1
|
Wang Y, Li P, Liang Y, Wang D. ANO6 Targets TMEM30A to Regulate Endoplasmic Reticulum Stress-Induced Lipid Peroxidation and Ferroptosis in Alzheimer's Cells. Cell Biochem Biophys 2025:10.1007/s12013-025-01748-9. [PMID: 40221538 DOI: 10.1007/s12013-025-01748-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, and the role of ANO6 in its progression remains largely unexplored. GSE118553 database was analyzed for ANO6 expression in AD. A total of 1 μmol/L Aβ1-42 treated SH-SY5Y cells were constructed as a cell model of AD. qRT-PCR and ELISA were used to detect the expression of ANO6, GPX4, ATF6, GRP78, IREIα expression and lipid peroxidation level. Endoplasmic reticulum(ER) stress was induced by using clindamycin and lipid peroxidation indicators were detected. ANO6 was concurrently regulated in ER stress induced by clindamycin treatment. The STRING-DB database was utilized to predict potential target molecules of ANO6, while Western blot analysis was conducted to detect the expression levels of TMEM30A and evaluate the impact of ANO6-targeted TMEM30A on the protein levels within the PERK-eIF2α-ATF4-CHOP pathway. ANO6 was highly expressed in AD model, Aβ1-42 induced ANO6 enrichment in SH-SY5Y cells. ANO6 interference increased the proliferation level of AD model cells, decreased the levels of GPX4, an indicator of ferroptosis, and lipid peroxidation, and down-regulated the expression of the ER stress-related proteins ATF6, GRP78, and IREIα. Clotrimazole-induced ER stress in AD model cells showed elevated expression of ANO6. ANO6 could target and inhibit TMEM30A to affect PERK-eIF2α-ATF4-CHOP pathway activity, regulate ER stress-dependent ferroptosis, and reduce neuronal loss injury. ANO6 can target inhibition of TMEM30A affecting PERK- IF2α- ATF4- CHOP pathway activity, modulate ER stress-dependent ferroptosis-induced AD progression to reduce neuronal loss injury.
Collapse
Affiliation(s)
- Ying Wang
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China.
| | - Penghui Li
- College of Basic Medicine, Qiqihar Medical University, Qiqihar, China
| | - Yonghan Liang
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Dandan Wang
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
2
|
Müller E, Enzlein T, Niemeyer D, von Ammon L, Stumpo K, Biber K, Klein C, Hopf C. Exploring the Aβ Plaque Microenvironment in Alzheimer's Disease Model Mice by Multimodal Lipid-Protein-Histology Imaging on a Benchtop Mass Spectrometer. Pharmaceuticals (Basel) 2025; 18:252. [PMID: 40006065 PMCID: PMC11860057 DOI: 10.3390/ph18020252] [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: 11/27/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Amyloid-β (Aβ) plaque deposits in the brain are a hallmark of Alzheimer's disease (AD) neuropathology. Plaques consist of complex mixtures of peptides like Aβ1-42 and characteristic lipids such as gangliosides, and they are targeted by reactive microglia and astrocytes. Background: In pharmaceutical research and development, it is a formidable challenge to contextualize the different biomolecular classes and cell types of the Aβ plaque microenvironment in a coherent experimental workflow on a single tissue section and on a benchtop imaging reader. Methods: Here, we developed a workflow that combines lipid MALDI mass spectrometry imaging using a vacuum-stable matrix with histopathology stains and with the MALDI HiPLEX immunohistochemistry of plaques and multiple protein markers on a benchtop imaging mass spectrometer. The three data layers consisting of lipids, protein markers, and histology could be co-registered and evaluated together. Results: Multimodal data analysis suggested the extensive co-localization of Aβ plaques with the peptide precursor protein, with a defined subset of lipids and with reactive glia cells on a single brain section in APPPS1 mice. Plaque-associated lipids like ganglioside GM2 and phosphatidylinositol PI38:4 isoforms were readily identified using the tandem MS capabilities of the mass spectrometer. Conclusions: Altogether, our data suggests that complex pathology involving multiple lipids, proteins and cell types can be interrogated by this spatial multiomics workflow on a user-friendly benchtop mass spectrometer.
Collapse
Affiliation(s)
- Elisabeth Müller
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (E.M.); (T.E.); (L.v.A.)
- Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Thomas Enzlein
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (E.M.); (T.E.); (L.v.A.)
| | | | - Livia von Ammon
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (E.M.); (T.E.); (L.v.A.)
| | | | - Knut Biber
- AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany
| | - Corinna Klein
- AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (E.M.); (T.E.); (L.v.A.)
- Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
- Mannheim Center for Translational Neurosciences (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
3
|
Zhang T, Yang J, Xu H, Cao Y, Du X, Guo L, Liang B, Su L, Chai L, Yuan Q, Hu L. Acorus tatarinowii alleviates D-galactose-induced Alzheimer's-like disease cognitive impairment and Aβ-induced pericytes dysfunction in mice. Brain Res 2025; 1847:149312. [PMID: 39515743 DOI: 10.1016/j.brainres.2024.149312] [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: 06/27/2024] [Revised: 09/26/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Pericytes regulate cerebral blood flow (CBF) and excess amyloid in the brain. Pericyte dysfunction may contribute to the pathology of Alzheimer's disease (AD). Acorus tatarinowii (AT), a Chinese medicine commonly used to treat AD, protects the central nervous system. However, whether AT can regulate pericyte function and ameliorate cognitive dysfunction remains unclear. We employed a novel target recognition assay, quantitative measurement of CBF, hematoxylin and eosin staining, immunofluorescence staining, and Western blot to investigate the role of AT in improving cognitive function in patients with AD. Additionally, we investigated the therapeutic potential of β-Asarone, the primary active compound in AT, for treating AD by modulating pericyte function using transmission electron microscopy, silver staining, electrical impedance, and other methodologies. The results revealed that administration of AT effectively alleviated the cognitive impairments induced by D-galactose in mice, as evidenced by enhanced CBF, improved histological characteristics of damaged brain tissue cells, increased expression of platelet-derived growth factor-β (PDGF-β), decreased Aβ accumulation via enhanced lipoprotein receptor-related protein 1 (LRP1), and reduced beta-site APP-cleaving enzyme 1 (BACE1). β-Asarone treatment mitigated ROS release and BACE1 expression while elevating the cell index in Aβ1-40 injured mouse brain vascular pericytes (MBVP). These findings suggest that AT has the potential to enhance CBF and mitigate pericellular dysfunction, thereby ameliorating Aβ deposition in the brain and improving cognitive impairment in patients with AD.
Collapse
Affiliation(s)
- Tong Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Juan Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haiying Xu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yushuang Cao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyuan Du
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lichen Guo
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bing Liang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Linlin Su
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lijuan Chai
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qing Yuan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Limin Hu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
4
|
Wang N, Cui J, Sun Z, Chen F, He X. Exploring the protective effect and molecular mechanism of betulin in Alzheimer's disease based on network pharmacology, molecular docking and experimental validation. Mol Med Rep 2024; 30:232. [PMID: 39392030 PMCID: PMC11529172 DOI: 10.3892/mmr.2024.13356] [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: 05/31/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that impairs learning and memory, with high rates of mortality. Birch bark has been traditionally used in the treatment of various skin ailments. Betulin (BT) is a key compound of birch bark that exhibits diverse pharmacological benefits and therapeutic potential in AD. However, the therapeutic effects and molecular mechanisms of BT in AD remain unclear. The present study aimed to predict the potential therapeutic targets of BT in the treatment of AD, and to determine the specific underlying molecular mechanisms through network pharmacology analysis and experimental validation. PharmMapper was used to predict the target genes of BT, and four disease databases were searched to screen for AD targets. The intersection targets were identified using the jveen website. Drug‑disease target protein‑protein interaction networks and hub genes were obtained and visualized using the Search Tool for the Retrieval of Interacting Genes/Proteins database and Cytoscape. The Database for Annotation, Visualization and Integrated Discovery was used for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and AutoDock was used for molecular docking analysis of BT and hub genes. Subsequently, the network‑predicted mechanisms of BT in AD were verified in vitro. A total of 495 BT and 1,386 AD targets were identified, and 120 were identified as potential targets of BT in the treatment of AD. The results of the molecular docking analysis revealed a strong binding affinity between BT and the hub genes. In addition, enrichment analyses of GO and KEGG pathways indicated that the neuroprotective effects of BT mainly involved the 'PI3K‑Akt signaling pathway'. The results of in vitro experiments demonstrated that pretreatment with BT for 2 h may ameliorate formaldehyde (FA)‑induced cytotoxicity and morphological changes in HT22 cells, and decrease FA‑induced Tau hyperphosphorylation and reactive oxygen species levels. Furthermore, the PI3K/AKT signaling pathway was activated and the expression levels of downstream proteins, namely GSK3β, Bcl‑2 and Bax, were modified following pre‑treatment with BT. Overall, the results of network pharmacology and in vitro analyses revealed that BT may reduce FA‑induced AD‑like pathology by modulating the PI3K/AKT signaling pathway, highlighting it as a potential multi‑target drug for the treatment of AD.
Collapse
Affiliation(s)
- Na Wang
- Laboratory of Brain and Cognitive Science, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671003, P.R. China
| | - Jiali Cui
- Yunnan Institute of Materia Medica, Yunnan Province Company Key Laboratory for TCM and Ethnic Drug of New Drug Creation, Kunming, Yunnan 650111, P.R. China
| | - Ziteng Sun
- Laboratory of Brain and Cognitive Science, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671003, P.R. China
| | - Fan Chen
- Department of Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi, Jiangsu 214151, P.R. China
- Laboratory of Heart Disease Mechanism and Translational Research, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Xiaping He
- Laboratory of Brain and Cognitive Science, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671003, P.R. China
| |
Collapse
|
5
|
Jin Y, Du Q, Song M, Kang R, Zhou J, Zhang H, Ding Y. Amyloid-β-targeting immunotherapies for Alzheimer's disease. J Control Release 2024; 375:346-365. [PMID: 39271059 DOI: 10.1016/j.jconrel.2024.09.012] [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: 06/17/2024] [Revised: 07/24/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
Recent advances in clinical passive immunotherapy have provided compelling evidence that eliminating amyloid-β (Aβ) slows cognitive decline in Alzheimer's disease (AD). However, the modest benefits and side effects observed in clinical trials indicate that current immunotherapy therapy is not a panacea, highlighting the need for a deeper understanding of AD mechanisms and the significance of early intervention through optimized immunotherapy or immunoprevention. This review focuses on the centrality of Aβ pathology in AD and summarizes recent clinical progress in passive and active immunotherapies targeting Aβ, discussing their lessons and failures to inform future anti-Aβ biotherapeutics design. Various delivery strategies to optimize Aβ-targeting immunotherapies are outlined, highlighting their benefits and drawbacks in overcoming challenges such as poor stability and limited tissue accessibility of anti-Aβ biotherapeutics. Additionally, the perspectives and challenges of immunotherapy and immunoprevention targeting Aβ are concluded in the end, aiming to guide the development of next-generation anti-Aβ immunotherapeutic agents towards improved efficacy and safety.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qiaofei Du
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Mingjie Song
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixin Kang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jianping Zhou
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Huaqing Zhang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yang Ding
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
Xia Y, Wang X, Lin S, Dong TTX, Tsim KWK. Berberine and palmatine, acting as allosteric potential ligands of α7 nAChR, synergistically regulate inflammation and phagocytosis of microglial cells. FASEB J 2024; 38:e70094. [PMID: 39373933 DOI: 10.1096/fj.202302538rrrr] [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: 12/08/2023] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Berberine and palmatine are isoquinoline quaternary alkaloids derived from Chinese medicinal herbs. These alkaloids have shown promising synergy in inhibiting acetylcholinesterase (AChE), indicating their potential in treating Alzheimer's disease (AD). Besides, the anti-inflammatory effects of berberine and palmatine have been widely reported, although the underlying mechanism remains unclear. Here, we found that berberine and palmatine could induce calcium ion (Ca2+) influx via activating α7 nicotinic acetylcholine receptor (α7 nAChR) in cultured microglial cells, possibly serving as its allosteric potential ligands. Furthermore, we examined the synergistic anti-inflammatory effects of berberine and palmatine in the LPS-induced microglia, that significantly suppressed the production of TNF-α and iNOS. Notably, this suppression was reversed by co-treatment with a selective antagonist of α7 nAChR. Moreover, the alkaloid-induced microglial phagocytosis was shown to be mediated by the induction of Ca2+ influx through α7 nAChR and subsequent CaMKII-Rac1-dependent pathway. Additionally, the combination of berberine and palmatine, at low concentration, protected against the LPS-induced endoplasmic reticulum stress and mitochondrial dysfunction in microglia. These findings indicate the potential of berberine and palmatine, either individually or in combination, in contributing to anti-AD drug development, which provide valuable insights into the mechanisms by which natural products, such as plant alkaloids, exert their anti-AD effects.
Collapse
Affiliation(s)
- Yingjie Xia
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China
| | - Xiaoyang Wang
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China
| | - Shengying Lin
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China
| | - Tina T X Dong
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China
| | - Karl W K Tsim
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China
| |
Collapse
|
7
|
Li P, Bi X, Xu D, Meng Y, Xia Y, Cai J, Shen Y, Wang J, Chen J, Yin L, Wang B, Wu D, Li K. Activation of the hypoxia response in the aging cerebrovasculature protects males against cognitive impairment. Aging Cell 2024; 23:e14264. [PMID: 38953594 PMCID: PMC11464119 DOI: 10.1111/acel.14264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with a distinct sex bias. Age-related vascular alterations, a hallmark of AD onset and progression, are consistently associated with sexual dimorphism. Here, we conducted an integrative meta-analysis of 335,803 single-nucleus transcriptomes and 667 bulk transcriptomes from the vascular system in AD and normal aging to address the underlying sex-dependent vascular aging in AD. All vascular cell types in male AD patients exhibited an activated hypoxia response and downstream signaling pathways including angiogenesis. The female AD vasculature is characterized by increased antigen presentation and decreased angiogenesis. We further confirmed that these sex-biased alterations in the cerebral vascular emerged and were primarily determined in the early stages of AD. Sex-stratified analysis of normal vascular aging revealed that angiogenesis and various stress-response genes were downregulated concurrently with female aging. Conversely, the hypoxia response increased steadily in males upon aging. An investigation of upstream driver transcription factors (TFs) revealed that altered communication between estrogen receptor alpha (ESR1) and hypoxia induced factors during menopause contributes to the inhibition of angiogenesis during normal female vascular aging. Additionally, inhibition of CREB1, a TF that targets estrogen, is also related to female AD. Overall, our study revealed a distinct cerebral vascular profile in females and males, and revealed novel targets for precision medicine therapy for AD.
Collapse
Affiliation(s)
- Peihu Li
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Xiaoman Bi
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Dahua Xu
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | | | - Yucheng Xia
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Jiale Cai
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Yutong Shen
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Jiaqi Wang
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Jiazhu Chen
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Lamei Yin
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Bo Wang
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Deng Wu
- School of Life Sciences, Faculty of ScienceThe Chinese University of Hong KongHong KongChina
| | - Kongning Li
- College of Biomedical Information and EngineeringHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| |
Collapse
|
8
|
Davoudi F, Shadjou N, Darroudi M. Interactions of memantine and rivastigmine with graphene oxide nanocarrier and beta-amyloid protein using molecular docking and in-silico methods. Heliyon 2024; 10:e37702. [PMID: 39309765 PMCID: PMC11416293 DOI: 10.1016/j.heliyon.2024.e37702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is characterized by the accumulation of beta-amyloid plaques and neurofibrillary tangles. Effective therapeutic strategies involve inhibiting the formation of beta-amyloid aggregates and destabilizing existing ones. A significant challenge in current treatments is the inability of therapeutic agents to cross the blood-brain barrier, a limitation addressed by employing drug nanocarriers. This study investigates the interactions between memantine, rivastigmine, beta-amyloid structures, and graphene oxide nanocarriers using molecular docking and in silico methods. The goal is to enhance drug development through cost-effective and efficient computational techniques. Results indicate that the binding energies for memantine-beta-amyloid and rivastigmine-beta-amyloid complexes are -9.03 kcal/mol and -7.81 kcal/mol, respectively, suggesting superior stability for the memantine-beta-amyloid complex. The electrostatic energies are -1.91 kcal/mol for memantine and -0.81 kcal/mol for rivastigmine, further supporting the greater stability of the memantine complex. Additionally, memantine's interaction with graphene oxide results in more negative adsorption energy (-92.47 kJ/mol) compared to rivastigmine (-86.36 kJ/mol), indicating a stronger binding affinity. The charge transfer (Q) values are -0.41 kJ/mol for memantine and -0.33 kJ/mol for rivastigmine. The negative enthalpy (ΔH) of -85.71 kJ/mol and Gibbs free energy (ΔG) of -41.52 kJ/mol for the memantine-graphene oxide interaction suggest a spontaneous process. Both memantine and rivastigmine display similar electronic properties, but memantine shows a more effective interaction with graphene oxide, likely due to its amine functional group and spatial configuration. The adsorption energy analysis confirms that memantine forms a more stable complex with graphene oxide than rivastigmine.
Collapse
Affiliation(s)
- Fateme Davoudi
- Department of Nanotechnology, Faculty of Chemistry, Urmia University, Urmia, Iran
- Institute of Nanotechnology, Urmia University, Urmia, Iran
| | - Nasrin Shadjou
- Department of Nanotechnology, Faculty of Chemistry, Urmia University, Urmia, Iran
| | - Mahdieh Darroudi
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
9
|
Shen Y, Liu F, Zhang M. Therapeutic potential of plant-derived natural compounds in Alzheimer's disease: Targeting microglia-mediated neuroinflammation. Biomed Pharmacother 2024; 178:117235. [PMID: 39094545 DOI: 10.1016/j.biopha.2024.117235] [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: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Microglia are resident immune cells of the central nervous system (CNS) with roles in sensing, housekeeping, and defense. Exploring the role of microglia in the occurrence and development of Alzheimer's disease (AD) and the possible therapeutic mechanism of plant-derived natural compounds (PDNCs) that regulate microglia-associated neuroinflammation may potentially help in elucidating the pathogenesis of AD and provide novel insights for its treatment. This review explores the role of abnormal microglial activation and its dominant neuroinflammatory response, as well as the activation of their target receptors and signaling pathways in AD pathogenesis. Additionally, we report an update on the potential pharmacological mechanisms of multiple PDNCs in modulating microglia-associated neuroinflammation in AD treatment. Dysregulated activation of microglial receptors and their downstream pathways impaired immune homeostasis in animal models of AD. Multiple signaling pathways, such as mitogen-activated protein kinase (MAPK), nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and Toll-like receptors, play important roles in microglial activation and can exacerbate microglia-mediated neuroinflammation. PDNCs, such as magnolol, stigmasterol, matrine, naringenin, naringin, and resveratrol, can delay the progression of AD by inhibiting the proinflammatory receptors of microglia, activating its anti-inflammatory receptors, regulating the receptors related to β-amyloid (Aβ) clearance, reversing immune dysregulation, and maintaining the immune homeostasis of microglial downstream pathways. This review summarizes the mechanisms by which microglia cause chronic inflammation in AD and evaluates the beneficial effects of PDNCs on immune regulation in AD by regulating microglial receptors and their downstream pathways.
Collapse
Affiliation(s)
- Yanyan Shen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| | - Fang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, China
| | - Mingjie Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
10
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [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: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
11
|
Zhang J, Chen Y, Zhao Y, Wang P, Ding H, Liu C, Lyu J, Le W. Terahertz Irradiation Improves Cognitive Impairments and Attenuates Alzheimer's Neuropathology in the APP SWE/PS1 DE9 Mouse: A Novel Therapeutic Intervention for Alzheimer's Disease. Neurosci Bull 2024; 40:857-871. [PMID: 37971654 PMCID: PMC11250709 DOI: 10.1007/s12264-023-01145-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the deposition of amyloid-β (Aβ), neurofibrillary tangles, neuroinflammation, and neurodegeneration in the brain. In recent years, considering the unsatisfied benefits of pharmacological therapies, non-pharmacological therapy has become a research hotspot for AD intervention. Terahertz (THz) waves with a range between microwave and infrared regions in the electromagnetic spectrum and high permeability to a wide range of materials have great potential in the bioengineering field. However, its biological impacts on the central nervous system, under either physiological or pathological conditions, are poorly investigated. In this study, we first measured the 0.14 THz waves penetration across the skull of a C57BL/6 mouse and found the percentage of THz penetration to be ~70%, guaranteeing that THz waves can reach the relevant brain regions. We then exposed the APPSWE/PS1DE9 mouse model of AD to repeated low-frequency THz waves on the head. We demonstrated that THz waves treatment significantly improved the cognitive impairment and alleviated AD neuropathology including Aβ deposition and tau hyperphosphorylation in the AD mice. Moreover, THz waves treatment effectively attenuated mitochondrial impairment, neuroinflammation, and neuronal loss in the AD mouse brain. Our findings reveal previously unappreciated beneficial effects of THz waves treatment in AD and suggest that THz waves may have the potential to be used as a novel therapeutic intervention for this devastating disease.
Collapse
Affiliation(s)
- Jun Zhang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yarui Zhao
- School of Physics, Dalian University of Technology, Dalian, 116024, China
| | - Panpan Wang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Hongbin Ding
- School of Physics, Dalian University of Technology, Dalian, 116024, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Junhong Lyu
- Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China.
- Department of Neurology and Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Medical School, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
12
|
Gao XY, Zhou CX, Li HM, Cheng M, Chen D, Li ZY, Feng B, Song J. Correlation between cerebral neurotransmitters levels by proton magnetic resonance spectroscopy and HbA1c in patients with type 2 diabetes. World J Diabetes 2024; 15:1263-1271. [PMID: 38983812 PMCID: PMC11229970 DOI: 10.4239/wjd.v15.i6.1263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/28/2024] [Accepted: 04/24/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Cognitive dysfunction is the main manifestation of central neuropathy. Although cognitive impairments tend to be overlooked in patients with diabetes mellitus (DM), there is a growing body of evidence linking DM to cognitive dysfunction. Hyperglycemia is closely related to neurological abnormalities, while often disregarded in clinical practice. Changes in cerebral neurotransmitter levels are associated with a variety of neurological abnormalities and may be closely related to blood glucose control in patients with type 2 DM (T2DM). AIM To evaluate the concentrations of cerebral neurotransmitters in T2DM patients exhibiting different hemoglobin A1c (HbA1c) levels. METHODS A total of 130 T2DM patients were enrolled at the Department of Endocrinology of Shanghai East Hospital. The participants were divided into four groups according to their HbA1c levels using the interquartile method, namely Q1 (< 7.875%), Q2 (7.875%-9.050%), Q3 (9.050%-11.200%) and Q4 (≥ 11.200%). Clinical data were collected and measured, including age, height, weight, neck/waist/hip circumferences, blood pressure, comorbidities, duration of DM, and biochemical indicators. Meanwhile, neurotransmitters in the left hippocampus and left brainstem area were detected by proton magnetic resonance spectroscopy. RESULTS The HbA1c level was significantly associated with urinary microalbumin (mALB), triglyceride, low-density lipoprotein cholesterol (LDL-C), homeostasis model assessment of insulin resistance (HOMA-IR), and beta cell function (HOMA-β), N-acetylaspartate/creatine (NAA/Cr), and NAA/choline (NAA/Cho). Spearman correlation analysis showed that mALB, LDL-C, HOMA-IR and NAA/Cr in the left brainstem area were positively correlated with the level of HbA1c (P < 0.05), whereas HOMA-β was negatively correlated with the HbA1c level (P < 0.05). Ordered multiple logistic regression analysis showed that NAA/Cho [Odds ratio (OR): 1.608, 95% confidence interval (95%CI): 1.004-2.578, P < 0.05], LDL-C (OR: 1.627, 95%CI: 1.119-2.370, P < 0.05), and HOMA-IR (OR: 1.107, 95%CI: 1.031-1.188, P < 0.01) were independent predictors of poor glycemic control. CONCLUSION The cerebral neurotransmitter concentrations in the left brainstem area in patients with T2DM are closely related to glycemic control, which may be the basis for the changes in cognitive function in diabetic patients.
Collapse
Affiliation(s)
- Xiang-Yu Gao
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, Shandong Province, China
| | - Chen-Xia Zhou
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hong-Mei Li
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Min Cheng
- Department of Immunization Program, Huangdao District Center for Disease Prevention and Control, Qingdao 266400, Shandong Province, China
| | - Da Chen
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zi-Yi Li
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bo Feng
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jun Song
- Department of Endocrinology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
13
|
Lin R, Jin L, Xue Y, Zhang Z, Huang H, Chen D, Liu Q, Mao Z, Wu Z, Tao Q. Hybrid Membrane-Coated Nanoparticles for Precise Targeting and Synergistic Therapy in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306675. [PMID: 38647399 PMCID: PMC11200089 DOI: 10.1002/advs.202306675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/01/2024] [Indexed: 04/25/2024]
Abstract
The blood brain barrier (BBB) limits the application of most therapeutic drugs for neurological diseases (NDs). Hybrid cell membrane-coated nanoparticles derived from different cell types can mimic the surface properties and functionalities of the source cells, further enhancing their targeting precision and therapeutic efficacy. Neuroinflammation has been increasingly recognized as a critical factor in the pathogenesis of various NDs, especially Alzheimer's disease (AD). In this study, a novel cell membrane coating is designed by hybridizing the membrane from platelets and chemokine (C-C motif) receptor 2 (CCR2) cells are overexpressed to cross the BBB and target neuroinflammatory lesions. Past unsuccessful endeavors in AD drug development underscore the challenge of achieving favorable outcomes when utilizing single-mechanism drugs.Two drugs with different mechanisms of actions into liposomes are successfully loaded to realize multitargeting treatment. In a transgenic mouse model for familial AD (5xFAD), the administration of these drug-loaded hybrid cell membrane liposomes results in a significant reduction in amyloid plaque deposition, neuroinflammation, and cognitive impairments. Collectively, the hybrid cell membrane-coated nanomaterials offer new opportunities for precise drug delivery and disease-specific targeting, which represent a versatile platform for targeted therapy in AD.
Collapse
Affiliation(s)
- Rong‐Rong Lin
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| | - Lu‐Lu Jin
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Yan‐Yan Xue
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| | - Zhe‐Sheng Zhang
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| | - Hui‐Feng Huang
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| | - Dian‐Fu Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| | - Qian Liu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| | - Zheng‐Wei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Zhi‐Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
- MOE Frontier Science Center for Brain Science and Brain‐Machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhou310058China
- CAS Center for Excellence in Brain Science and Intelligence TechnologyShanghai200031China
| | - Qing‐Qing Tao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Liangzhu LaboratoryZhejiang University School of MedicineHangzhou310009China
| |
Collapse
|
14
|
Tan RX, Li WH, Pang JM, Zhong SM, Huang XY, Deng JZ, Zhou LY, Wu JQ, Wang XQ. Design, synthesis, and evaluation of 2,2'-bipyridyl derivatives as bifunctional agents against Alzheimer's disease. Mol Divers 2024; 28:1225-1238. [PMID: 37119457 DOI: 10.1007/s11030-023-10651-5] [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: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 05/01/2023]
Abstract
Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disease. Metal ion dyshomeostasis and Aβ aggregation have been proposed to contribute to AD progression. Metal ions can bind to Aβ and promote Aβ aggregation, and ultimately lead to neuronal death. Bifunctional (metal chelation and Aβ interaction) compounds are showing promise against AD. In this work, eleven new 3,3'-diamino-2,2'-bipyridine derivatives 4a-4k were synthesized, and evaluated as bifunctional agents for AD treatment. In vitro Aβ aggregation inhibition assay confirmed that most of the synthesized compounds exhibited significant self-induced Aβ1-42 aggregation inhibition. Among them, compound 4d displayed the best inhibitory potency of self-induced Aβ1-42 aggregation with IC50 value of 9.4 µM, and it could selectively chelate with Cu2+ and exhibited 66.2% inhibition of Cu2+-induced Aβ1-42 aggregation. Meanwhile, compound 4d showed strong neuroprotective activity against Aβ1-42 and Cu2+-treated Aβ1-42 induced cell damage. Moreover, compound 4d in high dose significantly reversed Aβ-induced memory impairment in mice.
Collapse
Affiliation(s)
- Ren-Xian Tan
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Wei-Hao Li
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jia-Min Pang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Si-Min Zhong
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Xin-Yi Huang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jun-Ze Deng
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Lu-Yi Zhou
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jia-Qiang Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China
| | - Xiao-Qin Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
15
|
Guan XJ, Deng ZQ, Liu J, Su CF, Tong BCK, Zhu Z, Sreenivasmurthy SG, Kan YX, Lu KJ, Chu CPK, Pi RB, Cheung KH, Iyaswamy A, Song JX, Li M. Corynoxine promotes TFEB/TFE3-mediated autophagy and alleviates Aβ pathology in Alzheimer's disease models. Acta Pharmacol Sin 2024; 45:900-913. [PMID: 38225393 PMCID: PMC11053156 DOI: 10.1038/s41401-023-01197-1] [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: 05/01/2023] [Accepted: 11/09/2023] [Indexed: 01/17/2024]
Abstract
Autophagy impairment is a key factor in Alzheimer's disease (AD) pathogenesis. TFEB (transcription factor EB) and TFE3 (transcription factor binding to IGHM enhancer 3) are nuclear transcription factors that regulate autophagy and lysosomal biogenesis. We previously showed that corynoxine (Cory), a Chinese medicine compound, protects neurons from Parkinson's disease (PD) by activating autophagy. In this study, we investigated the effect of Cory on AD models in vivo and in vitro. We found that Cory improved learning and memory function, increased neuronal autophagy and lysosomal biogenesis, and reduced pathogenic APP-CTFs levels in 5xFAD mice model. Cory activated TFEB/TFE3 by inhibiting AKT/mTOR signaling and stimulating lysosomal calcium release via transient receptor potential mucolipin 1 (TRPML1). Moreover, we demonstrated that TFEB/TFE3 knockdown abolished Cory-induced APP-CTFs degradation in N2aSwedAPP cells. Our findings suggest that Cory promotes TFEB/TFE3-mediated autophagy and alleviates Aβ pathology in AD models.
Collapse
Affiliation(s)
- Xin-Jie Guan
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Zhi-Qiang Deng
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Cheng-Fu Su
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Benjamin Chun-Kit Tong
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhou Zhu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Sravan Gopalkrishnashetty Sreenivasmurthy
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yu-Xuan Kan
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Ke-Jia Lu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Carol Pui-Kei Chu
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Rong-Biao Pi
- School of Medicine, Sun Yat-sen University (Shenzhen), Shenzhen, 518107, China
| | - King-Ho Cheung
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China.
| | - Ju-Xian Song
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Min Li
- Mr. & Mrs. Ko Chi Ming Centre for Parkinson's Disease Research (CPDR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, 518057, China.
| |
Collapse
|
16
|
Choi JK, Kwon OY, Lee SH. Oral Administration of Bifidobacterium lactis Ameliorates Cognitive Deficits in Mice Intracerebroventricularly Administered Amyloid Beta via Regulation the Activation of Mitogen-activated Protein Kinases. Food Sci Anim Resour 2024; 44:607-619. [PMID: 38765290 PMCID: PMC11097010 DOI: 10.5851/kosfa.2024.e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 05/22/2024] Open
Abstract
Probiotics are functional microorganisms that exhibit various biological activities, such as allergic reactions, inflammation, and aging. The aim of this study is to evaluate the effects of Bifidobacterium lactis CBT BL3 (BL) on the amyloid beta (Aβ)-mediated cognitive impairments. Oral administration of live BL to intracerebroventricularly Aβ-injected mice significantly attenuated short- and long-term memory loss estimated using the Y-maze and Morris water maze tests. We found that expression of apoptosis-related proteins such as caspase-9, caspase-3, and cleaved poly (ADP-ribose) polymerase was significantly elevated in the brain tissues of Aβ-injected mouse brains when compared to that of the control mouse group. Interestingly, these expression levels were significantly decreased in the brain tissue of mice fed BL for 6 wk. In addition, the abnormal over-phosphorylation of mitogen-activated protein kinases (MAPKs) such as ERK1/2, p38 MAPK, and JNK in the brain tissue of intracerebroventricularly Aβ-injected mice was significantly attenuated by oral administration of BL. Taken together, the results indicate that Aβ-induced cognitive impairment may be ameliorated by the oral administration of BL by controlling the activation of MAPKs/apoptosis in the brain. This study strongly suggests that BL can be developed as a functional probiotic to attenuate Aβ-mediated cognitive deficits.
Collapse
Affiliation(s)
- Jong Kyu Choi
- Department of Nano-Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Oh Yun Kwon
- Department of Nano-Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute of New Drug Development, Incheon National University, Incheon 22012, Korea
| | - Seung Ho Lee
- Department of Nano-Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute of New Drug Development, Incheon National University, Incheon 22012, Korea
| |
Collapse
|
17
|
Muthu SA, Qureshi A, Sharma R, Bisaria I, Parvez S, Grover S, Ahmad B. Redesigning the kinetics of lysozyme amyloid aggregation by cephalosporin molecules. J Biomol Struct Dyn 2024:1-16. [PMID: 38682862 DOI: 10.1080/07391102.2024.2335304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/20/2024] [Indexed: 05/01/2024]
Abstract
In lysozyme amyloidosis, fibrillar aggregates of lysozyme are associated with severe renal, hepatic, and gastrointestinal manifestations, with no definite therapy. Current drugs are now being tested in amyloidosis clinical trials as aggregation inhibitors to mitigate disease progression. The tetracycline group among antimicrobials in use is in phase II of clinical trials, whereas some macrolides and cephalosporins have shown neuroprotection. In the present study, two cephalosporins, ceftazidime (CZD) and cefotaxime (CXM), and a glycopeptide, vancomycin (VNC), are evaluated for inhibition of amyloid aggregation of hen egg white lysozyme (HEWL) under two conditions (i) 4 M guanidine hydrochloride (GuHCl) at pH 6.5 and 37° C, (ii) At pH 1.5 and 65 °C. Fluorescence quench titration and molecular docking methods report that CZD, CXM, and VNC interact more strongly with the partially folded intermediates (PFI) in comparison to the protein's natural state (N). However, only CZD and CXM proficiently inhibit the aggregation. Transmission electron microscopy, tinctorial assessments, and aggregation kinetics all support oligomer-level inhibition. Transition structures in CZD-HEWL and CXM-HEWL aggregation are shown by circular dichroism (CD). On the other hand, kinetic variables and soluble fraction assays point to a localized association of monomers. Intrinsic fluorescence (IF),1-Anilino 8-naphthalene sulphonic acid, and CD demonstrate structural and conformational modifications redesigning the PFI. GuHCl-induced unfolding and differential scanning fluorimetry suggested that the PFI monomers bound to CZD and CXM exhibited partial stability. Our results present two mechanisms that function in both solution conditions, creating a novel avenue for the screening of putative inhibitors for drug repurposing. We extend our proposed mechanisms in the designing of physical inhibitors of amyloid aggregation considering shorter time frames and foolproof methods.
Collapse
Affiliation(s)
- Shivani A Muthu
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
- Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi, India
| | - Afnaan Qureshi
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Rahul Sharma
- Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi, India
| | - Ishita Bisaria
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Sonam Grover
- Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi, India
| | - Basir Ahmad
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
18
|
Jaye S, Sandau US, Saugstad JA. Clathrin mediated endocytosis in Alzheimer's disease: cell type specific involvement in amyloid beta pathology. Front Aging Neurosci 2024; 16:1378576. [PMID: 38694257 PMCID: PMC11061891 DOI: 10.3389/fnagi.2024.1378576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
This review provides a comprehensive examination of the role of clathrin-mediated endocytosis (CME) in Alzheimer's disease (AD) pathogenesis, emphasizing its impact across various cellular contexts beyond neuronal dysfunction. In neurons, dysregulated CME contributes to synaptic dysfunction, amyloid beta (Aβ) processing, and Tau pathology, highlighting its involvement in early AD pathogenesis. Furthermore, CME alterations extend to non-neuronal cell types, including astrocytes and microglia, which play crucial roles in Aβ clearance and neuroinflammation. Dysregulated CME in these cells underscores its broader implications in AD pathophysiology. Despite significant progress, further research is needed to elucidate the precise mechanisms underlying CME dysregulation in AD and its therapeutic implications. Overall, understanding the complex interplay between CME and AD across diverse cell types holds promise for identifying novel therapeutic targets and interventions.
Collapse
Affiliation(s)
| | | | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
19
|
Trujillo-Rangel WÁ, Acuña-Vaca S, Padilla-Ponce DJ, García-Mercado FG, Torres-Mendoza BM, Pacheco-Moises FP, Escoto-Delgadillo M, García-Benavides L, Delgado-Lara DLC. Modulation of the Circadian Rhythm and Oxidative Stress as Molecular Targets to Improve Vascular Dementia: A Pharmacological Perspective. Int J Mol Sci 2024; 25:4401. [PMID: 38673986 PMCID: PMC11050388 DOI: 10.3390/ijms25084401] [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: 03/03/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The circadian rhythms generated by the master biological clock located in the brain's hypothalamus influence central physiological processes. At the molecular level, a core set of clock genes interact to form transcription-translation feedback loops that provide the molecular basis of the circadian rhythm. In animal models of disease, a desynchronization of clock genes in peripheral tissues with the central master clock has been detected. Interestingly, patients with vascular dementia have sleep disorders and irregular sleep patterns. These alterations in circadian rhythms impact hormonal levels, cardiovascular health (including blood pressure regulation and blood vessel function), and the pattern of expression and activity of antioxidant enzymes. Additionally, oxidative stress in vascular dementia can arise from ischemia-reperfusion injury, amyloid-beta production, the abnormal phosphorylation of tau protein, and alterations in neurotransmitters, among others. Several signaling pathways are involved in the pathogenesis of vascular dementia. While the precise mechanisms linking circadian rhythms and vascular dementia are still being studied, there is evidence to suggest that maintaining healthy sleep patterns and supporting proper circadian rhythm function may be important for reducing the risk of vascular dementia. Here, we reviewed the main mechanisms of action of molecular targets related to the circadian cycle and oxidative stress in vascular dementia.
Collapse
Affiliation(s)
- Walter Ángel Trujillo-Rangel
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555, Ejido San José Tateposco, Tonalá 45425, Jalisco, Mexico; (W.Á.T.-R.); (D.J.P.-P.); (F.G.G.-M.); (L.G.-B.)
- Departamento de Formación Universitaria Ciencias de la Salud, Universidad Autónoma de Guadalajara, Av. Patria 1201, Lomas del Valle, Zapopan 45129, Jalisco, Mexico;
| | - Sofía Acuña-Vaca
- Departamento de Formación Universitaria Ciencias de la Salud, Universidad Autónoma de Guadalajara, Av. Patria 1201, Lomas del Valle, Zapopan 45129, Jalisco, Mexico;
| | - Danna Jocelyn Padilla-Ponce
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555, Ejido San José Tateposco, Tonalá 45425, Jalisco, Mexico; (W.Á.T.-R.); (D.J.P.-P.); (F.G.G.-M.); (L.G.-B.)
| | - Florencia Guillermina García-Mercado
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555, Ejido San José Tateposco, Tonalá 45425, Jalisco, Mexico; (W.Á.T.-R.); (D.J.P.-P.); (F.G.G.-M.); (L.G.-B.)
| | - Blanca Miriam Torres-Mendoza
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada 800, Colonia Independencia, Guadalajara 44340, Jalisco, Mexico; (B.M.T.-M.); (M.E.-D.)
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Colonia Independencia, Guadalajara 44340, Jalisco, Mexico
| | - Fermín P. Pacheco-Moises
- Departamento de Química, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. Marcelino García Barragán No. 1421, Guadalajara 44430, Jalisco, Mexico;
| | - Martha Escoto-Delgadillo
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada 800, Colonia Independencia, Guadalajara 44340, Jalisco, Mexico; (B.M.T.-M.); (M.E.-D.)
- Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez No. 2100, Zapopan 45200, Jalisco, Mexico
| | - Leonel García-Benavides
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555, Ejido San José Tateposco, Tonalá 45425, Jalisco, Mexico; (W.Á.T.-R.); (D.J.P.-P.); (F.G.G.-M.); (L.G.-B.)
| | - Daniela L. C. Delgado-Lara
- Departamento de Formación Universitaria Ciencias de la Salud, Universidad Autónoma de Guadalajara, Av. Patria 1201, Lomas del Valle, Zapopan 45129, Jalisco, Mexico;
| |
Collapse
|
20
|
Cohen J, Mathew A, Dourvetakis KD, Sanchez-Guerrero E, Pangeni RP, Gurusamy N, Aenlle KK, Ravindran G, Twahir A, Isler D, Sosa-Garcia SR, Llizo A, Bested AC, Theoharides TC, Klimas NG, Kempuraj D. Recent Research Trends in Neuroinflammatory and Neurodegenerative Disorders. Cells 2024; 13:511. [PMID: 38534355 PMCID: PMC10969521 DOI: 10.3390/cells13060511] [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: 12/25/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Neuroinflammatory and neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI) and Amyotrophic lateral sclerosis (ALS) are chronic major health disorders. The exact mechanism of the neuroimmune dysfunctions of these disease pathogeneses is currently not clearly understood. These disorders show dysregulated neuroimmune and inflammatory responses, including activation of neurons, glial cells, and neurovascular unit damage associated with excessive release of proinflammatory cytokines, chemokines, neurotoxic mediators, and infiltration of peripheral immune cells into the brain, as well as entry of inflammatory mediators through damaged neurovascular endothelial cells, blood-brain barrier and tight junction proteins. Activation of glial cells and immune cells leads to the release of many inflammatory and neurotoxic molecules that cause neuroinflammation and neurodegeneration. Gulf War Illness (GWI) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are chronic disorders that are also associated with neuroimmune dysfunctions. Currently, there are no effective disease-modifying therapeutic options available for these diseases. Human induced pluripotent stem cell (iPSC)-derived neurons, astrocytes, microglia, endothelial cells and pericytes are currently used for many disease models for drug discovery. This review highlights certain recent trends in neuroinflammatory responses and iPSC-derived brain cell applications in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Jessica Cohen
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Annette Mathew
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kirk D Dourvetakis
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Estella Sanchez-Guerrero
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Rajendra P Pangeni
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kristina K Aenlle
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Geeta Ravindran
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Dylan Isler
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Sara Rukmini Sosa-Garcia
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Axel Llizo
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Alison C Bested
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| |
Collapse
|
21
|
Mishra V, Yadav D, Solanki KS, Koul B, Song M. A Review on the Protective Effects of Probiotics against Alzheimer's Disease. BIOLOGY 2023; 13:8. [PMID: 38248439 PMCID: PMC10813289 DOI: 10.3390/biology13010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024]
Abstract
This review summarizes the protective effects of probiotics against Alzheimer's disease (AD), one of the most common neurodegenerative disorders affecting older adults. This disease is characterized by the deposition of tau and amyloid β peptide (Aβ) in different parts of the brain. Symptoms observed in patients with AD include struggles with writing, speech, memory, and knowledge. The gut microbiota reportedly plays an important role in brain functioning due to its bidirectional communication with the gut via the gut-brain axis. The emotional and cognitive centers in the brain are linked to the functions of the peripheral intestinal system via this gut-brain axis. Dysbiosis has been linked to neurodegenerative disorders, indicating the significance of gut homeostasis for proper brain function. Probiotics play an important role in protecting against the symptoms of AD as they restore gut-brain homeostasis to a great extent. This review summarizes the characteristics, status of gut-brain axis, and significance of gut microbiota in AD. Review and research articles related to the role of probiotics in the treatment of AD were searched in the PubMed database. Recent studies conducted using animal models were given preference. Recent clinical trials were searched for separately. Several studies conducted on animal and human models clearly explain the benefits of probiotics in improving cognition and memory in experimental subjects. Based on these studies, novel therapeutic approaches can be designed for the treatment of patients with AD.
Collapse
Affiliation(s)
- Vibhuti Mishra
- School of Studies in Biochemistry, Jiwaji University, Gwalior 474003, India;
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Kavita Singh Solanki
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA;
| | - Bhupendra Koul
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India;
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
22
|
Bai Q, Sun D, Zeng Y, Zhu J, Zhang C, Zhang X, Chen L, Zhou X, Ye L, Tang Y, Liu Y, Morozova-Roche LA. Effect of Proinflammatory S100A9 Protein on Migration and Proliferation of Microglial Cells. J Mol Neurosci 2023; 73:983-995. [PMID: 37947991 DOI: 10.1007/s12031-023-02168-1] [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: 05/19/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial disease affecting aging population worldwide. Neuroinflammation became a focus of research as one of the major pathologic processes relating to the disease onset and progression. Proinflammatory S100A9 is the central culprit in the amyloid-neuroinflammatory cascade implicated in AD and other neurodegenerative diseases. We studied the effect of S100A9 on microglial BV-2 cell proliferation and migration. The responses of BV-2 cells to S100A9 stimulation were monitored in real-time using live cell microscopy, transcriptome sequencing, immunofluorescence staining, western blot analysis, and ELISA. We observed that a low dose of S100A9 promotes migration and proliferation of BV-2 cells. However, acute inflammatory condition (i.e., high S100A9 doses) causes diminished cell viability; it is uncovered that S100A9 activates TLR-4 and TLR-7 signaling pathways, leading to TNF-α and IL-6 expression, which affect BV-2 cell migration and proliferation in a concentration-dependent manner. Interestingly, the effects of S100A9 are not only inhibited by TNF-α and IL-6 antibodies. The addition of amyloid-β (Aβ) 1-40 peptide resumes the capacities of BV-2 cells to the level of low S100A9 concentrations. Based on these results, we conclude that in contrast to the beneficial effects of low S100A9 dose, high S100A9 concentration leads to impaired mobility and proliferation of immune cells, reflecting neurotoxicity at acute inflammatory conditions. However, the formation of Aβ plaques may be a natural mechanism that rescues cells from the proinflammatory and cytotoxic effects of S100A9, especially considering that inflammation is one of the primary causes of AD.
Collapse
Affiliation(s)
- Qiao Bai
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Yang Zeng
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Jie Zhu
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Xiaoyin Zhang
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Li Chen
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Xin Zhou
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Liu Ye
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Yong Tang
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China
| | - Yonggang Liu
- Chongqing Medical University, 1 Medical College Road, Yu Zhong District, Chongqing, China.
| | | |
Collapse
|
23
|
Wu KJ, Wang WR, Cheng QH, Li H, Yan WZ, Zhou FR, Zhang RJ. Pyroptosis in neurodegenerative diseases: from bench to bedside. Cell Biol Toxicol 2023; 39:2467-2499. [PMID: 37491594 DOI: 10.1007/s10565-023-09820-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023]
Abstract
The central nervous system regulates all aspects of physiology to some extent. Neurodegenerative diseases (NDDs) lead to the progressive loss and dysfunction of neurons, which are particularly evident in Alzheimer's disease, Parkinson's disease, and many other conditions. NDDs are multifactorial diseases with complex pathogeneses, and there has been a rapid increase in the prevalence of NDDs. However, none of these diseases can be cured, making the development of novel treatment strategies an urgent necessity. Numerous studies have indicated how pyroptosis induces inflammation and affects many aspects of NDD. Therefore, components related to pyroptosis are potential therapeutic candidates and are attracting increasing attention. Here, we review the role of pyroptosis in the pathogenesis of NDDs and potential treatment options. Additionally, several of the current drugs and relevant inhibitors are discussed. Through this article, we provide theoretical support for exploring new therapeutic targets and updating clinical treatment strategies for NDDs. Notably, pyroptosis, a recently widely studied mode of cell death, is still under-researched compared to other traditional forms of cell death. Moreover, the focus of research has been on the onset and progression of NDDs, and the lack of organ-specific target discovery and drug development is a common problem for many basic studies. This urgent problem requires scientists and companies worldwide to collaborate in order to develop more effective drugs against NDDs.
Collapse
Affiliation(s)
- Ke-Jia Wu
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China
| | - Wan-Rong Wang
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China
| | - Qian-Hui Cheng
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China
| | - Hao Li
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China
| | - Wei-Zhen Yan
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China
| | - Fei-Ran Zhou
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China
| | - Rui-Jie Zhang
- College of Life Sciences, Anhui Medical University, Tanghe Road, Hefei, 230012, Anhui, People's Republic of China.
| |
Collapse
|
24
|
Feng J, Zheng Y, Guo M, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga MR, Wang X, Anadón A, Martínez MA. Oxidative stress, the blood-brain barrier and neurodegenerative diseases: The critical beneficial role of dietary antioxidants. Acta Pharm Sin B 2023; 13:3988-4024. [PMID: 37799389 PMCID: PMC10547923 DOI: 10.1016/j.apsb.2023.07.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/16/2023] [Accepted: 06/13/2023] [Indexed: 10/07/2023] Open
Abstract
In recent years, growing awareness of the role of oxidative stress in brain health has prompted antioxidants, especially dietary antioxidants, to receive growing attention as possible treatments strategies for patients with neurodegenerative diseases (NDs). The most widely studied dietary antioxidants include active substances such as vitamins, carotenoids, flavonoids and polyphenols. Dietary antioxidants are found in usually consumed foods such as fresh fruits, vegetables, nuts and oils and are gaining popularity due to recently growing awareness of their potential for preventive and protective agents against NDs, as well as their abundant natural sources, generally non-toxic nature, and ease of long-term consumption. This review article examines the role of oxidative stress in the development of NDs, explores the 'two-sidedness' of the blood-brain barrier (BBB) as a protective barrier to the nervous system and an impeding barrier to the use of antioxidants as drug medicinal products and/or dietary antioxidants supplements for prevention and therapy and reviews the BBB permeability of common dietary antioxidant suplements and their potential efficacy in the prevention and treatment of NDs. Finally, current challenges and future directions for the prevention and treatment of NDs using dietary antioxidants are discussed, and useful information on the prevention and treatment of NDs is provided.
Collapse
Affiliation(s)
- Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Youle Zheng
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| | - Mingyue Guo
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| |
Collapse
|
25
|
Yuan M, Feng Y, Zhao M, Xu T, Li L, Guo K, Hou D. Identification and verification of genes associated with hypoxia microenvironment in Alzheimer's disease. Sci Rep 2023; 13:16252. [PMID: 37759083 PMCID: PMC10533856 DOI: 10.1038/s41598-023-43595-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023] Open
Abstract
As the incidence of Alzheimer's disease (AD) increases year by year, more people begin to study this disease. In recent years, many studies on reactive oxygen species (ROS), neuroinflammation, autophagy, and other fields have confirmed that hypoxia is closely related to AD. However, no researchers have used bioinformatics methods to study the relationship between AD and hypoxia. Therefore, our study aimed to screen the role of hypoxia-related genes in AD and clarify their diagnostic significance. A total of 7681 differentially expressed genes (DEGs) were identified in GSE33000 by differential expression analysis and cluster analysis. Weighted gene co-expression network analysis (WGCNA) was used to detect 9 modules and 205 hub genes with high correlation coefficients. Next, machine learning algorithms were applied to 205 hub genes and four key genes were selected. Through the verification of external dataset and quantitative real-time PCR (qRT-PCR), the AD diagnostic model was established by ANTXR2, BDNF and NFKBIA. The bioinformatics analysis results suggest that hypoxia-related genes may increase the risk of AD. However, more in-depth studies are still needed to investigate their association, this article would guide the insights and directions for further research.
Collapse
Affiliation(s)
- Mingyang Yuan
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Yanjin Feng
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Mingri Zhao
- School of Life Sciences, Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, 410000, China
| | - Ting Xu
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Liuhong Li
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Ke Guo
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Deren Hou
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China.
| |
Collapse
|
26
|
Wiatrak B, Jawień P, Szeląg A, Jęśkowiak-Kossakowska I. Does Inflammation Play a Major Role in the Pathogenesis of Alzheimer's Disease? Neuromolecular Med 2023; 25:330-335. [PMID: 37027081 PMCID: PMC10514153 DOI: 10.1007/s12017-023-08741-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/03/2023] [Indexed: 04/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease leading to dementia for which no effective medicine exists. Currently, the goal of therapy is only to slow down the inevitable progression of the disease and reduce some symptoms. AD causes the accumulation of proteins with the pathological structure of Aβ and tau and the induction of inflammation of nerves in the brain, which lead to the death of neurons. The activated microglial cells produce pro-inflammatory cytokines that induce a chronic inflammatory response and mediate synapse damage and the neuronal death. Neuroinflammation has been an often ignored aspect of ongoing AD research. There are more and more scientific papers taking into account the aspect of neuroinflammation in the pathogenesis of AD, although there are no unambiguous results regarding the impact of comorbidities or gender differences. This publication concerns a critical look at the role of inflammation in the progression of AD, based on the results of our own in vitro studies using model cell cultures and other researchers.
Collapse
Affiliation(s)
- Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland
| | - Paulina Jawień
- Department of Biostructure and Animal Physiology, Wroclaw University of Environmental and Life Sciences, Norwida 25/27, 50-375 Wroclaw, Poland
| | - Adam Szeląg
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland
| | - Izabela Jęśkowiak-Kossakowska
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wrocław, Poland
| |
Collapse
|
27
|
Choudhury N, Chen L, Al-Harthi L, Hu XT. Hyperactivity of medial prefrontal cortex pyramidal neurons occurs in a mouse model of early-stage Alzheimer's disease without β-amyloid accumulation. Front Pharmacol 2023; 14:1194869. [PMID: 37465526 PMCID: PMC10350500 DOI: 10.3389/fphar.2023.1194869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023] Open
Abstract
The normal function of the medial prefrontal cortex (mPFC) is essential for regulating neurocognition, but it is disrupted in the early stages of Alzheimer's disease (AD) before the accumulation of Aβ and the appearance of symptoms. Despite this, little is known about how the functional activity of medial prefrontal cortex pyramidal neurons changes as Alzheimer's disease progresses during aging. We used electrophysiological techniques (patch-clamping) to assess the functional activity of medial prefrontal cortex pyramidal neurons in the brain of 3xTg-Alzheimer's disease mice modeling early-stage Alzheimer's disease without Aβ accumulation. Our results indicate that firing rate and the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) were significantly increased in medial prefrontal cortex neurons from young Alzheimer's disease mice (4-5-month, equivalent of <30-year-old humans) compared to age-matched control mice. Blocking ionotropic glutamatergic NMDA receptors, which regulate neuronal excitability and Ca2+ homeostasis, abolished this neuronal hyperactivity. There were no changes in Ca2+ influx through the voltage-gated Ca2+ channels (VGCCs) or inhibitory postsynaptic activity in medial prefrontal cortex neurons from young Alzheimer's disease mice compared to controls. Additionally, acute exposure to Aβ42 potentiated medial prefrontal cortex neuronal hyperactivity in young Alzheimer's disease mice but had no effects on controls. These findings indicate that the hyperactivity of medial prefrontal cortex pyramidal neurons at early-stage Alzheimer's disease is induced by an abnormal increase in presynaptic glutamate release and postsynaptic NMDA receptor activity, which initiates neuronal Ca2+ dyshomeostasis. Additionally, because accumulated Aβ forms unconventional but functional Ca2+ channels in medial prefrontal cortex neurons in the late stage of Alzheimer's disease, our study also suggests an exacerbated Ca2+ dyshomeostasis in medial prefrontal cortex pyramidal neurons following overactivation of such VGCCs.
Collapse
Affiliation(s)
| | | | | | - Xiu-Ti Hu
- Department of Microbial Pathogens and Immunity, Rush University Medical Centre, Chicago, IL, United States
| |
Collapse
|
28
|
Wang M, Chen X, Niu L, Xu J, Yu H, Xu X, Yang Q, Xiang Y, Le W. APP swe /PS1 ΔE9 mice exhibit low oxygen saturation and alterations of erythrocytes preceding the neuropathology and cognitive deficiency during Alzheimer's disease. CNS Neurosci Ther 2023; 29:1889-1897. [PMID: 36883266 PMCID: PMC10546960 DOI: 10.1111/cns.14147] [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: 09/02/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/09/2023] Open
Abstract
AIM The molecular mechanism underlying Alzheimer's disease (AD) pathologies remains unclear. The brain is extremely sensitive to oxygen deprivation, and brief interruptions in oxygen supply may lead to permanent brain damage. The objective here was to access the red blood cell (RBC) physiological alterations and the changes in blood oxygen saturation of an AD model as well as to explore the possible mechanism underlying these pathologies. METHODS We used female APPswe /PS1ΔE9 mice as AD models. Data were collected at the age of 3, 6, and 9 months. In addition to examining classic features of AD, namely cognitive deficiency and Aβ depositions, 24 h blood oxygen saturation was monitored by Plus oximeters in real time. In addition, RBC physiological parameters were measured by blood cell counter using peripheral blood from the epicanthal veins. Furthermore, in the mechanism investigations, the expression of phosphorylated band 3 protein was examined by a series of Western blot analyses, and the levels of soluble Aβ40 and Aβ42 on the membrane of RBCs were determined by ELISA. RESULTS Our results showed that the blood oxygen saturation in the AD mice was significantly reduced as early as at 3 months of age, preceding the neuropathological changes and cognitive impairments. Meanwhile, the expression of phosphorylated band 3 protein and levels of soluble Aβ40 and Aβ42 were all elevated in the erythrocytes of the AD mice. CONCLUSION APPswe /PS1ΔE9 mice exhibited decreased oxygen saturation together with reduced RBC counts and hemoglobin concentrations at the early stage, which may aid in the development of predictive markers for AD diagnosis. The increased expression of band 3 protein and elevated Aβ40 and Aβ42 levels may contribute to the deformation of RBCs and, in turn, cause the subsequent AD development.
Collapse
Affiliation(s)
- Manli Wang
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Long Niu
- Center for Clinical Research on Neurological Diseases, the First Affiliated HospitalDalian Medical UniversityDalianChina
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Jianli Xu
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Hang Yu
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Xiaojiao Xu
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Qiu Yang
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Yang Xiang
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Chinese Academy of Sciences Sichuan Translational Medicine Research HospitalChengduChina
| |
Collapse
|
29
|
Kim HJ, Kim REY, Kim S, Lee SK, Lee HW, Shin C. Earlier chronotype in midlife as a predictor of accelerated brain aging: a population-based longitudinal cohort study. Sleep 2023; 46:zsad108. [PMID: 37061816 DOI: 10.1093/sleep/zsad108] [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: 05/14/2022] [Revised: 02/28/2023] [Indexed: 04/17/2023] Open
Abstract
STUDY OBJECTIVES Evidence suggests that sleep-wake cycle disruption could be an early manifestation of neurodegeneration and might even be a risk factor for developing diseases in healthy adults. We investigated the impact of circadian phase change on structural and functional brain deterioration in a late-adulthood population. METHODS We analyzed the data of 1874 participants (mean age 58.6 ± 6.3 years, 50.3% female) from the Korean Genome and Epidemiology Study, who were identified as cognitively unimpaired. The mid-sleep time on free days corrected for sleep debt on workdays (MSFsc) at baseline was adopted as an indicator of the chronotype and used to categorize the participants into three groups. The relationships between the chronotype and longitudinal changes in the gray matter volume (GMV) and cognitive function were investigated (mean interval: 4.2 ± 0.5 years). RESULTS The mean MSFsc of the participants was 2:45 am. The earlier MSFsc was linearly associated with smaller right entorhinal GMV (β [SE] = 0.02 [0.01]; p = .001) and lower visual memory function test scores at baseline. Longitudinally, the earlier MSFsc at baseline was only significantly associated with more rapid atrophy in the temporal lobe (β [SE] = 0.18 [0.07]; p = .018) and not with other brain lobes or subregions. Moreover, the earlier MSFsc was associated with more deteriorated verbal learning and visual memory function test scores. CONCLUSIONS An earlier chronotype in midlife, measured using a questionnaire, can be a valuable indicator for individuals who should be closely monitored for the development of neurodegenerative disorders.
Collapse
Affiliation(s)
- Hyeon Jin Kim
- Department of Neurology, Korea University Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Departments of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, Republic of Korea
| | - Regina E Y Kim
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Soriul Kim
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seung Ku Lee
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
| | - Hyang Woon Lee
- Departments of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, Republic of Korea
- Computational Medicine, Graduate Programs in System Health Science & Engineering and Artificial Intelligence Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Chol Shin
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Biomedical Research Center, Korea University Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Tang B, Wang Y, Ren J. Basic information about memantine and its treatment of Alzheimer's disease and other clinical applications. IBRAIN 2023; 9:340-348. [PMID: 37786758 PMCID: PMC10527776 DOI: 10.1002/ibra.12098] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 10/04/2023]
Abstract
Memantine is a noncompetitive moderate-affinity strong voltage-dependent N-methyl-D-aspartate receptor antagonist. It has been used to treat Alzheimer's disease (AD) since 1989. In 2018, it became the second most commonly used drug for the treatment of dementia in the world. AD is nonreversible, and memantine can only relieve the symptoms of AD but not cure it. Over the past half-century, memantine's research and clinical application have been extensively developed. In this review, the basic composition of memantine, the mechanism and limitations of memantine in the treatment of AD, memantine combination therapy, comparison of memantine with other drugs for AD, and clinical studies of memantine in other diseases are reviewed to provide a valuable reference for further research and application of memantine for the treatment of AD.
Collapse
Affiliation(s)
- Bin‐Can Tang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Ya‐Ting Wang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Jie Ren
- Department of NeuroscienceThe University of SheffieldSheffieldUK
| |
Collapse
|
31
|
Sun Y, Wang K, Zhao W. Gut microbiota in perioperative neurocognitive disorders: current evidence and future directions. Front Immunol 2023; 14:1178691. [PMID: 37215136 PMCID: PMC10192759 DOI: 10.3389/fimmu.2023.1178691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/13/2023] [Indexed: 05/24/2023] Open
Abstract
Perioperative neurocognitive disorders (PND) is a common surgical anesthesia complication characterized by impairment of memory, attention, language understanding and social ability, which can lead to a decline in the quality of life of patients, prolong the hospitalization period and increase the mortality rate. PND has a high incidence rate, which has a great impact on postoperative recovery and quality of life of patients, and has caused a heavy economic burden to society and families. In recent years, PND has become an important public health problem. The high risk population of PND is more prone to gut microbiota imbalance, and gut microbiota may also affect the inflammatory response of the central nervous system through the microbiota-gut-brain axis. Meanwhile, Neuroinflammation and immune activation are important mechanisms of PND. Regulating gut microbiota through probiotics or fecal bacteria transplantation can significantly reduce neuroinflammation, reduce the abnormal activation of immune system and prevent the occurrence of PND. This review summarizes the research progress of gut microbiota and PND, providing basis for the prevention and treatment of PND.
Collapse
|
32
|
Sha S, Chaigneau T, Krantic S. Pre-symptomatic synaptic dysfunction and longitudinal decay of hippocampal synaptic function in APPPS1 mouse model of Alzheimer's disease is sex-independent. Brain Res Bull 2023; 198:36-49. [PMID: 37080395 DOI: 10.1016/j.brainresbull.2023.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/19/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Alzheimer's disease (AD) is an incurable, age-related and progressive neurodegenerative disease characterized by cognitive impairments. Deficits in synaptic plasticity were reported in various models of AD-like pathology and are considered as an early contributing factor of cognitive impairment. However, the majority of previous studies were focused on overt, symptomatic stages of pathology and assessed long-term potentiation (LTP), whereas long-term depression (LTD) was much less investigated and the precise nature of its involvement remains poorly defined. To better understand the earliest synaptic dysfunctions along the pre-symptomatic stage of AD-like pathology, we performed a detailed analysis of underlying mechanisms and quantified basal synaptic activity, presynaptic release probability, and synaptic plasticity such as post-tetanic potentiation (PTP), as well as LTP and LTD. These parameters were studied in APPPS1 mouse model at two time points (early- and mid-) along the pre-symptomatic stage, which were compared with alterations monitored at two later time-points, i.e. the onset of cognitive deficits and the overt stage of full-blown pathology. Because sex is known to be an instrumental biological parameter in AD pathophysiology, all alterations were assessed in both males and females. Our data show that, as compared to wild-type (WT) littermates, initial neuronal hyperexcitability, seen at early pre-symptomatic stage shifts subsequently towards hypoexcitability at mid-pre-symptomatic stage and remains impaired at advanced stages. The pre-symptomatic changes also involve increased synaptic plasticity as assessed by paired-pulse facilitation (PPF), which returns to basal level at the onset of pathology and remains stable afterwards. Synaptic plasticity is impaired by mid-pre-symptomatic stage and manifests as lowered LTP and absence of LTD induction, the latter being reported here for the first time. Observed LTP and LTD impairments both persist in older APPPS1 mice. Remarkably, none of the observed differences was gender-dependent. Altogether, our data evidence that major impairments in basal synaptic efficacy and plasticity are detectable already during mid-pre-symptomatic stage of AD-like pathogenesis and likely involve hyperexcitability as the underlying mechanism. Our study also uncovers synaptic alterations that may become critical read-outs for testing the efficiency of novel, pre-symptomatic stage-targeted therapies for AD.
Collapse
Affiliation(s)
- Sha Sha
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France; Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Thomas Chaigneau
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Slavica Krantic
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France.
| |
Collapse
|
33
|
Ono H, Nishijima Y, Ohta S. Therapeutic Inhalation of Hydrogen Gas for Alzheimer’s Disease Patients and Subsequent Long-Term Follow-Up as a Disease-Modifying Treatment: An Open Label Pilot Study. Pharmaceuticals (Basel) 2023; 16:ph16030434. [PMID: 36986533 PMCID: PMC10057981 DOI: 10.3390/ph16030434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
(1) Background: Alzheimer’s disease (AD) is a progressive and fatal neurodegenerative disorder. Hydrogen gas (H2) is a therapeutic medical gas with multiple functions such as anti-oxidant, anti-inflammation, anti-cell death, and the stimulation of energy metabolism. To develop a disease-modifying treatment for AD through multifactorial mechanisms, an open label pilot study on H2 treatment was conducted. (2) Methods: Eight patients with AD inhaled 3% H2 gas for one hour twice daily for 6 months and then followed for 1 year without inhaling H2 gas. The patients were clinically assessed using the Alzheimer’s Disease Assessment Scale-cognitive subscale (ADAS-cog). To objectively assess the neuron integrity, diffusion tensor imaging (DTI) with advanced magnetic resonance imaging (MRI) was applied to neuron bundles passing through the hippocampus. (3) Results: The mean individual ADAS-cog change showed significant improvement after 6 months of H2 treatment (−4.1) vs. untreated patients (+2.6). As assessed by DTI, H2 treatment significantly improved the integrity of neurons passing through the hippocampus vs. the initial stage. The improvement by ADAS-cog and DTI assessments were maintained during the follow-up after 6 months (significantly) or 1 year (non-significantly). (4) Conclusions: This study suggests that H2 treatment not only relieves temporary symptoms, but also has disease-modifying effects, despite its limitations.
Collapse
Affiliation(s)
- Hirohisa Ono
- Departments of Neurosurgery and Neurology, Nishijima Hospital, Ohoka, 2835-7, Numazu City 410-0022, Japan
- Correspondence: (H.O.); (S.O.); Tel.: +81-80-5658-5858 (H.O.); +81-90-9824-2970 (S.O.); Fax: +81-44-434-2336 (S.O.)
| | - Yoji Nishijima
- Departments of Neurosurgery and Neurology, Nishijima Hospital, Ohoka, 2835-7, Numazu City 410-0022, Japan
| | - Shigeo Ohta
- Department of Neurology Medicine, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Correspondence: (H.O.); (S.O.); Tel.: +81-80-5658-5858 (H.O.); +81-90-9824-2970 (S.O.); Fax: +81-44-434-2336 (S.O.)
| |
Collapse
|
34
|
The Long and the Short of Current Nanomedicines for Treating Alzheimer's Disease. J Transl Int Med 2023; 10:294-296. [PMID: 36860633 PMCID: PMC9969564 DOI: 10.2478/jtim-2021-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
35
|
Alkahtani S, AL-Johani NS, Alarifi S. Mechanistic Insights, Treatment Paradigms, and Clinical Progress in Neurological Disorders: Current and Future Prospects. Int J Mol Sci 2023; 24:1340. [PMID: 36674852 PMCID: PMC9865061 DOI: 10.3390/ijms24021340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Neurodegenerative diseases (NDs) are a major cause of disability and are related to brain development. The neurological signs of brain lesions can vary from mild clinical shortfalls to more delicate and severe neurological/behavioral symptoms and learning disabilities, which are progressive. In this paper, we have tried to summarize a collective view of various NDs and their possible therapeutic outcomes. These diseases often occur as a consequence of the misfolding of proteins post-translation, as well as the dysfunctional trafficking of proteins. In the treatment of neurological disorders, a challenging hurdle to cross regarding drug delivery is the blood-brain barrier (BBB). The BBB plays a unique role in maintaining the homeostasis of the central nervous system (CNS) by exchanging components between the circulations and shielding the brain from neurotoxic pathogens and detrimental compounds. Here, we outline the current knowledge about BBB deterioration in the evolving brain, its origin, and therapeutic interventions. Additionally, we summarize the physiological scenarios of the BBB and its role in various cerebrovascular diseases. Overall, this information provides a detailed account of BBB functioning and the development of relevant treatments for neurological disorders. This paper will definitely help readers working in the field of neurological scientific communities.
Collapse
Affiliation(s)
- Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
36
|
Wang ML, Zou QQ, Sun Z, Wei XE, Li PY, Wu X, Li YH. Associations of MRI-visible perivascular spaces with longitudinal cognitive decline across the Alzheimer's disease spectrum. Alzheimers Res Ther 2022; 14:185. [PMID: 36514127 PMCID: PMC9746143 DOI: 10.1186/s13195-022-01136-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the characteristics and associations of MRI-visible perivascular spaces (PVS) with clinical progression and longitudinal cognitive decline across the Alzheimer's disease spectrum. METHODS We included 1429 participants (641 [44.86%] female) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. PVS number and grade in the centrum semiovale (CSO-PVS), basal ganglia (BG-PVS), and hippocampus (HP-PVS) were compared among the control (CN), mild cognitive impairment (MCI), and Alzheimer's disease (AD) groups. PVS were tested as predictors of diagnostic progression (i.e., CN to MCI/AD or MCI to AD) and longitudinal changes in the 13-item Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-Cog 13), Mini-Mental State Examination (MMSE), memory (ADNI-MEM), and executive function (ADNI-EF) using multiple linear regression, linear mixed-effects, and Cox proportional hazards modeling. RESULTS Compared with CN subjects, MCI and AD subjects had more CSO-PVS, both in number (p < 0.001) and grade (p < 0.001). However, there was no significant difference in BG-PVS and HP-PVS across the AD spectrum (p > 0.05). Individuals with moderate and frequent/severe CSO-PVS had a higher diagnostic conversion risk than individuals with no/mild CSO-PVS (log-rank p < 0.001 for all) in the combined CN and MCI group. Further Cox regression analyses revealed that moderate and frequent/severe CSO-PVS were associated with a higher risk of diagnostic conversion (HR = 2.007, 95% CI = 1.382-2.914, p < 0.001; HR = 2.676, 95% CI = 1.830-3.911, p < 0.001, respectively). A higher CSO-PVS number was associated with baseline cognitive performance and longitudinal cognitive decline in all cognitive tests (p < 0.05 for all). CONCLUSIONS CSO-PVS were more common in MCI and AD and were associated with cognitive decline across the AD spectrum.
Collapse
Affiliation(s)
- Ming-Liang Wang
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, 200233, China
| | - Qiao-Qiao Zou
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, 200233, China
| | - Zheng Sun
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, 200233, China
| | - Xiao-Er Wei
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, 200233, China
| | - Peng-Yang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Xue Wu
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yue-Hua Li
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600, Yi Shan Road, Shanghai, 200233, China.
| |
Collapse
|
37
|
Luo SM, Li LY, Guo LZ, Wang L, Wang YF, Chen N, Wang E. Dexmedetomidine exerts an anti-inflammatory effect via α2 adrenoceptors to alleviate cognitive dysfunction in 5xFAD mice. Front Aging Neurosci 2022; 14:978768. [PMID: 36204551 PMCID: PMC9531904 DOI: 10.3389/fnagi.2022.978768] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/29/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Inflammation promotes the progression of Alzheimer’s disease (AD). In this study, we explored the effect of dexmedetomidine on inflammation and cognitive function in a mouse model of AD. Methods: 5xFAD mice were intragastrically administered saline, dexmedetomidine, or dexmedetomidine and yohimbine for 14 days. The effects of dexmedetomidine on the acquisition and retention of memory in the Morris water-maze test and Y maze were evaluated. The deposition of amyloid beta protein (Abeta) and cytokine levels in the hippocampus were assessed. The expression of Bace1 protein and NFκB-p65 protein was assessed by Western blotting. Results: Compared with WT mice, 5xFAD mice exhibited cognitive impairment in the Morris water maze test and Y maze test. Cognitive decline was alleviated by dexmedetomidine and this was reversed by the α2 adrenoceptor antagonist yohimbine. Compared with saline treatment, dexmedetomidine led to a reduction in the Abeta deposition area (p < 0.05) and in the mean gray value (p < 0.01) in the hippocampus of 5xFAD mice. Compared with saline treatment, dexmedetomidine inhibited the activation of astrocytes and microglia in the hippocampal DG of 5xFAD mice and reduced the area of GFAP (p < 0.01) and IBA1 (p < 0.01). The level of IL-1β in the hippocampus decreased significantly after dexmedetomidine treatment compared with saline treatment in 5xFAD mice (p < 0.01). Yohimbine neutralized the effects of dexmedetomidine. Dexmedetomidine inhibited the expression of BACE1 and NF-κB p65 (p < 0.01), and these changes were reversed by yohimbine treatment. Conclusion: Dexmedetomidine alleviates cognitive decline, inhibits neuroinflammation, and prevents the deposition of Abeta in 5xFAD mice. The effect is mediated by the α2 adrenoceptor-mediated anti-inflammatory pathway. Dexmedetomidine may be effective for the treatment of AD and a better choice for the sedation of AD.
Collapse
Affiliation(s)
- Su-mei Luo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Long-yan Li
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
| | - Li-zhe Guo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Lu Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Yan-feng Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Na Chen
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - E. Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
- *Correspondence: E. Wang
| |
Collapse
|
38
|
Efficacy of Donepezil Hydrochloride plus Olanzapine for Senile Dementia and Its Effect on the Recovery of Cognitive Function. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4156312. [PMID: 35990821 PMCID: PMC9388230 DOI: 10.1155/2022/4156312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/14/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022]
Abstract
Objective To investigate the efficacy of donepezil hydrochloride plus olanzapine for senile dementia and its effect on the recovery of cognitive function. Methods A total of 60 patients with senile dementia admitted to our hospital from April 2020 to July 2021 were recruited and assigned to receive either olanzapine alone (observation group) or donepezil hydrochloride plus olanzapine (experimental group) via the random number table method, with 30 patients in each group. Results The combined therapy resulted in significantly higher clinical efficacy versus monotherapy of olanzapine (P < 0.05). Before treatment, the difference in the scores of cognitive function between the two groups did not come up to the statistical standard (P > 0.05). Donepezil hydrochloride plus olanzapine was associated with significantly higher scores of cognitive function in patients versus olanzapine alone (P < 0.05). The two groups had a similar incidence of adverse reactions (P > 0.05). Conclusion Donepezil hydrochloride plus olanzapine substantially enhances the recovery of cognitive function of patients with senile dementia and features a manageable safety. Further trials are, however, required prior to clinical promotion.
Collapse
|
39
|
Trinh PNH, Baltos JA, Hellyer SD, May LT, Gregory KJ. Adenosine receptor signalling in Alzheimer’s disease. Purinergic Signal 2022; 18:359-381. [PMID: 35870032 PMCID: PMC9391555 DOI: 10.1007/s11302-022-09883-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/02/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common dementia in the elderly and its increasing prevalence presents treatment challenges. Despite a better understanding of the disease, the current mainstay of treatment cannot modify pathogenesis or effectively address the associated cognitive and memory deficits. Emerging evidence suggests adenosine G protein-coupled receptors (GPCRs) are promising therapeutic targets for Alzheimer’s disease. The adenosine A1 and A2A receptors are expressed in the human brain and have a proposed involvement in the pathogenesis of dementia. Targeting these receptors preclinically can mitigate pathogenic β-amyloid and tau neurotoxicity whilst improving cognition and memory. In this review, we provide an accessible summary of the literature on Alzheimer’s disease and the therapeutic potential of A1 and A2A receptors. Although there are no available medicines targeting these receptors approved for treating dementia, we provide insights into some novel strategies, including allosterism and the targeting of oligomers, which may increase drug discovery success and enhance the therapeutic response.
Collapse
Affiliation(s)
- Phuc N. H. Trinh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Shane D. Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
| | - Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Parkville, 3052 Australia
| |
Collapse
|
40
|
Islam F, Nafady MH, Islam MR, Saha S, Rashid S, Akter A, Or-Rashid MH, Akhtar MF, Perveen A, Md Ashraf G, Rahman MH, Hussein Sweilam S. Resveratrol and neuroprotection: an insight into prospective therapeutic approaches against Alzheimer's disease from bench to bedside. Mol Neurobiol 2022; 59:4384-4404. [PMID: 35545730 DOI: 10.1007/s12035-022-02859-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and cognitive impairment; yet, there is currently no treatment. A buildup of Aβ, tau protein phosphorylation, oxidative stress, and inflammation in AD is pathogenic. The accumulation of amyloid-beta (Aβ) peptides in these neurocognitive areas is a significant characteristic of the disease. Therefore, inhibiting Aβ peptide aggregation has been proposed as the critical therapeutic approach for AD treatment. Resveratrol has been demonstrated in multiple studies to have a neuroprotective, anti-inflammatory, and antioxidant characteristic and the ability to minimize Aβ peptides aggregation and toxicity in the hippocampus of Alzheimer's patients, stimulating neurogenesis and inhibiting hippocampal degeneration. Furthermore, resveratrol's antioxidant effect promotes neuronal development by activating the silent information regulator-1 (SIRT1), which can protect against the detrimental effects of oxidative stress. Resveratrol-induced SIRT1 activation is becoming more crucial in developing novel therapeutic options for AD and other diseases that have neurodegenerative characteristics. This review highlighted a better knowledge of resveratrol's mechanism of action and its promising therapeutic efficacy in treating AD. We also highlighted the therapeutic potential of resveratrol as an AD therapeutic agent, which is effective against neurodegenerative disorders.
Collapse
Grants
- This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia, as well as Egyptian Russian University, Badr City, Egypt.
- Al-Kharj This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia, as well as Egyptian Russian University, Badr City, Egypt.
- Saudi Arabia This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia, as well as Egyptian Russian University, Badr City, Egypt.
- as well as Egyptian Russian University This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia, as well as Egyptian Russian University, Badr City, Egypt.
- Badr City This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia, as well as Egyptian Russian University, Badr City, Egypt.
- Egypt. This publication was supported by the Deanship of Scientific Research at Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia, as well as Egyptian Russian University, Badr City, Egypt.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mohamed H Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza, Egypt
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Susmita Saha
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Salma Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Harun- Or-Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore Campus, Lahore, Pakistan
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Uttar Pradesh, Mirzapur Pole, Saharanpur, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka, 1213, Bangladesh
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Gangwon-do, Wonju, 26426, South Korea
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City 11829, Egypt.
| |
Collapse
|
41
|
Kang N, Luan Y, Jiang Y, Cheng W, Liu Y, Su Z, Liu Y, Tan P. Neuroprotective Effects of Oligosaccharides in Rehmanniae Radix on Transgenic Caenorhabditis elegans Models for Alzheimer’s Disease. Front Pharmacol 2022; 13:878631. [PMID: 35784741 PMCID: PMC9247152 DOI: 10.3389/fphar.2022.878631] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
Rehmanniae Radix (RR, the dried tuberous roots of Rehmannia glutinosa (Gaertn.) DC.) is an important traditional Chinese medicine distributed in Henan, Hebei, Inner Mongolia, and Northeast in China. RR is frequently used to treat diabetes mellitus, cardiovascular disease, osteoporosis and aging-related diseases in a class of prescriptions. The oligosaccharides and catalpol in RR have been confirmed to have neuroprotective effects. However, there are few studies on the anti-Alzheimer’s disease (AD) effect of oligosaccharides in Rehmanniae Radix (ORR). The chemical components and pharmacological effects of dried Rehmannia Radix (DRR) and prepared Rehmannia Radix (PRR) are different because of the different processing methods. ORR has neuroprotective potential, such as improving learning and memory in rats. Therefore, this study aimed to prove the importance of oligosaccharides in DRR (ODRR) and PRR (OPRR) for AD based on the Caenorhabditis elegans (C. elegans) model and the different roles of ODRR and OPRR in the treatment of AD. In this study, we used paralysis assays, lifespan and stress resistance assays, bacterial growth curve, developmental and behavioral parameters, and ability of learning and memory to explore the effects of ODRR and OPRR on anti-AD and anti-aging. Furthermore, the accumulation of reactive oxygen species (ROS); deposition of Aβ; and expression of amy-1, sir-2.1, daf-16, sod-3, skn-1, and hsp-16.2 were analyzed to confirm the efficacy of ODRR and OPRR. OPRR was more effective than ODRR in delaying the paralysis, improving learning ability, and prolonging the lifespan of C. elegans. Further mechanism studies showed that the accumulation of ROS, aggregation, and toxicity of Aβ were reduced, suggesting that ORR alleviated Aβ-induced toxicity, in part, through antioxidant activity and Aβ aggregation inhibiting. The expression of amy-1 was downregulated, and sir-2.1, daf-16, sod-3, and hsp-16.2 were upregulated. Thus, ORR could have a possible therapeutic effect on AD by modulating the expression of amy-1, sir-2.1, daf-16, sod-3, and hsp-16.2. Furthermore, ORR promoted the nuclear localization of daf-16 and further increased the expression of sod-3 and hsp-16.2, which significantly contributed to inhibiting the Aβ toxicity and enhancing oxidative stress resistance. In summary, the study provided a new idea for the development of ORR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peng Tan
- *Correspondence: Yonggang Liu, ; Peng Tan,
| |
Collapse
|
42
|
|
43
|
Yang B, He M, Chen X, Sun M, Pan T, Xu X, Zhang X, Gong Q, Zhao Y, Jin Z, Cheng Z. Acupuncture Effect Assessment in APP/PS1 Transgenic Mice: On Regulating Learning-Memory Abilities, Gut Microbiota, and Microbial Metabolites. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1527159. [PMID: 35432583 PMCID: PMC9012623 DOI: 10.1155/2022/1527159] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a brain illness that affects learning and memory capacities over time. In recent investigations, acupuncture has been shown to be an effective alternative treatment for AD. We investigated the effect of acupuncture on learning and memory abilities using a water maze in APP/PS1 transgenic mice. The amounts of Aβ and tau protein in mice's hippocampal tissue were determined using Western blot. The levels of IL-1β, IL-10, LPS and TNF-α in mice's serum were measured using ELISA. The variations of gut microbiota in mice's feces were determined using the 16SrDNA technique, and the metabolites were examined using a untargeted metabolomics methodology. The results showed that acupuncture treatment improved mice's learning and memory abilities substantially. Acupuncture therapy regulated the Aβ and tau protein concentration as well as the levels of IL-10 and LPS. Acupuncture treatment influenced the mouse microbiota and metabolites and had been linked to six biochemical pathways. This study adds to our understanding of the effect of acupuncture on AD and opens the door to further research into the alterations of intestinal bacteria in the presence of AD.
Collapse
Affiliation(s)
- Bo Yang
- Liaoning University of Traditional Chinese Medicine, No. 79, Chongshan Eastern Rd, Huanggu District, 110847 Shenyang, China
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Min He
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Xinhua Chen
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Mengmeng Sun
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Ting Pan
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Xiaohong Xu
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Xuesong Zhang
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Qing Gong
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Ye Zhao
- Northwest University, No. 229, Taibai North Road, Xi' an 710069, China
| | - Ziqi Jin
- Changchun University of Chinese Medicine, No. 1035, Boshuo Rd, Jingyue Economic Development District, 130117 Changchun, China
| | - Zedong Cheng
- Liaoning University of Traditional Chinese Medicine, No. 79, Chongshan Eastern Rd, Huanggu District, 110847 Shenyang, China
| |
Collapse
|
44
|
Kou JJ, Shi JZ, He YY, Hao JJ, Zhang HY, Luo DM, Song JK, Yan Y, Xie XM, Du GH, Pang XB. Luteolin alleviates cognitive impairment in Alzheimer's disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol Sin 2022; 43:840-849. [PMID: 34267346 DOI: 10.1038/s41401-021-00702-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
Luteolin is a flavonoid in a variety of fruits, vegetables, and herbs, which has shown anti-inflammatory, antioxidant, and anti-cancer neuroprotective activities. In this study, we investigated the potential beneficial effects of luteolin on memory deficits and neuroinflammation in a triple-transgenic mouse model of Alzheimer's disease (AD) (3 × Tg-AD). The mice were treated with luteolin (20, 40 mg · kg-1 · d-1, ip) for 3 weeks. We showed that luteolin treatment dose-dependently improved spatial learning, ameliorated memory deficits in 3 × Tg-AD mice, accompanied by inhibiting astrocyte overactivation (GFAP) and neuroinflammation (TNF-α, IL-1β, IL-6, NO, COX-2, and iNOS protein), and decreasing the expression of endoplasmic reticulum (ER) stress markers GRP78 and IRE1α in brain tissues. In rat C6 glioma cells, treatment with luteolin (1, 10 µM) dose-dependently inhibited LPS-induced cell proliferation, excessive release of inflammatory cytokines, and increase of ER stress marker GRP78. In conclusion, luteolin is an effective agent in the treatment of learning and memory deficits in 3 × Tg-AD mice, which may be attributable to the inhibition of ER stress in astrocytes and subsequent neuroinflammation. These results provide the experimental basis for further research and development of luteolin as a therapeutic agent for AD.
Collapse
|
45
|
Gong B, Ji W, Chen X, Li P, Cheng W, Zhao Y, He B, Zhuang J, Gao J, Yin Y. Recent Advancements in Strategies for Abnormal Protein Clearance in Alzheimer's Disease. Mini Rev Med Chem 2022; 22:2260-2270. [PMID: 35156576 DOI: 10.2174/1389557522666220214092824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/06/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
:
Alzheimer's disease (AD) is a intricate neurodegenerative disease with chronic and progressive development whose typical neuropathological features encompasses senile plaques and neurofibrillary tangles respectively formed by the extracellular deposition of amyloid-beta (Aβ) and the intracellular accumulation of hyperphosphorylated tau protein in the brain, particularly in limbic and cortical regions. The pathological changes are considered to be caused by the loss of Aβ and tau protein clearance mechanisms under pathological conditions, which leads to an imbalance between the rates of clearance and production. Consequently, the main strategies for treating AD aim to reduce the production of Aβ and hyperphosphorylated tau protein in the brain, inhibit their accumulation, or accelerate their clearance. Although drugs utilizing these therapeutic strategies have been studied successively, their therapeutic effects have generally been less than ideal. Fortunately, recent advances have been made in clearance strategies for these abnormally expressed proteins, including immunotherapies and nanomedicines targeting Aβ or tau, which could represent an important breakthrough for treating AD. Here, we review recent development of the strategies for the removal of abnormal proteins and provide new ideas and methods for treating AD.
Collapse
Affiliation(s)
- Baofeng Gong
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Wenbo Ji
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Xiaohan Chen
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Peng Li
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Wenbin Cheng
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Yuchen Zhao
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Bin He
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Jianhua Zhuang
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - Jie Gao
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| | - You Yin
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China; b Institute of Translational Medicine, Shanghai University, Shanghai, 200444, Chin
| |
Collapse
|
46
|
Zhu ZY, Liu YD, Gong Y, Jin W, Topchiy E, Turdi S, Gao YF, Culver B, Wang SY, Ge W, Zha WL, Ren J, Pei ZH, Qin X. Mitochondrial aldehyde dehydrogenase (ALDH2) rescues cardiac contractile dysfunction in an APP/PS1 murine model of Alzheimer's disease via inhibition of ACSL4-dependent ferroptosis. Acta Pharmacol Sin 2022; 43:39-49. [PMID: 33767380 PMCID: PMC8724276 DOI: 10.1038/s41401-021-00635-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/26/2021] [Indexed: 02/01/2023]
Abstract
Alzheimer's disease (AD) is associated with high incidence of cardiovascular events but the mechanism remains elusive. Our previous study reveals a tight correlation between cardiac dysfunction and low mitochondrial aldehyde dehydrogenase (ALDH2) activity in elderly AD patients. In the present study we investigated the effect of ALDH2 overexpression on cardiac function in APP/PS1 mouse model of AD. Global ALDH2 transgenic mice were crossed with APP/PS1 mutant mice to generate the ALDH2-APP/PS1 mutant mice. Cognitive function, cardiac contractile, and morphological properties were assessed. We showed that APP/PS1 mice displayed significant cognitive deficit in Morris water maze test, myocardial ultrastructural, geometric (cardiac atrophy, interstitial fibrosis) and functional (reduced fractional shortening and cardiomyocyte contraction) anomalies along with oxidative stress, apoptosis, and inflammation in myocardium. ALDH2 transgene significantly attenuated or mitigated these anomalies. We also noted the markedly elevated levels of lipid peroxidation, the essential lipid peroxidation enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4), the transcriptional regulator for ACLS4 special protein 1 (SP1) and ferroptosis, evidenced by elevated NCOA4, decreased GPx4, and SLC7A11 in myocardium of APP/PS1 mutant mice; these effects were nullified by ALDH2 transgene. In cardiomyocytes isolated from WT mice and in H9C2 myoblasts in vitro, application of Aβ (20 μM) decreased cell survival, compromised cardiomyocyte contractile function, and induced lipid peroxidation; ALDH2 transgene or activator Alda-1 rescued Aβ-induced deteriorating effects. ALDH2-induced protection against Aβ-induced lipid peroxidation was mimicked by the SP1 inhibitor tolfenamic acid (TA) or the ACSL4 inhibitor triacsin C (TC), and mitigated by the lipid peroxidation inducer 5-hydroxyeicosatetraenoic acid (5-HETE) or the ferroptosis inducer erastin. These results demonstrate an essential role for ALDH2 in AD-induced cardiac anomalies through regulation of lipid peroxidation and ferroptosis.
Collapse
Affiliation(s)
- Zhi-Yun Zhu
- Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, China
| | - Yan-Dong Liu
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, 330009, China
| | - Yan Gong
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, 330009, China
| | - Wei Jin
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, 330009, China
| | - Elena Topchiy
- University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Subat Turdi
- University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Yue-Feng Gao
- University of Wyoming College of Health Sciences, Laramie, WY, USA
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100083, China
| | - Bruce Culver
- University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Shu-Yi Wang
- University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Wei Ge
- Department of General Practice, Xijing Hospital, the Air Force Military Medical University, Xi'an, 710032, China
| | - Wen-Liang Zha
- Department of Surgery, Clinic Medical College, Hubei University of Science and Technology, Xianning, 437100, China
- National Demonstration Center for Experimental General Medicine Education, Hubei University of Science and Technology, Xianning, 437100, China
| | - Jun Ren
- University of Wyoming College of Health Sciences, Laramie, WY, USA.
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
| | - Zhao-Hui Pei
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, 330009, China.
| | - Xing Qin
- University of Wyoming College of Health Sciences, Laramie, WY, USA.
- Department of Cardiology, Xijing Hospital, the Air Force Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
47
|
OUP accepted manuscript. Brain 2022; 145:2250-2275. [DOI: 10.1093/brain/awac096] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/13/2022] Open
|
48
|
Qiu-Yue X, Tian-Yuan Y, Xiao-Long W, Dong-Mei Q, Xiao-Rui C. Effects of Metformin on Modulating the Expression of Brain-related Genes of APP/PS1 Transgenic Mice based on Single Cell Sequencing. Curr Alzheimer Res 2022; 19:754-771. [PMID: 36464874 DOI: 10.2174/1567205020666221201143323] [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: 08/02/2022] [Revised: 10/26/2022] [Accepted: 11/12/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND Alzheimer's disease is the most common form of dementia, affecting millions of people worldwide. METHODS Here, we analyzed the effects of metformin on APP/PS1 transgenic mice by behavioral test and single-cell sequencing. RESULTS It showed that metformin can improve the spatial learning, memory function, and anxiety mood of APP/PS1 transgenic mice. We identified transcriptionally distinct subpopulations of nine major brain cell types. Metformin increased the differentiation of stem cells, decreased the proportion of cells in the G2 phase, enhanced the generation of neural stem cells and oligodendrocyte progenitor cells, and the tendency of neural stem cells to differentiate into astrocytes. Notably, 253 genes expressed abnormally in APP/PS1 transgenic mice and were reversed by metformin. Ttr, Uba52, and Rps21 are the top 3 genes in the cell-gene network with the highest node degree. Moreover, histochemistry showed the expressions of RPS15, Uba52, and RPL23a were consistent with the data from single-cell sequencing. Pathway and biological process enrichment analysis indicated metformin was involved in nervous system development and negative regulation of the apoptotic process. CONCLUSION Overall, metformin might play an important role in the differentiation and development and apoptotic process of the central nervous system by regulating the expression of Ttr, Uba52, Rps21, and other genes to improve cognition of APP/PS1 transgenic mice. These results provided a clue for elaborating on the molecular and cellular basis of metformin on AD.
Collapse
Affiliation(s)
- Xiao Qiu-Yue
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ye Tian-Yuan
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wang Xiao-Long
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qi Dong-Mei
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Cheng Xiao-Rui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
49
|
The Potential Role of Cytokines and Growth Factors in the Pathogenesis of Alzheimer's Disease. Cells 2021; 10:cells10102790. [PMID: 34685770 PMCID: PMC8534363 DOI: 10.3390/cells10102790] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most prominent neurodegenerative diseases, which impairs cognitive function in afflicted individuals. AD results in gradual decay of neuronal function as a consequence of diverse degenerating events. Several neuroimmune players (such as cytokines and growth factors that are key players in maintaining CNS homeostasis) turn aberrant during crosstalk between the innate and adaptive immunities. This aberrance underlies neuroinflammation and drives neuronal cells toward apoptotic decline. Neuroinflammation involves microglial activation and has been shown to exacerbate AD. This review attempted to elucidate the role of cytokines, growth factors, and associated mechanisms implicated in the course of AD, especially with neuroinflammation. We also evaluated the propensities and specific mechanism(s) of cytokines and growth factors impacting neuron upon apoptotic decline and further shed light on the availability and accessibility of cytokines across the blood-brain barrier and choroid plexus in AD pathophysiology. The pathogenic and the protective roles of macrophage migration and inhibitory factors, neurotrophic factors, hematopoietic-related growth factors, TAU phosphorylation, advanced glycation end products, complement system, and glial cells in AD and neuropsychiatric pathology were also discussed. Taken together, the emerging roles of these factors in AD pathology emphasize the importance of building novel strategies for an effective therapeutic/neuropsychiatric management of AD in clinics.
Collapse
|
50
|
Wang XQ, Zhou LY, Tan RX, Liang GP, Fang SX, Li W, Xie M, Wen YH, Wu JQ, Chen YP. Design, Synthesis, and Evaluation of Chalcone Derivatives as Multifunctional Agents against Alzheimer's Disease. Chem Biodivers 2021; 18:e2100341. [PMID: 34510699 DOI: 10.1002/cbdv.202100341] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/09/2021] [Indexed: 01/08/2023]
Abstract
Fifteen chalcone derivatives 3a-3o were synthesized, and evaluated as multifunctional agents against Alzheimer's disease. In vitro studies revealed that these compounds inhibited self-induced Aβ1-42 aggregation effectively ranged from 45.9-94.5 % at 20 μM, and acted as potential antioxidants. Their structure-activity relationships were summarized. In particular, (2E)-3-[4-(dimethylamino)phenyl]-1-(pyridin-2-yl)prop-2-en-1-one (3g) exhibited an excellent inhibitory activity of 94.5 % at 20 μM, and it could disassemble the self-induced Aβ1-42 aggregation fibrils with ratio of 57.1 % at 20 μM concentration. In addition, compound 3g displayed good chelating ability for Cu2+ , and could effectively inhibit and disaggregate Cu2+ -induced Aβ aggregation. Moreover, compound 3g exerted low cytotoxicity, significantly reversed Aβ1-42 -induced SH-SY5Y cell damage. More importantly, compound 3g remarkably ameliorated scopolamine-induced memory impairment in mice. In summary, all the results revealed compound 3g was a potential multifunctional agent for AD therapy.
Collapse
Affiliation(s)
- Xiao-Qin Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Lu-Yi Zhou
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Ren-Xian Tan
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Guo-Peng Liang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Si-Xian Fang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Wei Li
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Mei Xie
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yu-Hao Wen
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jia-Qiang Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China
| | - Yi-Ping Chen
- School of Pharmaceutical Sciences, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| |
Collapse
|