PMID: 38264653 PMCID: PMC10803428 DOI: 10.3389/fimmu.2023.1326651] [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: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
Tuberculous meningitis (TBM), the most severe form of tuberculosis, causes death in approximately 25% cases despite antibiotic therapy, and half of survivors are left with neurological disability. Mortality and morbidity are contributed to by a dysregulated immune response, and adjunctive host-directed therapies are required to modulate this response and improve outcomes. Developing such therapies relies on improved understanding of the host immune response to TBM. The historical challenges in TBM research of limited in vivo and in vitro models have been partially overcome by recent developments in proteomics, transcriptomics, and metabolomics, and the use of these technologies in nested substudies of large clinical trials. We review the current understanding of the human immune response in TBM. We begin with M. tuberculosis entry into the central nervous system (CNS), microglial infection and blood-brain and other CNS barrier dysfunction. We then outline the innate response, including the early cytokine response, role of canonical and non-canonical inflammasomes, eicosanoids and specialised pro-resolving mediators. Next, we review the adaptive response including T cells, microRNAs and B cells, followed by the role of the glutamate-GABA neurotransmitter cycle and the tryptophan pathway. We discuss host genetic immune factors, differences between adults and children, paradoxical reaction, and the impact of HIV-1 co-infection including immune reconstitution inflammatory syndrome. Promising immunomodulatory therapies, research gaps, ongoing challenges and future paths are discussed.
Collapse
Affiliation(s)
- James R. Barnacle
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Angharad G. Davis
- The Francis Crick Institute, London, United Kingdom
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Robert J. Wilkinson
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| |
Collapse
|
13
|
Freyssin A, Carles A, Guehairia S, Rubinstenn G, Maurice T. Fluoroethylnormemantine (FENM) shows synergistic protection in combination with a sigma-1 receptor agonist in a mouse model of Alzheimer's disease. Neuropharmacology 2024; 242:109733. [PMID: 37844867 DOI: 10.1016/j.neuropharm.2023.109733] [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: 08/02/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Fluoroethylnormemantine (FENM) is a Memantine derivative with anti-amnesic and neuroprotective activities showed in the Aβ25-35 pharmacological mouse model of Alzheimer's disease (AD). As AD is a complex multi-factorial neurodegenerative pathology, combination therapies relying on drugs acting through different pathways, have been suggested to more adequately address neuroprotection. As several agonists of the sigma-1 receptor (S1R), an intracellular chaperone, are presently in phase 2 or 3 clinical trials in neurodegenetrative diseases including AD, we examined the potentialities of S1R drug-based combinations with FENM, or Memantine. Aβ25-35-treated mice were treated with S1R agonists (PRE-084, Igmesine, Cutamesine) and/or FENM, or Memantine, during 7 days after intracerebroventricular administration of oligomerized Aβ25-35. Mice were then tested for spatial short-term memory on day 8 and non-spatial long-term memory on days 9-10, using the spontaneous alternation or passive avoidance tests, respectively. The FENM or Memantine combination with Donepezil, that non-selectively inhibits acetylcholinesterase and activates S1R, was also tested. The efficacy of combinations using maximal non-active or minimal active doses of S1R agonist or FENM was analyzed using calculations of the combination index, based on simple isobologram representation. Data showed that most of the FENM-based combinations led to synergistic protection against Aβ25-35-induced learning deficits, for both long- and short-term memory responses, with a higher efficiency on the latter. Memantine led to synergistic combination in short-term memory but poorly in long-term memory responses, with either PRE-084 or Donepezil. These study showed that drug combinations based on FENM and S1R agonists may lead to highly effective and synergistic protection in AD, particularly on short-term memory.
Collapse
Affiliation(s)
- Aline Freyssin
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France; ReST Therapeutics, Montpellier, France
| | - Allison Carles
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France
| | | | | | - Tangui Maurice
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France.
| |
Collapse
|
14
|
Sharma C, Mazumder A. A Comprehensive Review on Potential Molecular Drug Targets for the Management of Alzheimer's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:45-56. [PMID: 38305393 DOI: 10.2174/0118715249263300231116062740] [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: 06/20/2023] [Revised: 08/25/2023] [Accepted: 10/04/2023] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is an onset and incurable neurodegenerative disorder that has been linked to various genetic, environmental, and lifestyle factors. Recent research has revealed several potential targets for drug development, such as the prevention of Aβ production and removal, prevention of tau hyperphosphorylation, and keeping neurons alive. Drugs that target numerous ADrelated variables have been developed, and early results are encouraging. This review provides a concise map of the different receptor signaling pathways associated with Alzheimer's Disease, as well as insight into drug design based on these pathways. It discusses the molecular mechanisms of AD pathogenesis, such as oxidative stress, aging, Aβ turnover, thiol groups, and mitochondrial activities, and their role in the disease. It also reviews the potential drug targets, in vivo active agents, and docking studies done in AD and provides prospects for future drug development. This review intends to provide more clarity on the molecular processes that occur in Alzheimer's patient's brains, which can be of use in diagnosing and preventing the condition.
Collapse
Affiliation(s)
- Chanchal Sharma
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| |
Collapse
|
15
|
Yu SP, Jiang MQ, Shim SS, Pourkhodadad S, Wei L. Extrasynaptic NMDA receptors in acute and chronic excitotoxicity: implications for preventive treatments of ischemic stroke and late-onset Alzheimer's disease. Mol Neurodegener 2023; 18:43. [PMID: 37400870 DOI: 10.1186/s13024-023-00636-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/01/2023] [Indexed: 07/05/2023] Open
Abstract
Stroke and late-onset Alzheimer's disease (AD) are risk factors for each other; the comorbidity of these brain disorders in aging individuals represents a significant challenge in basic research and clinical practice. The similarities and differences between stroke and AD in terms of pathogenesis and pathophysiology, however, have rarely been comparably reviewed. Here, we discuss the research background and recent progresses that are important and informative for the comorbidity of stroke and late-onset AD and related dementia (ADRD). Glutamatergic NMDA receptor (NMDAR) activity and NMDAR-mediated Ca2+ influx are essential for neuronal function and cell survival. An ischemic insult, however, can cause rapid increases in glutamate concentration and excessive activation of NMDARs, leading to swift Ca2+ overload in neuronal cells and acute excitotoxicity within hours and days. On the other hand, mild upregulation of NMDAR activity, commonly seen in AD animal models and patients, is not immediately cytotoxic. Sustained NMDAR hyperactivity and Ca2+ dysregulation lasting from months to years, nevertheless, can be pathogenic for slowly evolving events, i.e. degenerative excitotoxicity, in the development of AD/ADRD. Specifically, Ca2+ influx mediated by extrasynaptic NMDARs (eNMDARs) and a downstream pathway mediated by transient receptor potential cation channel subfamily M member (TRPM) are primarily responsible for excitotoxicity. On the other hand, the NMDAR subunit GluN3A plays a "gatekeeper" role in NMDAR activity and a neuroprotective role against both acute and chronic excitotoxicity. Thus, ischemic stroke and AD share an NMDAR- and Ca2+-mediated pathogenic mechanism that provides a common receptor target for preventive and possibly disease-modifying therapies. Memantine (MEM) preferentially blocks eNMDARs and was approved by the Federal Drug Administration (FDA) for symptomatic treatment of moderate-to-severe AD with variable efficacy. According to the pathogenic role of eNMDARs, it is conceivable that MEM and other eNMDAR antagonists should be administered much earlier, preferably during the presymptomatic phases of AD/ADRD. This anti-AD treatment could simultaneously serve as a preconditioning strategy against stroke that attacks ≥ 50% of AD patients. Future research on the regulation of NMDARs, enduring control of eNMDARs, Ca2+ homeostasis, and downstream events will provide a promising opportunity to understand and treat the comorbidity of AD/ADRD and stroke.
Collapse
Affiliation(s)
- Shan P Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA.
| | - Michael Q Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Seong S Shim
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Soheila Pourkhodadad
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
16
|
Neha, Parvez S. Emerging therapeutics agents and recent advances in drug repurposing for Alzheimer's disease. Ageing Res Rev 2023; 85:101815. [PMID: 36529440 DOI: 10.1016/j.arr.2022.101815] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a multivariate and diversified disease and affects the most sensitive areas of the brain, the cerebral cortex, and the hippocampus. AD is a progressive age-related neurodegenerative disease most often associated with memory deficits and cognition that get more worsen over time. The central theory on the pathophysiological hallmark features of AD is characterized by the accumulation of amyloid β (Aβ) peptides, also associated with tau proteins (τ) dysfunctioning which leads to distorted microtubular structure, affects the cholinergic system, and mitochondrial biogenesis. This review emphasizes how simple it is to find novel treatments for AD and focuses on several recently developed medications through repurposing that can speed up traditional drug development.
Collapse
Affiliation(s)
- Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
17
|
Vrabec R, Blunden G, Cahlíková L. Natural Alkaloids as Multi-Target Compounds towards Factors Implicated in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054399. [PMID: 36901826 PMCID: PMC10003045 DOI: 10.3390/ijms24054399] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in elderly people; currently, there is no efficient treatment. Considering the increase in life expectancy worldwide AD rates are predicted to increase enormously, and thus the search for new AD drugs is urgently needed. A great amount of experimental and clinical evidence indicated that AD is a complex disorder characterized by widespread neurodegeneration of the CNS, with major involvement of the cholinergic system, causing progressive cognitive decline and dementia. The current treatment, based on the cholinergic hypothesis, is only symptomatic and mainly involves the restoration of acetylcholine (ACh) levels through the inhibition of acetylcholinesterase (AChE). Since the introduction of the Amaryllidaceae alkaloid galanthamine as an antidementia drug in 2001, alkaloids have been one of the most attractive groups for searching for new AD drugs. The present review aims to comprehensively summarize alkaloids of various origins as multi-target compounds for AD. From this point of view, the most promising compounds seem to be the β-carboline alkaloid harmine and several isoquinoline alkaloids since they can simultaneously inhibit several key enzymes of AD's pathophysiology. However, this topic remains open for further research on detailed mechanisms of action and the synthesis of potentially better semi-synthetic analogues.
Collapse
Affiliation(s)
- Rudolf Vrabec
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Gerald Blunden
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Lucie Cahlíková
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
- Correspondence:
| |
Collapse
|
18
|
Chen X, Gong T, Chen T, Xu C, Li Y, Song Q, Lin L, Oeltzschner G, Edden RAE, Xia Z, Wang G. Altered glutamate-glutamine and amide proton transfer-weighted values in the hippocampus of patients with amnestic mild cognitive impairment: A novel combined imaging diagnostic marker. Front Neurosci 2023; 17:1089300. [PMID: 36908797 PMCID: PMC9995585 DOI: 10.3389/fnins.2023.1089300] [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: 11/04/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Background and purpose Early diagnosis of amnestic mild cognitive impairment (aMCI) and timely management to delay the onset of Alzheimer's disease (AD) would benefit patients. Pathological metabolic changes of excitatory/inhibitory neurotransmitters and abnormal protein deposition in the hippocampus of aMCI may provide a new clue to imaging diagnosis. However, the diagnostic performance using these hippocampal metabolite measurements is still unclear. We aimed to quantify right hippocampal glutamate-glutamine (Glx) and gamma-aminobutyric acid (GABA) levels as well as protein-based amide proton transfer-weighted (APTw) signals of patients with aMCI and investigate the diagnostic performance of these metabolites. Methods In this cross-sectional study, 20 patients with aMCI and 20 age- and gender-matched healthy controls (HCs) underwent MEGA Point Resolved Spectroscopy (MEGA-PRESS) and APTw MR imaging at 3 T. GABA+, Glx, and APTw signals were measured in the right hippocampus. The GABA+ levels, Glx levels, Glx/GABA+ ratios, and APTw values were compared between the HCs and aMCI groups using the Mann-Whitney U test. Binary logistic regression and receiver operating characteristic (ROC) curve analyses were used to evaluate MEGA-PRESS and APTw parameters' diagnostic performance. Results Compared with HCs, patients with aMCI had significantly lower Glx levels in the right hippocampus (7.02 ± 1.41 i.u. vs. 5.81 ± 1.33 i.u., P = 0.018). No significant changes in the GABA+ levels were observed in patients with aMCI (HCs vs. aMCI: 2.54 ± 0.28 i.u. vs. 2.47 ± 0.36 i.u., P = 0.620). In addition, Glx/GABA+ ratios between the two groups (HCs vs. aMCI: 2.79 ± 0.60 vs. 2.37 ± 0.55, P = 0.035) were significantly different. Compared with HCs, patients with aMCI showed higher APTw values in the right hippocampus (0.99 ± 0.26% vs. 1.26% ± 0.28, P = 0.006). The ROC curve analysis showed that Glx, GABA+, Glx/GABA+, and APTw values had an area under the curve (AUC) of 0.72, 0.55, 0.70, and 0.75, respectively, for diagnosing aMCI. In the ROC curve analysis, the AUC of the combination of the parameters increased to 0.88, which is much higher than that observed in the univariate analysis (P < 0.05). Conclusion The combination of right hippocampal Glx levels and APTw values improved the diagnostic performance for aMCI, indicating it as a promising combined imaging diagnostic marker. Our study provided a potential imaging diagnostic strategy of aMCI, which may promote early detection of aMCI and facilitate timely intervention to delay the pathological progress toward AD.
Collapse
Affiliation(s)
- Xin Chen
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Neurology, Liaocheng People's Hospital, Liaocheng, China.,Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Gong
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tong Chen
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changyuan Xu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuchao Li
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qingxu Song
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | | | - Georg Oeltzschner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Richard A E Edden
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China.,Department of Neurology, Liaocheng Clinical School of Shandong First Medical University, Liaocheng, China
| | - Guangbin Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
19
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
20
|
Mandal PK, Roy RG, Samkaria A. Oxidative Stress: Glutathione and Its Potential to Protect Methionine-35 of Aβ Peptide from Oxidation. ACS OMEGA 2022; 7:27052-27061. [PMID: 35967059 PMCID: PMC9366984 DOI: 10.1021/acsomega.2c02760] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/07/2022] [Indexed: 05/15/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with heterogeneous etiology. Intracellular neurofibrillary tangles caused by tau (τ) protein phosphorylation and extracellular senile plaques caused by aggregation of amyloid-beta (Aβ) peptide are characteristic histopathological hallmarks of AD. Oxidative stress (OS) is also suggested to play a role in the pathophysiology of AD. The antioxidant glutathione (GSH) is able to mitigate OS through the detoxification of free radicals. The clearance of these free radicals is reported to be affected when there is a decline in GSH levels in AD. These radicals further react with the methionine-35 (M-35) residue of Aβ and facilitate its subsequent oligomerization. This review presents a plausible model indicating the role of master antioxidant GSH to protect M35 of Aβ1-40/Aβ1-42 from oxidation in pathological conditions as compared to healthy controls.
Collapse
Affiliation(s)
- Pravat K. Mandal
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon Haryana 122051, India
- Florey
Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Parkville, VIC 3052, Australia
| | - Rimil Guha Roy
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon Haryana 122051, India
| | - Avantika Samkaria
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon Haryana 122051, India
| |
Collapse
|
21
|
Rummel NG, Butterfield DA. Altered Metabolism in Alzheimer Disease Brain: Role of Oxidative Stress. Antioxid Redox Signal 2022; 36:1289-1305. [PMID: 34416829 PMCID: PMC9229240 DOI: 10.1089/ars.2021.0177] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Significance: Alzheimer disease (AD) is an all-too-common condition in the aging population. However, aging does not automatically equal neurodegeneration and memory decline. Recent Advances: This review article involves metabolic changes in the AD brain that are related to oxidative stress. Selected pathways are identified as potential targets for intervention in AD. Critical Issues: One of the main factors of AD is the oxidative imbalance within the central nervous system, causing a disruption in metabolic processes. Reactive oxygen species (ROS) are a natural consequence of many cellular processes, especially those associated with mitochondria, such as the electron transport chain. Some ROS, when kept under control and maintained at reasonable levels, often play roles in cell signaling. The cellular damage of ROS arises when oxidative imbalance occurs, in which case ROS are not controlled, leading to a myriad of alterations in cellular metabolic processes. These altered pathways include, among others, dysfunctional glycolysis, calcium regulation, lipid metabolism, mitochondrial processes, and mammalian target of rapamycin pathway dysregulation. Future Directions: Understanding how ROS can lead to these alterations can, ideally, elucidate therapeutic options for retarding AD progression in the aging population. Antioxid. Redox Signal. 36, 1289-1305.
Collapse
Affiliation(s)
- Nicole G Rummel
- Department of Chemistry and University of Kentucky, Lexington, Kentucky, USA
| | - D Allan Butterfield
- Department of Chemistry and University of Kentucky, Lexington, Kentucky, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
22
|
Wu S, Zhou J, Zhang H, Barger SW. Serine Racemase Expression Differentiates Aging from Alzheimer's Brain. Curr Alzheimer Res 2022; 19:494-502. [PMID: 35929621 DOI: 10.2174/1567205019666220805105106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/27/2023]
Abstract
Aging is an inevitable process characterized by progressive loss of physiological integrity and increased susceptibility to cancer, diabetes, cardiovascular, and neurodegenerative diseases; aging is the primary risk factor for Alzheimer's disease (AD), the most common cause of dementia. AD is characterized by brain pathology, including extracellular deposition of amyloid aggregation and intracellular accumulation of neurofibrillary tangles composed of hyperphosphorylated tau protein. In addition, losses of synapses and a wide range of neurons are pivotal pathologies in the AD brain. Accumulating evidence demonstrates hypoactivation of hippocampal neural networks in the aging brain, whereas AD-related mild cognitive impairment (AD-MCI) begins with hyperactivation, followed by a diminution of hippocampal activity as AD develops. The biphasic trends of the activity of the hippocampal neural network are consistent with the alteration of N-methyl-D-aspartate receptor (NMDA-R) activity from aging to prodromal (AD-MCI) to mid-/late stage AD. D-serine, a product of racemization catalyzed by serine racemase (SR), is an important co-agonist of the NMDA-R which is involved in synaptic events including neurotransmission, synaptogenesis, long-term potentiation (LTP), development, and excitotoxicity. SR and D-serine are decreased in the hippocampus of the aging brain, correlating with impairment of cognitive function. By contrast, SR is increased in AD brain, which is associated with a greater degree of cognitive dysfunction. Emerging studies suggest that D-serine levels in the brain or in cerebral spinal fluid from AD patients are higher than in age-matched controls, but the results are inconsistent. Very recently, serum D-serine levels in AD were reported to correlate with sex and clinical dementia rating (CDR) stage. This review will discuss alterations of NMDA-R and SR in aging and AD brain, and the mechanisms underlying the differential regulation of SR will be probed. Collectively, we propose that SR may be a molecular switch that distinguishes the effects of aging from those of AD on the brain.
Collapse
Affiliation(s)
- Shengzhou Wu
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Wenzhou, Zhejiang 325003, P.R. China
| | - Jing Zhou
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Wenzhou, Zhejiang 325003, P.R. China
| | - He Zhang
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Wenzhou, Zhejiang 325003, P.R. China.,College of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang, P.R. China
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock AR, USA.,Geriatric Research, Education & Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock AR, USA
| |
Collapse
|
23
|
Mendes D, Peixoto F, Oliveira MM, Andrade PB, Videira RA. Mitochondria research and neurodegenerative diseases: on the track to understanding the biological world of high complexity. Mitochondrion 2022; 65:67-79. [PMID: 35623557 DOI: 10.1016/j.mito.2022.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/20/2022] [Accepted: 05/22/2022] [Indexed: 12/18/2022]
Abstract
From the simple unicellular eukaryote to the highly complex multicellular organism like Human, mitochondrion emerges as a ubiquitous player to ensure the organism's functionality. It is popularly known as "the powerhouse of the cell" by its key role in ATP generation. However, our understanding of the physiological relevance of mitochondria is being challenged by data obtained in different fields. In this review, a short history of the mitochondria research field is presented, stressing the findings and questions that allowed the knowledge advances, and put mitochondrion as the main player of safeguarding organism life as well as a key to solve the puzzle of the neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniela Mendes
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, no 228, Porto 4050-313, Portugal
| | - Francisco Peixoto
- Chemistry Center - Vila Real (CQ-VR), Biological and Environment Department, School of Life and Environmental Sciences, University of Trás-os-Montes e Alto Douro, UTAD, P.O. Box 1013, 5001-801 Vila Real, Portugal
| | - Maria M Oliveira
- Chemistry Center - Vila Real (CQ-VR), Chemistry Department, School of Life and Environmental Sciences, University of Trás-os-Montes e Alto Douro, UTAD, P.O. Box 1013, 5001-801 Vila Real, Portugal
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, no 228, Porto 4050-313, Portugal
| | - Romeu A Videira
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, no 228, Porto 4050-313, Portugal.
| |
Collapse
|
24
|
Faßbender RV, Goedecke J, Visser-Vandewalle V, Fink GR, Onur OA. [Brain Stimulation for the Treatment of Dementia]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2022; 90:336-342. [PMID: 35483888 DOI: 10.1055/a-1787-0335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Due to the increasing number of cases of Alzheimer's disease and the relatively moderate success with the available symptomatic and causal pharmacological therapies, there is a considerable need to explore non-pharmacological treatment options. In the field of non-invasive brain stimulation (NIBS), various methods have been investigated, particularly transcranial magnetic stimulation and transcranial electrical stimulation. In addition, deep brain stimulation (DBS) is currently being researched as an innovative method for targeted neuromodulation. Both non-invasive and invasive approaches aim to modulate neuronal activity and improve cognitive-mnestic functions. Secondary mechanisms such as long-term potentiation in NIBS or neurogenesis in DBS could also achieve long-term positive effects. Preclinical and clinical studies have already shown promising results in patients in early stages of Alzheimer's disease. However, inconsistent study and stimulation protocols and small sample sizes make it difficult to assess efficacy. Further research is warranted to enable the use of non-invasive or invasive neuromodulatory approaches in clinical practice in the near future.
Collapse
Affiliation(s)
- Ronja V Faßbender
- Klinik und Poliklinik für Neurologie, Medizinische Fakultät und Uniklinik Köln, Universität zu Köln, Köln, Germany.,Institut für Neurowissenschaften (INM-3), Forschungszentrum Jülich, Jülich, Germany
| | - Jana Goedecke
- Klinik und Poliklinik für Neurologie, Medizinische Fakultät und Uniklinik Köln, Universität zu Köln, Köln, Germany
| | - Veerle Visser-Vandewalle
- Klinik und Poliklinik für Stereotaxie und Funktionelle Neurochirurgie, Medizinische Fakultät und Uniklinik Köln, Universität zu Köln, Köln, Germany
| | - Gereon R Fink
- Klinik und Poliklinik für Neurologie, Medizinische Fakultät und Uniklinik Köln, Universität zu Köln, Köln, Germany.,Institut für Neurowissenschaften (INM-3), Forschungszentrum Jülich, Jülich, Germany
| | - Oezguer A Onur
- Klinik und Poliklinik für Neurologie, Medizinische Fakultät und Uniklinik Köln, Universität zu Köln, Köln, Germany.,Institut für Neurowissenschaften (INM-3), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
25
|
Sharma M, Tiwari V, Chaturvedi S, Wahajuddin M, Shukla S, Panda JJ. Self-Fluorescent Lone Tryptophan Nanoparticles as Theranostic Agents Against Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2022; 14:13079-13093. [PMID: 35263093 DOI: 10.1021/acsami.2c01090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Aggregation of β-amyloid (Aβ42) peptide in the neural extracellular space leads to cellular dysfunction, resulting in Alzheimer's disease (AD). The hydrophobic core of the amyloidogenic Aβ42 peptide contains aromatic residues that play an important role in the self-assembly and subsequent aggregation of the peptide. Hence, targeting these hydrophobic core residues by potent low molecular agents can be a promising therapeutic approach toward AD. In the current work, we have developed self-fluorescent solo tryptophan nanoparticles (TNPs) as nanotheranostic systems against AD. We demonstrated that TNPs could significantly inhibit as well as disrupt the fibrils formed by both Aβ42 peptide and another reductionist approach-based amyloid model dipeptide, phenylalanine-phenylalanine (FF). More importantly, these nanostructures were nontoxic to neural cells and could protect the neurons from Aβ42 peptide and FF aggregate-induced cytotoxicity. In addition, efficacy studies performed in animal model further revealed that the TNPs could rescue spatial and learning memory in intracerebroventricular streptozotocin-administration-induced AD phenotype in rats. Moreover, our pharmacokinetics study further established the BBB permeability and brain delivery potency of TNPs. The inherent excellent fluorescent properties of these nanoparticles could be exploited further to use them as imaging modalities for tagging and detecting FF and Aβ42 peptide fibrils. Overall, our results clearly illustrated that the solo TNPs could serve as promising nanotheranostic agents for AD therapy.
Collapse
Affiliation(s)
- Manju Sharma
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| | - Virendra Tiwari
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Swati Chaturvedi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Shubha Shukla
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| |
Collapse
|
26
|
Babaei P. NMDA and AMPA receptors dysregulation in Alzheimer's disease. Eur J Pharmacol 2021; 908:174310. [PMID: 34265291 DOI: 10.1016/j.ejphar.2021.174310] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition characterized by cognitive dysfunction and synaptic failure. The current therapeutic approaches are mainly focused on symptomatic treatment and possess limited effectiveness in addressing the pathophysiology of AD. It is known that neurodegeneration is negatively correlated with synaptic plasticity. This negative correlation highlights glutamatergic neurotransmission via N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors and (AMPA) receptors as a critical mediator of synaptic plasticity. Despite this favorable role, extensive extracellular glutamate concentration induces excitotoxicity and neurodegeneration. NMDA receptors containing GluN2A subunits are located at synaptic sites, implicated in the protective pathways. In comparison, GluN2B containing receptors are located mainly at extrasynaptic sites and increase neuronal vulnerability. AMPA receptors are consistently endocytosed and recycled back to the membrane. An increase in the rate of endocytosis has been implicated as a part of AD pathophysiology through inducing long-term depression (LTD) and synaptic disintegration. In the present review, we focused on the mechanisms of glutamatergic system dysregulation in AD, particularly on its interaction with amyloid-beta. We concluded that assigning a specific role to an individual subtype of either NMDA receptors or AMPA receptors might be an oversimplification as they are not static receptors. Therefore, any imbalance between synaptic and extrasynaptic NMDA receptors and a reduced number of surface AMPA receptors will lead to synaptopathy.
Collapse
Affiliation(s)
- Parvin Babaei
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Cellular &Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
27
|
Khan AF, Adewale Q, Baumeister TR, Carbonell F, Zilles K, Palomero-Gallagher N, Iturria-Medina Y. Personalized brain models identify neurotransmitter receptor changes in Alzheimer's disease. Brain 2021; 145:1785-1804. [PMID: 34605898 DOI: 10.1093/brain/awab375] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 11/12/2022] Open
Abstract
Alzheimer's disease (AD) involves many neurobiological alterations from molecular to macroscopic spatial scales, but we currently lack integrative, mechanistic brain models characterizing how factors across different biological scales interact to cause clinical deterioration in a way that is subject-specific or personalized. Neurotransmitter receptors, as important signaling molecules and potential drug targets, are key mediators of interactions between many neurobiological processes altered in AD. We present a neurotransmitter receptor-enriched multifactorial brain model, which integrates spatial distribution patterns of 15 neurotransmitter receptors from post-mortem autoradiography with multiple in-vivo neuroimaging modalities (tau, amyloid-β and glucose PET, and structural, functional and arterial spin labeling MRI) in a personalized, generative, whole-brain formulation. Applying this data-driven model to a heterogeneous aged population (N = 423, ADNI data), we observed that personalized receptor-neuroimaging interactions explained about 70% (± 20%) of the across-population variance in longitudinal changes to the six neuroimaging modalities, and up to 39.7% (P < 0.003, FWE-corrected) of inter-individual variability in AD cognitive deterioration via an axis primarily affecting executive function. Notably, based on their contribution to the clinical severity in AD, we found significant functional alterations to glutamatergic interactions affecting tau accumulation and neural activity dysfunction, and GABAergic interactions concurrently affecting neural activity dysfunction, amyloid and tau distributions, as well as significant cholinergic receptor effects on tau accumulation. Overall, GABAergic alterations had the largest effect on cognitive impairment (particularly executive function) in our AD cohort (N = 25). Furthermore, we demonstrate the clinical applicability of this approach by characterizing subjects based on individualized 'fingerprints' of receptor alterations. This study introduces the first robust, data-driven framework for integrating several neurotransmitter receptors, multi-modal neuroimaging and clinical data in a flexible and interpretable brain model. It enables further understanding of the mechanistic neuropathological basis of neurodegenerative progression and heterogeneity, and constitutes a promising step towards implementing personalized, neurotransmitter-based treatments.
Collapse
Affiliation(s)
- Ahmed Faraz Khan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | - Quadri Adewale
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | - Tobias R Baumeister
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | | | - Karl Zilles
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany.,Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany.,Department of Psychiatry, Psychotherapy, and Psychosomatics, Medical Faculty, RWTH Aachen, 52074 Aachen, Germany.,JARA, Translational Brain Medicine, 52074 Aachen, Germany
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | | |
Collapse
|
28
|
Yeung JHY, Palpagama TH, Wood OWG, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. EAAT2 Expression in the Hippocampus, Subiculum, Entorhinal Cortex and Superior Temporal Gyrus in Alzheimer's Disease. Front Cell Neurosci 2021; 15:702824. [PMID: 34588956 PMCID: PMC8475191 DOI: 10.3389/fncel.2021.702824] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/23/2021] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a neuropathological disorder characterized by the presence and accumulation of amyloid-beta plaques and neurofibrillary tangles. Glutamate dysregulation and the concept of glutamatergic excitotoxicity have been frequently described in the pathogenesis of a variety of neurodegenerative disorders and are postulated to play a major role in the progression of AD. In particular, alterations in homeostatic mechanisms, such as glutamate uptake, have been implicated in AD. An association with excitatory amino acid transporter 2 (EAAT2), the main glutamate uptake transporter, dysfunction has also been described. Several animal and few human studies examined EAAT2 expression in multiple brain regions in AD but studies of the hippocampus, the most severely affected brain region, are scarce. Therefore, this study aims to assess alterations in the expression of EAAT2 qualitatively and quantitatively through DAB immunohistochemistry (IHC) and immunofluorescence within the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus (STG) regions, between human AD and control cases. Although no significant EAAT2 density changes were observed between control and AD cases, there appeared to be increased transporter expression most likely localized to fine astrocytic branches in the neuropil as seen on both DAB IHC and immunofluorescence. Therefore, individual astrocytes are not outlined by EAAT2 staining and are not easily recognizable in the CA1–3 and dentate gyrus regions of AD cases, but the altered expression patterns observed between AD and control hippocampal cases could indicate alterations in glutamate recycling and potentially disturbed glutamatergic homeostasis. In conclusion, no significant EAAT2 density changes were found between control and AD cases, but the observed spatial differences in transporter expression and their functional significance will have to be further explored.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Oliver W G Wood
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
Abstract
The detection of neurotransmitter release from reprogrammed human cell is an important demonstration of their functionality. Electrochemistry has the distinct advantages over alternative methods that it allows for the measuring of the analyte of interest at a high temporal resolution. This is necessary for fast events, such as neurotransmitter release and reuptake, which happen in the order of milliseconds to seconds. The precise description of these kinetic events can lead to insights into the function of cells in health and disease and allows for the exploration of events that might be missed using methods that look at absolute concentration values or methods that have a slower sampling rate. In the present chapter, we describe the use of constant potential amperometry and enzyme-coated multielectrode arrays for the detection of glutamate in vitro. These biosensors have the distinct advantage of "self-referencing," a method providing high selectivity while retaining outstanding temporal resolution. Here, we provide a step-by-step user guide for a commercially available system and its application for in vitro systems such as reprogrammed cells.
Collapse
|
30
|
Mancini G, Dias C, Lourenço CF, Laranjinha J, de Bem A, Ledo A. A High Fat/Cholesterol Diet Recapitulates Some Alzheimer's Disease-Like Features in Mice: Focus on Hippocampal Mitochondrial Dysfunction. J Alzheimers Dis 2021; 82:1619-1633. [PMID: 34219714 DOI: 10.3233/jad-210122] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ample evidence from clinical and pre-clinical studies suggests mid-life hypercholesterolemia as a risk factor for developing Alzheimer's disease (AD) at a later age. Hypercholesterolemia induced by dietary habits can lead to vascular perturbations that increase the risk of developing sporadic AD. OBJECTIVE To investigate the effects of a high fat/cholesterol diet (HFCD) as a risk factor for AD by using a rodent model of AD and its correspondent control (healthy animals). METHODS We compared the effect of a HFCD in normal mice (non-transgenic mice, NTg) and the triple transgenic mouse model of AD (3xTgAD). We evaluated cognitive performance in relation to changes in oxidative metabolism and neuron-derived nitric oxide (•NO) concentration dynamics in hippocampal slices as well as histochemical staining of markers of the neurovascular unit. RESULTS In NTg, the HFCD produced only moderate hypercholesterolemia but significant decline in spatial memory was observed. A tendency for decrease in •NO production was accompanied by compromised mitochondrial function with decrease in spare respiratory capacity. In 3xTgAD mice, a robust increase in plasma cholesterol levels with the HFCD did not worsen cognitive performance but did induce compromise of mitochondrial function and significantly decreased •NO production. We found increased staining of biomarkers for astrocyte endfeet and endothelial cells in 3xTgAD hippocampi, which was further increased by the HFCD. CONCLUSION A short term (8 weeks) intervention with HFCD can produce an AD-like phenotype even in the absence of overt systemic hypercholesterolemia and highlights mitochondrial dysfunction as a link between hypercholesterolemia and sporadic AD.
Collapse
Affiliation(s)
- Gianni Mancini
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Candida Dias
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Catia F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Joao Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Andreza de Bem
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
31
|
Yeung JHY, Walby JL, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. Glutamatergic receptor expression changes in the Alzheimer's disease hippocampus and entorhinal cortex. Brain Pathol 2021; 31:e13005. [PMID: 34269494 PMCID: PMC8549033 DOI: 10.1111/bpa.13005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's Disease (AD) is the leading form of dementia worldwide. Currently, the pathological mechanisms underlying AD are not well understood. Although the glutamatergic system is extensively implicated in its pathophysiology, there is a gap in knowledge regarding the expression of glutamate receptors in the AD brain. This study aimed to characterize the expression of specific glutamate receptor subunits in post‐mortem human brain tissue using immunohistochemistry and confocal microscopy. Free‐floating immunohistochemistry and confocal laser scanning microscopy were used to quantify the density of glutamate receptor subunits GluA2, GluN1, and GluN2A in specific cell layers of the hippocampal sub‐regions, subiculum, entorhinal cortex, and superior temporal gyrus. Quantification of GluA2 expression in human post‐mortem hippocampus revealed a significant increase in the stratum (str.) moleculare of the dentate gyrus (DG) in AD compared with control. Increased GluN1 receptor expression was found in the str. moleculare and hilus of the DG, str. oriens of the CA2 and CA3, str. pyramidale of the CA2, and str. radiatum of the CA1, CA2, and CA3 subregions and the entorhinal cortex. GluN2A expression was significantly increased in AD compared with control in the str. oriens, str. pyramidale, and str. radiatum of the CA1 subregion. These findings indicate that the expression of glutamatergic receptor subunits shows brain region‐specific changes in AD, suggesting possible pathological receptor functioning. These results provide evidence of specific glutamatergic receptor subunit changes in the AD hippocampus and entorhinal cortex, indicating the requirement for further research to elucidate the pathophysiological mechanisms it entails, and further highlight the potential of glutamatergic receptor subunits as therapeutic targets.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joshua L Walby
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
32
|
Roy J, Tsui KC, Ng J, Fung ML, Lim LW. Regulation of Melatonin and Neurotransmission in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22136841. [PMID: 34202125 PMCID: PMC8268832 DOI: 10.3390/ijms22136841] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease is a neurodegenerative disorder associated with age, and is characterized by pathological markers such as amyloid-beta plaques and neurofibrillary tangles. Symptoms of AD include cognitive impairments, anxiety and depression. It has also been shown that individuals with AD have impaired neurotransmission, which may result from the accumulation of amyloid plaques and neurofibrillary tangles. Preclinical studies showed that melatonin, a monoaminergic neurotransmitter released from the pineal gland, is able to ameliorate AD pathologies and restore cognitive impairments. Theoretically, inhibition of the pathological progression of AD by melatonin treatment should also restore the impaired neurotransmission. This review aims to explore the impact of AD on neurotransmission, and whether and how melatonin can enhance neurotransmission via improving AD pathology.
Collapse
|
33
|
Zhao A, Ma B, Xu L, Yao M, Zhang Y, Xue B, Ren J, Chang D, Liu J. Jiedu Tongluo Granules Ameliorates Post-stroke Depression Rat Model via Regulating NMDAR/BDNF Signaling Pathway. Front Pharmacol 2021; 12:662003. [PMID: 34093193 PMCID: PMC8173625 DOI: 10.3389/fphar.2021.662003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/28/2021] [Indexed: 01/26/2023] Open
Abstract
Post-stroke depression (PSD) is one of the most common stroke complications, which seriously affects stroke’s therapeutic effect and brings great pain for patients. The pathological mechanism of PSD has not been revealed. Jiedu Tongluo granules (JDTLG) is an effective traditional Chinese medicine for PSD treatment which is widely used in clinical treatment. JDTLG has a significant therapeutic effect against PSD, but the mechanism is still unclear. The PSD rat model was established by carotid artery embolization combined with chronic sleep deprivation followed by treating with JDTLG. Neurobehavioral and neurofunctional experiments were engaged in studying the neural function of rats. Histomorphology, proteomics, and western blotting researches were performed to investigate the potential molecular mechanisms related to JDTLG therapy. Oral treatment of JDTLG could significantly improve the symptoms of neurological deficit and depression symptoms of PSD rats. Proteomic analysis identified several processes that may involve the regulation of JDTLG on the PSD animal model, including energy metabolism, nervous system, and N-methyl-D-aspartate receptor (NMDAR)/brain-derived neurotrophic factor (BDNF) signal pathway. Our results showed that JDTLG could reduce glutamate (Glu) level and increase gamma-aminobutyric acid (GABA) level via regulating the NMDAR/BDNF pathway, which may play a vital role in the occurrence and development of PSD.
Collapse
Affiliation(s)
- Aimei Zhao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Bo Ma
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Xu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Mingjiang Yao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yehao Zhang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingjie Xue
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Junguo Ren
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Dennis Chang
- NICM, Western Sydney University, Penrith, NSW, Australia
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
34
|
Ashourpour F, Jafari A, Babaei P. Chronic administration of Tat-GluR23Y ameliorates cognitive dysfunction targeting CREB signaling in rats with amyloid beta neurotoxicity. Metab Brain Dis 2021; 36:701-709. [PMID: 33420884 DOI: 10.1007/s11011-020-00662-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 12/25/2020] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is behaviorally characterized by memory impairments, and pathologically by amyloid β1-42 (Aβ1-42) plaques and tangles. Aβ binds to excitatory synapses and disrupts their transmission due to dysregulation of the glutamate receptors. Here we hypothesized that chronic inhibition of the endocytosis of AMPA receptors together with GluN2B subunit of NMDA receptors might improve cognition deficit induced by Aβ(1-42) neurotoxicity. Forty male Wistar rats were used in this study and divided into 5 groups: Saline + Saline, Aβ+Saline, Aβ+Ifen (Ifenprodil, 3 nmol /2 weeks), Aβ+GluR23Y (Tat-GluR23Y 3 μmol/kg/2 weeks) and Aβ+Ifen+GluR23Y (same doses and durations). Aβ(1-42) neurotoxicity was induced by intracerebroventricular (ICV) injection of Aβ1-42 (2 μg/μl/side), and then animals received the related treatments for 14 days. Cognitive performance of rats and hippocampal level of cAMP-response element-binding (CREB) were evaluated using Morris Water Maze (MWM), and western blotting respectively. Obtained data from the acquisition trials were analyzed by two way Anova and Student T test. Also one way Analysis of variance (ANOVA) with post hoc Tuckey were used to clarify between groups differences in probe test. The Group receiving Aβ, showed significant cognition deficit (long latency to platform and short total time spent in target quadrant (TTS), parallel with lower level of hippocampal CREB, versus vehicle group. While, Aβ+ GluR23Y exhibited the shortest latency to platform and the longest TTS during the probe test, parallel with the higher hippocampal level of CREB compared with other groups. The present study provides evidence that chronic administration of Tat-GluR23Y; an inhibitor of GluA2-AMPARs endocytosis, successfully restores spatial memory impaired by amyloid beta neurotoxicity targeting CREB signaling pathway.
Collapse
Affiliation(s)
- Fatemeh Ashourpour
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, 8th Km of Rasht -Tehran road, Guilan University Complex, Rasht, Guilan, 41996-13769, Iran
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvin Babaei
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, 8th Km of Rasht -Tehran road, Guilan University Complex, Rasht, Guilan, 41996-13769, Iran.
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
35
|
Zhang X, Ma C, Sun L, He Z, Feng Y, Li X, Gan J, Chen X. Effect of policosanol from insect wax on amyloid β-peptide-induced toxicity in a transgenic Caenorhabditis elegans model of Alzheimer's disease. BMC Complement Med Ther 2021; 21:103. [PMID: 33785017 PMCID: PMC8011155 DOI: 10.1186/s12906-021-03278-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 03/17/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD), an age-related neurodegenerative disorder and a serious public health concern, is mainly caused by β-amyloid (Aβ)-induced toxicity. Currently, a limited number of drugs are effective against AD, and only a few are used for its treatment. According to traditional Chinese medicine, white wax is mainly composed of policosanol, hexacosanol, and octacosanol. Policosanol has been shown to reduce lipid levels in blood and alleviate the symptoms associated with diabetic complications and neurodegenerative disorders, such as Parkinson's disease and AD. However, the efficacy of policosanol depends on the purity and composition of the preparation, and the therapeutic efficacy of policosanol derived from insect wax (PIW) in AD is unknown. METHODS Here, we identified the main components of PIW and investigated the effects of PIW on Aβ-induced toxicity and life-span in a transgenic Caenorhabditis elegans model of AD, CL4176. Furthermore, we estimated the expression of amyloid precursor-like protein (apl-1) and the genes involved in various pathways associated with longevity and alleviation of AD-related symptoms in PIW-fed CL4176. RESULTS PIW mainly consists of tetracosanol, hexacosanol, octacosanol, and triacontanol; it could decrease the Aβ-induced paralysis rate from 86.87 to 66.97% (P < 0.01) and extend the life-span from 6.2 d to 7.8 d (P < 0.001) in CL4176 worms. Furthermore, PIW downregulated apl-1, a gene known to be associated with the levels of Aβ deposits in C. elegans. Additionally, our results showed that PIW modulated the expression of genes associated with longevity-related pathways such as heat shock response, anti-oxidative stress, and glutamine cysteine synthetase. CONCLUSION Our findings suggest that PIW may be a potential therapeutic agent for the prevention and treatment of AD. However, its effects on murine models and patients with AD need to be explored further.
Collapse
Affiliation(s)
- Xin Zhang
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| | - Chenjing Ma
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| | - Long Sun
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| | - Zhao He
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| | - Ying Feng
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China.
| | - Xian Li
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| | - Jin Gan
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| | - Xiaoming Chen
- The Key Laboratory of Cultivating and Utilization of Resource Insects of State Forestry Administration, Research Institute of Resource Insects, Chinese Academy of Forestry, Kunming, 650224, China
| |
Collapse
|
36
|
Olajide OJ, Suvanto ME, Chapman CA. Molecular mechanisms of neurodegeneration in the entorhinal cortex that underlie its selective vulnerability during the pathogenesis of Alzheimer's disease. Biol Open 2021; 10:bio056796. [PMID: 33495355 PMCID: PMC7860115 DOI: 10.1242/bio.056796] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The entorhinal cortex (EC) is a vital component of the medial temporal lobe, and its contributions to cognitive processes and memory formation are supported through its extensive interconnections with the hippocampal formation. During the pathogenesis of Alzheimer's disease (AD), many of the earliest degenerative changes are seen within the EC. Neurodegeneration in the EC and hippocampus during AD has been clearly linked to impairments in memory and cognitive function, and a growing body of evidence indicates that molecular and functional neurodegeneration within the EC may play a primary role in cognitive decline in the early phases of AD. Defining the mechanisms underlying molecular neurodegeneration in the EC is crucial to determining its contributions to the pathogenesis of AD. Surprisingly few studies have focused on understanding the mechanisms of molecular neurodegeneration and selective vulnerability within the EC. However, there have been advancements indicating that early dysregulation of cellular and molecular signaling pathways in the EC involve neurodegenerative cascades including oxidative stress, neuroinflammation, glia activation, stress kinases activation, and neuronal loss. Dysfunction within the EC can impact the function of the hippocampus, which relies on entorhinal inputs, and further degeneration within the hippocampus can compound this effect, leading to severe cognitive disruption. This review assesses the molecular and cellular mechanisms underlying early degeneration in the EC during AD. These mechanisms may underlie the selective vulnerability of neuronal subpopulations in this brain region to the disease development and contribute both directly and indirectly to cognitive loss.This paper has an associated Future Leader to Watch interview with the first author of the article.
Collapse
Affiliation(s)
- Olayemi Joseph Olajide
- Division of Neurobiology, Department of Anatomy, University of Ilorin, Ilorin, Nigeria, PMB 1515
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada H4B 1R6
| | - Marcus E Suvanto
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada H4B 1R6
| | - Clifton Andrew Chapman
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada H4B 1R6
| |
Collapse
|
37
|
Olajide OJ, Gbadamosi IT, Yawson EO, Arogundade T, Lewu FS, Ogunrinola KY, Adigun OO, Bamisi O, Lambe E, Arietarhire LO, Oluyomi OO, Idowu OK, Kareem R, Asogwa NT, Adeniyi PA. Hippocampal Degeneration and Behavioral Impairment During Alzheimer-Like Pathogenesis Involves Glutamate Excitotoxicity. J Mol Neurosci 2021; 71:1205-1220. [PMID: 33420680 DOI: 10.1007/s12031-020-01747-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022]
Abstract
The hallmarks of Alzheimer's disease (AD) pathology include senile plaques accumulation and neurofibrillary tangles, which is thought to underlie synaptic failure. Recent evidence however supports that synaptic failure in AD may instead be instigated by enhanced N-methyl-D-aspartate (NMDA) activity, via a reciprocal relationship between soluble amyloid-β (Aβ) accumulation and increased glutamate agonist. While previous studies have shown Aβ-mediated alterations to the glutamatergic system during AD, the underlying etiology of excitotoxic glutamate-induced changes has not been explored. Here, we investigated the acute effects of stereotaxic dentate gyrus (DG) glutamate injection on behavior and molecular expression of specific proteins and neurochemicals modulating hippocampal functions. Dependence of glutamate-mediated effects on NMDA receptor (NMDAR) hyperactivation was tested using NMDARs antagonist memantine. DG of Wistar rats (12-weeks-old) were bilaterally microinjected with glutamate (500 mM) with or without daily intraperitoneal (i.p.) memantine injection (20 mg/kg) for 14 days, while controls received either intrahippocampal/i.p. PBS or i.p. memantine. Behavioral characterization in open field and Y-maze revealed that glutamate evoked anxiogenic responses and perturbed spatial memory were inhibited by memantine. In glutamate-treated rats, increased NO expression was accompanied by marked reduction in profiles of glutathione-s-transferase and glutathione peroxidase. Similarly, glutamate-mediated increase in acetylcholinesterase expression corroborated downregulation of synaptophysin and PSD-95, coupled with initiation of reactive astrogliosis (GFAP). While neurofilament immunolocalization/immunoexpression was unperturbed, we found glutamate-mediated reduction in neurogenic markers Ki67 and PCNA immunoexpression, with a decrease in NR2B protein expression, whereas mGluR1 remains unchanged. In addition, increased expression of apoptotic regulatory proteins p53 and Bax was seen in glutamate infused rats, corroborating chromatolytic degeneration of granule neurons in the DG. Interestingly, memantine abrogated most of the degenerative changes associated with glutamate excitotoxicity in this study. Taken together, our findings causally link acute glutamate dyshomeostasis in the DG with development of AD-related behavioral impairment and molecular neurodegeneration.
Collapse
Affiliation(s)
- Olayemi Joseph Olajide
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria. .,Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada.
| | - Ismail Tayo Gbadamosi
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Central Research Laboratories Ltd, 132b University Road, Ilorin, Nigeria
| | - Emmanuel Olusola Yawson
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Department of Anatomy, Faculty of Basic Medical Sciences, Redeemer's University, Ede, Nigeria
| | - Tolulope Arogundade
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Department of Anatomy, Faculty of Basic Medical Sciences, Adeleke University, Ede, Nigeria
| | - Folashade Susan Lewu
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Kehinde Yomi Ogunrinola
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Department of Anatomy, School of Post-Basic Nursing, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Oluwaseun Olaniyi Adigun
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olawande Bamisi
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Department of Anatomy, Faculty of Basic Medical Sciences, Ekiti State University, Ado Ekiti, Nigeria
| | - Ezra Lambe
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Leviticus Ogbenevurinrin Arietarhire
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olushola Oladapo Oluyomi
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olumayowa Kolawole Idowu
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Rukayat Kareem
- Department of Anatomy, Division of Neurobiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Nnaemeka Tobechukwu Asogwa
- Central Research Laboratories Ltd, 132b University Road, Ilorin, Nigeria.,Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Ilorin, Nigeria
| | - Philip Adeyemi Adeniyi
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
38
|
Liu Y, Chen Z, Li B, Yao H, Zarka M, Welch J, Sachdev P, Bridge W, Braidy N. Supplementation with γ-glutamylcysteine (γ-GC) lessens oxidative stress, brain inflammation and amyloid pathology and improves spatial memory in a murine model of AD. Neurochem Int 2020; 144:104931. [PMID: 33276023 DOI: 10.1016/j.neuint.2020.104931] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/02/2020] [Accepted: 11/29/2020] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The accumulation of oxidative stress, neuroinflammation and abnormal aggregation of amyloid β-peptide (Aβ) have been shown to induce synaptic dysfunction and memory deficits in Alzheimer's disease (AD). Cellular depletion of the major endogenous antioxidant Glutathione (GSH) has been linked to cognitive decline and the development of AD pathology. Supplementation with γ-glutamylcysteine (γ-GC), the immediate precursor and the limiting substrate for GSH biosynthesis, can transiently augment cellular GSH levels by bypassing the regulation of GSH homeostasis. METHODS In the present study, we investigated the effect of dietary supplementation of γ-GC on oxidative stress and Aβ pathology in the brains of APP/PS1 mice. The APP/PS1 mice were fed γ-GC from 3 months of age with biomarkers of apoptosis and cell death, oxidative stress, neuroinflammation and Aβ load being assessed at 6 months of age. RESULTS Our data showed that supplementation with γ-GC lowered the levels of brain lipid peroxidation, protein carbonyls and apoptosis, increased both total GSH and the glutathione/glutathione disulphide (GSH/GSSG) ratio and replenished ATP and the activities of the antioxidant enzymes (superoxide dismutase (SOD), catalase, glutamine synthetase and glutathione peroxidase (GPX)), the latter being a key regulator of ferroptosis. Brain Aβ load was lower and acetylcholinesterase (AChE) activity was markedly improved compared to APP/PS1 mice fed a standard chow diet. Alteration in brain cytokine levels and matrix metalloproteinase enzymes MMP-2 and MMP-9 suggested that γ-GC may lower inflammation and enhance Aβ plaque clearance in vivo. Spatial memory was also improved by γ-GC as determined using the Morris water maze. CONCLUSION Our data collectively suggested that supplementation with γ-GC may represent a novel strategy for the treatment and/or prevention of cognitive impairment and neurodegeneration.
Collapse
Affiliation(s)
- Yue Liu
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Guangdong Mental Health Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zheng Chen
- School of Medicine, Huzhou University, Huzhou Central Hospital Huzhou, China
| | - Ben Li
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Hua Yao
- Institute of Life Sciences and Institute of Neuroscience, Wenzhou University, Wenzhou, China
| | - Martin Zarka
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Jeffrey Welch
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Wallace Bridge
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
39
|
Butterfield DA. Brain lipid peroxidation and alzheimer disease: Synergy between the Butterfield and Mattson laboratories. Ageing Res Rev 2020; 64:101049. [PMID: 32205035 PMCID: PMC7502429 DOI: 10.1016/j.arr.2020.101049] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/08/2020] [Accepted: 03/12/2020] [Indexed: 02/05/2023]
Abstract
Brains from persons with Alzheimer disease (AD) and its earlier stage, amnestic mild cognitive impairment (MCI), exhibit high levels of oxidative damage, including that to phospholipids. One type of oxidative damage is lipid peroxidation, the most important index of which is protein-bound 4-hydroxy-2-trans-nonenal (HNE). This highly reactive alkenal changes the conformations and lowers the activities of brain proteins to which HNE is covalently bound. Evidence exists that suggests that lipid peroxidation is the first type of oxidative damage associated with amyloid β-peptide (Aβ), a 38-42 amino acid peptide that is highly neurotoxic and critical to the pathophysiology of AD. The Butterfield laboratory is one of, if not the, first research group to show that Aβ42 oligomers led to lipid peroxidation and to demonstrate this modification in brains of subjects with AD and MCI. The Mattson laboratory, particularly when Dr. Mattson was a faculty member at the University of Kentucky, also showed evidence for lipid peroxidation associated with Aβ peptides, mostly in in vitro systems. Consequently, there is synergy between our two laboratories. Since this special tribute issue of Aging Research Reviews is dedicated to the career of Dr. Mattson, a review of some aspects of this synergy of lipid peroxidation and its relevance to AD, as well as the role of lipid peroxidation in the progression of this dementing disorder seems germane. Accordingly, this review outlines some of the individual and/or complementary research on lipid peroxidation related to AD published from our two laboratories either separately or jointly.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University Of Kentucky, Lexington, KY, 40506, United States.
| |
Collapse
|
40
|
Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases. ATMOSPHERE 2020. [DOI: 10.3390/atmos11101098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence links air pollution (AP) exposure to effects on the central nervous system structure and function. Particulate matter AP, especially the ultrafine (nanoparticle) components, can carry numerous metal and trace element contaminants that can reach the brain in utero and after birth. Excess brain exposure to either essential or non-essential elements can result in brain dyshomeostasis, which has been implicated in both neurodevelopmental disorders (NDDs; autism spectrum disorder, schizophrenia, and attention deficit hyperactivity disorder) and neurodegenerative diseases (NDGDs; Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis). This review summarizes the current understanding of the extent to which the inhalational or intranasal instillation of metals reproduces in vivo the shared features of NDDs and NDGDs, including enlarged lateral ventricles, alterations in myelination, glutamatergic dysfunction, neuronal cell death, inflammation, microglial activation, oxidative stress, mitochondrial dysfunction, altered social behaviors, cognitive dysfunction, and impulsivity. Although evidence is limited to date, neuronal cell death, oxidative stress, and mitochondrial dysfunction are reproduced by numerous metals. Understanding the specific contribution of metals/trace elements to this neurotoxicity can guide the development of more realistic animal exposure models of human AP exposure and consequently lead to a more meaningful approach to mechanistic studies, potential intervention strategies, and regulatory requirements.
Collapse
|
41
|
Dong Y, Brewer GJ. Global Metabolic Shifts in Age and Alzheimer's Disease Mouse Brains Pivot at NAD+/NADH Redox Sites. J Alzheimers Dis 2020; 71:119-140. [PMID: 31356210 PMCID: PMC6839468 DOI: 10.3233/jad-190408] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Age and Alzheimer’s disease (AD) share some common features such as cognitive impairments, memory loss, metabolic disturbances, bioenergetic deficits, and inflammation. Yet little is known on how systematic shifts in metabolic networks depend on age and AD. In this work, we investigated the global metabolomic alterations in non-transgenic (NTg) and triple-transgenic (3xTg-AD) mouse brain hippocampus as a function of age by using untargeted Ultrahigh Performance Liquid Chromatography-tandem Mass Spectroscopy (UPLC-MS/MS). We observed common metabolic patterns with aging in both NTg and 3xTg-AD brains involved in energy-generating pathways, fatty acids oxidation, glutamate, and sphingolipid metabolism. We found age-related downregulation of metabolites from reactions in glycolysis that consumed ATP and in the TCA cycle, especially at NAD+/NADH-dependent redox sites, where age- and AD-associated limitations in the free NADH may alter reactions. Conversely, metabolites increased in glycolytic reactions in which ATP is produced. With age, inputs to the TCA cycle were increased including fatty acid β-oxidation and glutamine. Overall age- and AD-related changes were > 2-fold when comparing the declines of upstream metabolites of NAD+/NADH-dependent reactions to the increases of downstream metabolites (p = 10-5, n = 8 redox reactions). Inflammatory metabolites such as ceramides and sphingosine-1-phosphate also increased with age. Age-related decreases in glutamate, GABA, and sphingolipid were seen which worsened with AD genetic load in 3xTg-AD brains, possibly contributing to synaptic, learning- and memory-related deficits. The data support the novel hypothesis that age- and AD-associated metabolic shifts respond to NAD(P)+/NAD(P)H redox-dependent reactions, which may contribute to decreased energetic capacity.
Collapse
Affiliation(s)
- Yue Dong
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Gregory J Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.,MIND Institute, Center for Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
| |
Collapse
|
42
|
Rathnayake AU, Abuine R, Kim YJ, Byun HG. Anti-Alzheimer's Materials Isolated from Marine Bio-resources: A Review. Curr Alzheimer Res 2020; 16:895-906. [PMID: 31647396 DOI: 10.2174/1567205016666191024144044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 08/12/2019] [Accepted: 09/08/2019] [Indexed: 12/15/2022]
Abstract
The most common type of dementia found in the elderly population is Alzheimer's disease. The disease not only impacts the patients and their families but also the society therefore, the main focus of researchers is to search new bioactive materials for treating AD. The marine environment is a rich source of functional ingredients and to date, we can find sufficient research relating to anti- Alzheimer's compounds isolated from marine environment. Therefore, this review focuses on the anti- Alzheimer's material from marine bio-resources and then expounds on the anti-Alzheimer's compounds from marine seaweed, marine animal and marine microorganisms. Moreover, because of the complexity of the disease, different hypothesizes have been elaborated and active compounds have been isolated to inhibit different stages of pathophysiological mechanisms. Sulfated polysaccharides, glycoprotein, and enzymatic hydrolysates from marine seaweeds, peptides, dietary omega-3 polyunsaturated fatty acids and skeletal polysaccharide from marine animals and secondary metabolites from marine microorganism are summarized in this review under the anti-Alzheimer's compounds from the marine.
Collapse
Affiliation(s)
| | - Racheal Abuine
- Department of Marine Biotechnology, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Yong-Jae Kim
- Department of Marine Biotechnology, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Hee-Guk Byun
- Department of Marine Biotechnology, Gangneung-Wonju National University, Gangneung 25457, Korea
| |
Collapse
|
43
|
Martínez-Cué C, Rueda N. Signalling Pathways Implicated in Alzheimer's Disease Neurodegeneration in Individuals with and without Down Syndrome. Int J Mol Sci 2020; 21:E6906. [PMID: 32962300 PMCID: PMC7555886 DOI: 10.3390/ijms21186906] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer's disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain;
| | | |
Collapse
|
44
|
Kaniakova M, Nepovimova E, Kleteckova L, Skrenkova K, Holubova K, Chrienova Z, Hepnarova V, Kucera T, Kobrlova T, Vales K, Korabecny J, Soukup O, Horak M. Combination of Memantine and 6-Chlorotacrine as Novel Multi-Target Compound against Alzheimer's Disease. Curr Alzheimer Res 2020; 16:821-833. [PMID: 30819076 DOI: 10.2174/1567205016666190228122218] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/04/2019] [Accepted: 01/31/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia in the elderly. It is characterized as a multi-factorial disorder with a prevalent genetic component. Due to the unknown etiology, current treatment based on acetylcholinesterase (AChE) inhibitors and N-methyl-D-aspartate receptors (NMDAR) antagonist is effective only temporary. It seems that curative treatment will necessarily be complex due to the multifactorial nature of the disease. In this context, the so-called "multi-targeting" approach has been established. OBJECTIVES The aim of this study was to develop a multi-target-directed ligand (MTDL) combining the support for the cholinergic system by inhibition of AChE and at the same time ameliorating the burden caused by glutamate excitotoxicity mediated by the NMDAR receptors. METHODS We have applied common approaches of organic chemistry to prepare a hybrid of 6-chlorotacrine and memantine. Then, we investigated its blocking ability towards AChE and NMDRS in vitro, as well as its neuroprotective efficacy in vivo in the model of NMDA-induced lessions. We also studied cytotoxic potential of the compound and predicted the ability to cross the blood-brain barrier. RESULTS A novel molecule formed by combination of 6-chlorotacrine and memantine proved to be a promising multipotent hybrid capable of blocking the action of AChE as well as NMDARs. The presented hybrid surpassed the AChE inhibitory activity of the parent compound 6-Cl-THA twofold. According to results it has been revealed that our novel hybrid blocks NMDARs in the same manner as memantine, potently inhibits AChE and is predicted to cross the blood-brain barrier via passive diffusion. Finally, the MTDL design strategy was indicated by in vivo results which showed that the novel 6-Cl-THA-memantine hybrid displayed a quantitatively better neuroprotective effect than the parent compound memantine. CONCLUSION We conclude that the combination of two pharmacophores with a synergistic mechanism of action into a single molecule offers great potential for the treatment of CNS disorders associated with cognitive decline and/or excitotoxicity mediated by NMDARs.
Collapse
Affiliation(s)
- Martina Kaniakova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague 4, Czech Republic.,Institute of Physiology of the Czech Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Lenka Kleteckova
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Kristyna Skrenkova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Kristina Holubova
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Zofia Chrienova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Vendula Hepnarova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, Hradec Kralove 500 05, Czech Republic
| | - Tomas Kucera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, Hradec Kralove 500 05, Czech Republic
| | - Tereza Kobrlova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, Hradec Kralove 500 05, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Karel Vales
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Martin Horak
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague 4, Czech Republic.,Institute of Physiology of the Czech Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4, Czech Republic
| |
Collapse
|
45
|
Swimming exercise improves cognitive and behavioral disorders in male NMRI mice with sporadic Alzheimer-like disease. Physiol Behav 2020; 223:113003. [DOI: 10.1016/j.physbeh.2020.113003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/16/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
|
46
|
Tobore TO. On the Etiopathogenesis and Pathophysiology of Alzheimer's Disease: A Comprehensive Theoretical Review. J Alzheimers Dis 2020; 68:417-437. [PMID: 30775973 DOI: 10.3233/jad-181052] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimers' disease (AD) is the most common cause of dementia, with an estimated 5 million new cases occurring annually. Among the elderly, AD shortens life expectancy, results in disability, decreases quality of life, and ultimately, leads to institutionalization. Despite extensive research in the last few decades, its heterogeneous pathophysiology and etiopathogenesis have made it difficult to develop an effective treatment and prevention strategy. Aging is the biggest risk factor for AD and evidence suggest that the total number of older people in the population is going to increase astronomically in the next decades. Also, there is evidence that air pollution and increasing income inequality may result in higher incidence and prevalence of AD. This makes the need for a comprehensive understanding of the etiopathogenesis and pathophysiology of the disease extremely critical. In this paper, a quintuple framework of thyroid dysfunction, vitamin D deficiency, sex hormones, and mitochondria dysfunction and oxidative stress are used to provide a comprehensive description of AD etiopathogenesis and pathophysiology. The individual role of each factor, their synergistic and genetic interactions, as well as the limitations of the framework are discussed.
Collapse
|
47
|
Yeung JHY, Calvo-Flores Guzmán B, Palpagama TH, Ethiraj J, Zhai Y, Tate WP, Peppercorn K, Waldvogel HJ, Faull RLM, Kwakowsky A. Amyloid-beta 1-42 induced glutamatergic receptor and transporter expression changes in the mouse hippocampus. J Neurochem 2020; 155:62-80. [PMID: 32491248 DOI: 10.1111/jnc.15099] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the leading type of dementia worldwide. With an increasing burden of an aging population coupled with the lack of any foreseeable cure, AD warrants the current intense research effort on the toxic effects of an increased concentration of beta-amyloid (Aβ) in the brain. Glutamate is the main excitatory brain neurotransmitter and it plays an essential role in the function and health of neurons and neuronal excitability. While previous studies have shown alterations in expression of glutamatergic signaling components in AD, the underlying mechanisms of these changes are not well understood. This is the first comprehensive anatomical study to characterize the subregion- and cell layer-specific long-term effect of Aβ1-42 on the expression of specific glutamate receptors and transporters in the mouse hippocampus, using immunohistochemistry with confocal microscopy. Outcomes are examined 30 days after Aβ1-42 stereotactic injection in aged male C57BL/6 mice. We report significant decreases in density of the glutamate receptor subunit GluA1 and the vesicular glutamate transporter (VGluT) 1 in the conus ammonis 1 region of the hippocampus in the Aβ1-42 injected mice compared with artificial cerebrospinal fluid injected and naïve controls, notably in the stratum oriens and stratum radiatum. GluA1 subunit density also decreased within the dentate gyrus dorsal stratum moleculare in Aβ1-42 injected mice compared with artificial cerebrospinal fluid injected controls. These changes are consistent with findings previously reported in the human AD hippocampus. By contrast, glutamate receptor subunits GluA2, GluN1, GluN2A, and VGluT2 showed no changes in expression. These findings indicate that Aβ1-42 induces brain region and layer specific expression changes of the glutamatergic receptors and transporters, suggesting complex and spatial vulnerability of this pathway during development of AD neuropathology. Read the Editorial Highlight for this article on page 7. Cover Image for this issue: https://doi.org/10.1111/jnc.14763.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jayarjun Ethiraj
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ying Zhai
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Warren P Tate
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
48
|
Findley CA, Bartke A, Hascup KN, Hascup ER. Amyloid Beta-Related Alterations to Glutamate Signaling Dynamics During Alzheimer's Disease Progression. ASN Neuro 2020; 11:1759091419855541. [PMID: 31213067 PMCID: PMC6582288 DOI: 10.1177/1759091419855541] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) ranks sixth on the Centers for Disease Control and Prevention Top 10 Leading Causes of Death list for 2016, and the Alzheimer’s Association attributes 60% to 80% of dementia cases as AD related. AD pathology hallmarks include accumulation of senile plaques and neurofibrillary tangles; however, evidence supports that soluble amyloid beta (Aβ), rather than insoluble plaques, may instigate synaptic failure. Soluble Aβ accumulation results in depression of long-term potentiation leading to cognitive deficits commonly characterized in AD. The mechanisms through which Aβ incites cognitive decline have been extensively explored, with a growing body of evidence pointing to modulation of the glutamatergic system. The period of glutamatergic hypoactivation observed alongside long-term potentiation depression and cognitive deficits in later disease stages may be the consequence of a preceding period of increased glutamatergic activity. This review will explore the Aβ-related changes to the tripartite glutamate synapse resulting in altered cell signaling throughout disease progression, ultimately culminating in oxidative stress, synaptic dysfunction, and neuronal loss.
Collapse
Affiliation(s)
- Caleigh A Findley
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- 3 Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,4 Department of Molecular Biology, Microbiology & Biochemistry, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
49
|
Prasansuklab A, Brimson JM, Tencomnao T. Potential Thai medicinal plants for neurodegenerative diseases: A review focusing on the anti-glutamate toxicity effect. J Tradit Complement Med 2020; 10:301-308. [PMID: 32670825 PMCID: PMC7340876 DOI: 10.1016/j.jtcme.2020.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases (NDD) are a range of debilitating conditions of the brain involving progressive loss of neurons, many of which are still currently incurable despite enormous efforts on drug discovery and development in the past decade. As NDD is closely linked to old age, the rapid worldwide growth in the aging population contributes to an increasing number of people with one of these incurable diseases and therefore it is considered a significant global health issue. There is an urgent need for novel effective treatments for NDD, and many new research strategies are centered on traditional medicine as an alternative or complementary solution. Several previous findings have suggested that glutamate toxicity drives neurodegeneration in many NDD, and the medicinal plants with anti-glutamate toxicity properties can be potentially used for their treatment. In order to obtain data relating to natural products against glutamate toxicity, six candidate plant species of Thailand were identified. Studies utilizing these herbs were searched for using the herb name (Latin and common names) along with the term "glutamate" in the following databases across all available years: PubMed, Scopus, and Google Scholar. This review emphasizes the importance of glutamate toxicity in NDD and summarizes individual plants and their active constituents with the mechanism of action against glutamate toxicity-mediated neuronal cell death that could be a promising resource for future NDD therapy. TAXONOMY CLASSIFICATION BY EVISE Alzheimer's disease, Neurodegenerative diseases, Cell culture, Molecular Biology, Traditional herbal medicine, Oxidative stress, Glutamate neurotransmitter.
Collapse
Affiliation(s)
- Anchalee Prasansuklab
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - James M. Brimson
- Age-Related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tewin Tencomnao
- Age-Related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
50
|
Jeon SG, Yoo A, Chun DW, Hong SB, Chung H, Kim JI, Moon M. The Critical Role of Nurr1 as a Mediator and Therapeutic Target in Alzheimer's Disease-related Pathogenesis. Aging Dis 2020; 11:705-724. [PMID: 32489714 PMCID: PMC7220289 DOI: 10.14336/ad.2019.0718] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/18/2019] [Indexed: 01/16/2023] Open
Abstract
Several studies have revealed that the transcription factor nuclear receptor related 1 (Nurr1) plays several roles not only in the regulation of gene expression related to dopamine synthesis, but also in alternative splicing, and miRNA targeting. Moreover, it regulates cognitive functions and protects against inflammation-induced neuronal death. In particular, the role of Nurr1 in the pathogenesis of Parkinson's disease (PD) has been well investigated; for example, it has been shown that it restores behavioral and histological impairments in PD models. Although many studies have evaluated the connection between Nurr1 and PD pathogenesis, the role of Nurr1 in Alzheimer's disease (AD) remain to be studied. There have been several studies describing Nurr1 protein expression in the AD brain. However, only a few studies have examined the role of Nurr1 in the context of AD. Therefore, in this review, we highlight the overall effects of Nurr1 under the neuropathologic conditions related to AD. Furthermore, we suggest the possibility of using Nurr1 as a therapeutic target for AD or other neurodegenerative disorders.
Collapse
Affiliation(s)
- Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Anji Yoo
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Dong Wook Chun
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Hyunju Chung
- Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Jin-il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| |
Collapse
|