1
|
Li T, Xing HM, Qian HD, Gao Q, Xu SL, Ma H, Chi ZL. Small extracellular vesicles derived from human induced pluripotent stem cell-differentiated neural progenitor cells mitigate retinal ganglion cell degeneration in a mouse model of optic nerve injury. Neural Regen Res 2025; 20:587-597. [PMID: 38819069 PMCID: PMC11317950 DOI: 10.4103/nrr.nrr-d-23-01414] [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: 08/22/2023] [Revised: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00034/figure1/v/2024-05-28T214302Z/r/image-tiff Several studies have found that transplantation of neural progenitor cells (NPCs) promotes the survival of injured neurons. However, a poor integration rate and high risk of tumorigenicity after cell transplantation limits their clinical application. Small extracellular vesicles (sEVs) contain bioactive molecules for neuronal protection and regeneration. Previous studies have shown that stem/progenitor cell-derived sEVs can promote neuronal survival and recovery of neurological function in neurodegenerative eye diseases and other eye diseases. In this study, we intravitreally transplanted sEVs derived from human induced pluripotent stem cells (hiPSCs) and hiPSCs-differentiated NPCs (hiPSC-NPC) in a mouse model of optic nerve crush. Our results show that these intravitreally injected sEVs were ingested by retinal cells, especially those localized in the ganglion cell layer. Treatment with hiPSC-NPC-derived sEVs mitigated optic nerve crush-induced retinal ganglion cell degeneration, and regulated the retinal microenvironment by inhibiting excessive activation of microglia. Component analysis further revealed that hiPSC-NPC derived sEVs transported neuroprotective and anti-inflammatory miRNA cargos to target cells, which had protective effects on RGCs after optic nerve injury. These findings suggest that sEVs derived from hiPSC-NPC are a promising cell-free therapeutic strategy for optic neuropathy.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hui-Min Xing
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hai-Dong Qian
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qiao Gao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Sheng-Lan Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hua Ma
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zai-Long Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Rodríguez-Fernández L, Zorzo C, Arias JL. Photobiomodulation in the aging brain: a systematic review from animal models to humans. GeroScience 2024; 46:6583-6623. [PMID: 38861125 PMCID: PMC11493890 DOI: 10.1007/s11357-024-01231-y] [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: 02/28/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
Aging is a multifactorial biological process that may be associated with cognitive decline. Photobiomodulation (PBM) is a non-pharmacological therapy that shows promising results in the treatment or prevention of age-related cognitive impairments. The aim of this review is to compile the preclinical and clinical evidence of the effect of PBM during aging in healthy and pathological conditions, including behavioral analysis and neuropsychological assessment, as well as brain-related modifications. 37 studies were identified by searching in PubMed, Scopus, and PsycInfo databases. Most studies use wavelengths of 800, 810, or 1064 nm but intensity and days of application were highly variable. In animal studies, it has been shown improvements in spatial memory, episodic-like memory, social memory, while different results have been found in recognition memory. Locomotor activity improved in Parkinson disease models. In healthy aged humans, it has been outlined improvements in working memory, cognitive inhibition, and lexical/semantic access, while general cognition was mainly enhanced on Alzheimer disease or mild cognitive impairment. Anxiety assessment is scarce and shows mixed results. As for brain activity, results outline promising effects of PBM in reversing metabolic alterations and enhancing mitochondrial function, as evidenced by restored CCO activity and ATP levels. Additionally, PBM demonstrated neuroprotective, anti-inflammatory, immunomodulatory and hemodynamic effects. The findings suggest that PBM holds promise as a non-invasive intervention for enhancing cognitive function, and in the modulation of brain functional reorganization. It is necessary to develop standardized protocols for the correct, beneficial, and homogeneous use of PBM.
Collapse
Affiliation(s)
| | - Candela Zorzo
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain.
| | - Jorge L Arias
- ISPA, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
3
|
Lacerda RAV, Desio JAF, Kammers CM, Henkes S, Freitas de Sá M, de Souza EF, da Silva DM, Teixeira Pinheiro Gusmão C, Santos JCCD. Sleep disorders and risk of alzheimer's disease: A two-way road. Ageing Res Rev 2024; 101:102514. [PMID: 39317268 DOI: 10.1016/j.arr.2024.102514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Substantial sleep impairment in patients with Alzheimer's disease (AD) is one of the emerging points for continued efforts to better understand the disease. Individuals without cognitive decline, an important marker of the clinical phase of AD, may show early alterations in the sleep-wake cycle. The objective of this critical narrative review is to explore the bidirectional pathophysiological correlation between sleep disturbances and Alzheimer's Disease. Specifically, it examines how the disruption of sleep homeostasis in individuals without dementia could contribute to the pathogenesis of AD, and conversely, how neurodegeneration in individuals with Alzheimer's Disease might lead to dysregulation of the sleep-wake cycle. Recent scientific results indicate that sleep disturbances, particularly those related to impaired glymphatic clearance, may act as an important mechanism associated with the genesis of Alzheimer's Disease. Additionally, amyloid deposition and tau protein hyperphosphorylation, along with astrocytic hyperactivation, appear to trigger changes in neurotransmission dynamics in areas related to sleep, which may explain the onset of sleep disturbances in individuals with AD. Disruption of sleep homeostasis appears to be a modifiable risk factor in Alzheimer's disease. Whenever possible, the use of non-pharmacological strategies becomes important in this context. From a different perspective, additional research is needed to understand and treat the dysfunction of the sleep-wake cycle in individuals already affected by AD. Early recognition and correction of sleep disturbances in this population could potentially mitigate the progression of dementia and improve the quality of life for those with AD.
Collapse
Affiliation(s)
| | | | | | - Silvana Henkes
- Lutheran University of Brazil - ULBRA, Carazinho, RS, Brazil
| | | | | | | | | | - Júlio César Claudino Dos Santos
- Medical School of the Christus University Center - UNICHRISTUS, Fortaleza, CE, Brazil; Post-Graduate Program of Morphofunctional Sciences, Federal University of Ceara, Fortaleza, CE, Brazil; Unifacvest University Center - UNIFACVEST, Lages, SC, Brazil.
| |
Collapse
|
4
|
Singh I, Anand S, Gowda DJ, Kamath A, Singh AK. Caloric restriction mimetics improve gut microbiota: a promising neurotherapeutics approach for managing age-related neurodegenerative disorders. Biogerontology 2024; 25:899-922. [PMID: 39177917 PMCID: PMC11486790 DOI: 10.1007/s10522-024-10128-4] [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: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
The gut microbiota (GM) produces various molecules that regulate the physiological functionality of the brain through the gut-brain axis (GBA). Studies suggest that alteration in GBA may lead to the onset and progression of various neurological dysfunctions. Moreover, aging is one of the prominent causes that contribute to the alteration of GBA. With age, GM undergoes a shift in population size and species of microflora leading to changes in their secreted metabolites. These changes also hamper communications among the HPA (hypothalamic-pituitary-adrenal), ENS (enteric nervous system), and ANS (autonomic nervous system). A therapeutic intervention that has recently gained attention in improving health and maintaining communication between the gut and the brain is calorie restriction (CR), which also plays a critical role in autophagy and neurogenesis processes. However, its strict regime and lifelong commitment pose challenges. The need is to produce similar beneficial effects of CR without having its rigorous compliance. This led to an exploration of calorie restriction mimetics (CRMs) which could mimic CR's functions without limiting diet, providing long-term health benefits. CRMs ensure the efficient functioning of the GBA through gut bacteria and their metabolites i.e., short-chain fatty acids, bile acids, and neurotransmitters. This is particularly beneficial for elderly individuals, as the GM deteriorates with age and the body's ability to digest the toxic accumulates declines. In this review, we have explored the beneficial effect of CRMs in extending lifespan by enhancing the beneficial bacteria and their effects on metabolite production, physiological conditions, and neurological dysfunctions including neurodegenerative disorders.
Collapse
Affiliation(s)
- Ishika Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Shashi Anand
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Deepashree J Gowda
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Amitha Kamath
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India.
| |
Collapse
|
5
|
Chen L, Zhuang Z, Duan H, Lv D, Hong S, Chen P, He B, Shen Z. Corilagin improves cognitive impairment in APP/PS1 mice by reducing Aβ generation and enhancing synaptic plasticity. Eur J Pharmacol 2024; 981:176893. [PMID: 39134295 DOI: 10.1016/j.ejphar.2024.176893] [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: 02/24/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is closely associated with the neurotoxic effects of amyloid-β (Aβ), leading to synaptic damage, neuronal loss and cognitive dysfunction. Previous in vitro studies have demonstrated the potential of corilagin to counteract Aβ-induced oxidative stress, inflammatory injury, and β-site amyloid precursor protein cleaving enzyme-1 (BACE1) activity in Aβ production. However, the in vivo protective effects of corilagin on Alzheimer's disease remain unexplored. The purpose of this study was to investigate the protective effects of corilagin on APP/PS1 mice and the underlying mechanisms. The cognitive function of the mice was assessed by step-through passive avoidance and Morris water maze tests. Nissl staining was used to evaluate neuronal damage in the hippocampus. ELISA and Western blotting analyses were used to determine the associated protein expression. Transmission electron microscopy was utilized to observe the synaptic ultrastructure of hippocampal neurons. Golgi staining was applied to assess dendritic morphology and dendritic spine density in hippocampal pyramidal neurons. Immunohistochemistry and Western blotting were performed to examine the expression of synaptic-associated proteins. The results showed that corilagin improves learning and memory in APP/PS1 mice, reduces hippocampal neuron damage, inhibits BACE1 and reduces Aβ generation. It also improves synaptic plasticity and the expression of synaptic-associated proteins. Corilagin effectively reduces Aβ generation by inhibiting BACE1, ultimately reducing neuronal loss and enhancing synaptic plasticity to improve synaptic transmission. This study sheds light on the potential therapeutic role of corilagin in Alzheimer's disease.
Collapse
Affiliation(s)
- Linyi Chen
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Zhujun Zhuang
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Hengqian Duan
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Di Lv
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Shengxiong Hong
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China
| | - Peng Chen
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| | - Bo He
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| | - Zhiqiang Shen
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
6
|
Xie H, Wu F, Mao J, Wang Y, Zhu J, Zhou X, Hong K, Li B, Qiu X, Wen C. The role of microglia in neurological diseases with involvement of extracellular vesicles. Neurobiol Dis 2024; 202:106700. [PMID: 39401551 DOI: 10.1016/j.nbd.2024.106700] [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: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
As a subset of mononuclear phagocytes in the central nervous system, microglia play a crucial role in immune defense and homeostasis maintenance. Microglia can regulate their states in response to specific signals of health and pathology. Microglia-mediated neuroinflammation is a pathological hallmark of neurodegenerative diseases, neurological damage and neurological tumors, underscoring its key immunoregulatory role in these conditions. Intriguingly, a substantial body of research has indicated that extracellular vesicles can mediate intercellular communication by transporting cargoes from parental cells, a property that is also reflected in microenvironmental signaling networks involving microglia. Based on the microglial characteristics, we briefly outline the biological features of extracellular vesicles and focus on summarizing the integrative role played by microglia in the maintenance of nervous system homeostasis and progression of different neurological diseases. Extracellular vesicles may engage in the homeostasis maintenance and pathological process as a medium of intercellular communication. Here, we aim to provide new insights for further exploration of neurological disease pathogenesis, which may provide theoretical foundations for cell-free therapies.
Collapse
Affiliation(s)
- Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
7
|
Song Q, Li J, Li T, Li H. Nanomaterials that Aid in the Diagnosis and Treatment of Alzheimer's Disease, Resolving Blood-Brain Barrier Crossing Ability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403473. [PMID: 39101248 PMCID: PMC11481234 DOI: 10.1002/advs.202403473] [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: 05/06/2024] [Revised: 07/04/2024] [Indexed: 08/06/2024]
Abstract
As a form of dementia, Alzheimer's disease (AD) suffers from no efficacious cure, yet AD treatment is still imperative, as it ameliorates the symptoms or prevents it from deteriorating or maintains the current status to the longest extent. The human brain is the most sensitive and complex organ in the body, which is protected by the blood-brain barrier (BBB). This yet induces the difficulty in curing AD as the drugs or nanomaterials that are much inhibited from reaching the lesion site. Thus, BBB crossing capability of drug delivery system remains a significant challenge in the development of neurological therapeutics. Fortunately, nano-enabled delivery systems possess promising potential to achieve multifunctional diagnostics/therapeutics against various targets of AD owing to their intriguing advantages of nanocarriers, including easy multifunctionalization on surfaces, high surface-to-volume ratio with large payloads, and potential ability to cross the BBB, making them capable of conquering the limitations of conventional drug candidates. This review, which focuses on the BBB crossing ability of the multifunctional nanomaterials in AD diagnosis and treatment, will provide an insightful vision that is conducive to the development of AD-related nanomaterials.
Collapse
Affiliation(s)
- Qingting Song
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Junyou Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Ting Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Hung‐Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
8
|
Hussain M, Khan I, Chaudhary MN, Ali K, Mushtaq A, Jiang B, Zheng L, Pan Y, Hu J, Zou X. Phosphatidylserine: A comprehensive overview of synthesis, metabolism, and nutrition. Chem Phys Lipids 2024; 264:105422. [PMID: 39097133 DOI: 10.1016/j.chemphyslip.2024.105422] [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: 06/07/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Phosphatidylserine (PtdS) is classified as a glycerophospholipid and a primary anionic phospholipid and is particularly abundant in the inner leaflet of the plasma membrane in neural tissues. It is synthesized from phosphatidylcholine or phosphatidylethanolamine by exchanging the base head group with serine, and this reaction is catalyzed by PtdS synthase-1 and PtdS synthase-2 located in the endoplasmic reticulum. PtdS exposure on the outside surface of the cell is essential for eliminating apoptotic cells and initiating the blood clotting cascade. It is also a precursor of phosphatidylethanolamine, produced by PtdS decarboxylase in bacteria, yeast, and mammalian cells. Furthermore, PtdS acts as a cofactor for several necessary enzymes that participate in signaling pathways. Beyond these functions, several studies indicate that PtdS plays a role in various cerebral functions, including activating membrane signaling pathways, neuroinflammation, neurotransmission, and synaptic refinement associated with the central nervous system (CNS). This review discusses the occurrence of PtdS in nature and biosynthesis via enzymes and genes in plants, yeast, prokaryotes, mammalian cells, and the brain, and enzymatic synthesis through phospholipase D (PLD). Furthermore, we discuss metabolism, its role in the CNS, the fortification of foods, and supplementation for improving some memory functions, the results of which remain unclear. PtdS can be a potentially beneficial addition to foods for kids, seniors, athletes, and others, especially with the rising consumer trend favoring functional foods over conventional pills and capsules. Clinical studies have shown that PtdS is safe and well tolerated by patients.
Collapse
Affiliation(s)
- Mudassar Hussain
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Imad Khan
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Muneeba Naseer Chaudhary
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing, 400715, China
| | - Khubaib Ali
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Anam Mushtaq
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Bangzhi Jiang
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Lei Zheng
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yuechao Pan
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jijie Hu
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xiaoqiang Zou
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
9
|
Ghiasvand K, Amirfazli M, Moghimi P, Safari F, Takhshid MA. The role of neuron-like cell lines and primary neuron cell models in unraveling the complexity of neurodegenerative diseases: a comprehensive review. Mol Biol Rep 2024; 51:1024. [PMID: 39340590 DOI: 10.1007/s11033-024-09964-x] [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: 05/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs) are characterized by the progressive loss of neurons. As to developing effective therapeutic interventions, it is crucial to understand the underlying mechanisms of NDs. Cellular models have become invaluable tools for studying the complex pathogenesis of NDs, offering insights into disease mechanisms, determining potential therapeutic targets, and aiding in drug discovery. This review provides a comprehensive overview of various cellular models used in ND research, focusing on Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Cell lines, such as SH-SY5Y and PC12 cells, have emerged as valuable tools due to their ease of use, reproducibility, and scalability. Additionally, co-culture models, involving the growth of distinct cell types like neurons and astrocytes together, are highlighted for simulating brain interactions and microenvironment. While cell lines cannot fully replicate the complexity of the human brain, they provide a scalable method for examining important aspects of neurodegenerative diseases. Advancements in cell line technologies, including the incorporation of patient-specific genetic variants and improved co-culture models, hold promise for enhancing our understanding and expediting the development of effective treatments. Integrating multiple cellular models and advanced technologies offers the potential for significant progress in unraveling the intricacies of these debilitating diseases and improving patient outcomes.
Collapse
Affiliation(s)
- Kianoush Ghiasvand
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Amirfazli
- School of biological sciences, Illinois State University, Normal, United States of America
| | - Parvaneh Moghimi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Wang L, Yuan X, Cai Q, Chen Y, Jia Z, Mai Q, Liu J, Liu Y. Mitochondria-targeting Cu 2-xSe-TPP with dual enzyme activity alleviates Alzheimer's disease by modulating oxidative stress. Colloids Surf B Biointerfaces 2024; 245:114244. [PMID: 39366108 DOI: 10.1016/j.colsurfb.2024.114244] [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: 07/25/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024]
Abstract
Mitochondrial dysfunction in microglia has been implicated as a key pathogenesis of most neurodegenerative diseases including Alzheimer's disease (AD). Abnormal production of reactive oxygen species (ROS) and neuroinflammation caused by mitochondrial oxidative stress are important factors leading to neuronal death in AD. Herein, a "dual brake" strategy to synergistically halt mitochondrial dysfunction and neuroinflammation targeting mitochondria in microglia is proposed. To achieve this goal, (3-carboxypropyl) triphenyl-phosphonium bromide (TPP)-modified Cu2-xSe nanozymes (Cu2-xSe-TPP NPs) with dual enzyme-like activities was designed. Cu2-xSe-TPP NPs with superoxide dismutase-mimetic (SOD) and catalase-mimetic (CAT) activities can effectively scavenge ROS in the mitochondria of microglia and relieve mitochondrial oxidative stress. In vivo studies demonstrated that Cu2-xSe-TPP NPs can alleviate oxidative stress and promote neuroprotection in the hippocampus of AD model mice. In addition, Cu2-xSe-TPP NPs can regulate the polarization of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, promote Aβ phagocytosis and reshape the AD inflammatory microenvironment, thus effectively attenuating AD neuropathology and rescuing cognitive deficits in AD model mice. Taken together, this strategy preventing mitochondrial damage and remodeling the inflammatory microenvironment will provide a new perspective for AD therapy.
Collapse
Affiliation(s)
- Liqiang Wang
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen 518110, China; College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China
| | - Xiaoyu Yuan
- College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China
| | - Qianyu Cai
- College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China
| | - Yutong Chen
- College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China
| | - Zhi Jia
- College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China
| | - Qiongmei Mai
- College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China
| | - Jie Liu
- College of Chemistry and Materials Science of Jinan University, Guangzhou 510632, China.
| | - Yanan Liu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen 518110, China.
| |
Collapse
|
11
|
Lin S, Xue M, Sun J, Xu C, Wang T, Lian J, Lv M, Yang P, Sheng C, Cheng Z, Wang W. MRI Radiomics Nomogram for Predicting Disease Transition Time and Risk Stratification in Preclinical Alzheimer's Disease. Acad Radiol 2024:S1076-6332(24)00646-9. [PMID: 39332990 DOI: 10.1016/j.acra.2024.08.059] [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: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/29/2024]
Abstract
RATIONALE AND OBJECTIVES Accurate prediction of the progression of preclinical Alzheimer's disease (AD) is crucial for improving clinical management and disease prognosis. The objective of this study was to develop and validate clinical-radimoics integrated model to predict the time to progression (TTP) and disease risk stratification of preclinical AD. MATERIALS AND METHODS A total of 244 cases (mean age: 73.8 ± 5.5 years, 120 women) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database were randomly divided into the training cohort (n = 172) and validation cohort (n = 72) using a 7:3 ratio. Clinical factors were identified by univariate and multivariate COX regression. Radiomics features were extracted from GM, WM and CSF of T1WI images and selected by Spearman correlation analysis and least absolute shrinkage and selection operator (LASSO). Using selected clinical factors and radiomics features, the clinical, radimocis and clinical-radiomics nomogram models were developed for predicting the TTP. The performance of each model was assessed by C-index. The risk stratification ability and predicting efficacy of the clinical-radiomics model were utilizing the Kaplan-Meier curve and receiver operator characteristic (ROC) curve. RESULTS The C-index of clinical, radimocis and clinical-radiomics models were 0.852 (95% confidence interval[CI]:0.810-0.893), 0.863 (95%CI:0.816-0.910) and 0.903 (95%:0.870-0.936) in the training cohort and 0.725 (95%CI:0.630-0.820), 0.788 (95%CI:0.678-0.898), 0.813(95%CI:0.734-0.892) in the validation cohort. The AUCs of the multi-predictor nomogram at 1-, 3-, 5- and 7-year were 0.894, 0.908, 0.930, 0.979 in the training cohort and 0.671, 0.726, 0.839, 0.931 in the validation cohort. CONCLUSION In this study, we constructed a clinical-radimoics integrated model to predict the progression of preclinical AD and stratified the risk of disease progression in preclinical AD.
Collapse
Affiliation(s)
- Shuai Lin
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ming Xue
- Department of Radiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiali Sun
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Xu
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianqi Wang
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | - Min Lv
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ping Yang
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenjun Sheng
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zijian Cheng
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Wang
- Department of MRI, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
13
|
Lin N, Gao XY, Li X, Chu WM. Involvement of ubiquitination in Alzheimer's disease. Front Neurol 2024; 15:1459678. [PMID: 39301473 PMCID: PMC11412110 DOI: 10.3389/fneur.2024.1459678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024] Open
Abstract
The hallmark pathological features of Alzheimer's disease (AD) consist of senile plaques, which are formed by extracellular β-amyloid (Aβ) deposition, and neurofibrillary tangles, which are formed by the hyperphosphorylation of intra-neuronal tau proteins. With the increase in clinical studies, the in vivo imbalance of iron homeostasis and the dysfunction of synaptic plasticity have been confirmed to be involved in AD pathogenesis. All of these mechanisms are constituted by the abnormal accumulation of misfolded or conformationally altered protein aggregates, which in turn drive AD progression. Proteostatic imbalance has emerged as a key mechanism in the pathogenesis of AD. Ubiquitination modification is a major pathway for maintaining protein homeostasis, and protein degradation is primarily carried out by the ubiquitin-proteasome system (UPS). In this review, we provide an overview of the ubiquitination modification processes and related protein ubiquitination degradation pathways in AD, focusing on the microtubule-associated protein Tau, amyloid precursor protein (APP), divalent metal transporter protein 1 (DMT1), and α-amino-3-hyroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors. We also discuss recent advances in ubiquitination-based targeted therapy for AD, with the aim of contributing new ideas to the development of novel therapeutic interventions for AD.
Collapse
Affiliation(s)
- Nan Lin
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xi-Yan Gao
- The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiao Li
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wen-Ming Chu
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Sethi P, Bhaskar R, Singh KK, Gupta S, Han SS, Avinash D, Abomughaid MM, Koul A, Rani B, Ghosh S, Jha NK, Sinha JK. Exploring advancements in early detection of Alzheimer's disease with molecular assays and animal models. Ageing Res Rev 2024; 100:102411. [PMID: 38986845 DOI: 10.1016/j.arr.2024.102411] [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: 06/14/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Alzheimer's Disease (AD) is a challenging neurodegenerative condition, with overwhelming implications for affected individuals and healthcare systems worldwide. Animal models have played a crucial role in studying AD pathogenesis and testing therapeutic interventions. Remarkably, studies on the genetic factors affecting AD risk, such as APOE and TREM2, have provided valuable insights into disease mechanisms. Early diagnosis has emerged as a crucial factor in effective AD management, as demonstrated by clinical studies emphasizing the benefits of initiating treatment at early stages. Novel diagnostic technologies, including RNA sequencing of microglia, offer promising avenues for early detection and monitoring of AD progression. Therapeutic strategies remain to evolve, with a focus on targeting amyloid beta (Aβ) and tau pathology. Advances in animal models, such as APP-KI mice, and the advancement of anti-Aβ drugs signify progress towards more effective treatments. Therapeutically, the focus has shifted towards intricate approaches targeting multiple pathological pathways simultaneously. Strategies aimed at reducing Aβ plaque accumulation, inhibiting tau hyperphosphorylation, and modulating neuroinflammation are actively being explored, both in preclinical models and clinical trials. While challenges continue in developing validated animal models and translating preclinical findings to clinical success, the continuing efforts in understanding AD at molecular, cellular, and clinical levels offer hope for improved management and eventual prevention of this devastating disease.
Collapse
Affiliation(s)
- Paalki Sethi
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Apurva Koul
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University, Jaipur, Rajsthan, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India.
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140401, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
15
|
Huang Q, Wu W, Wen Y, Lu S, Zhao C. Potential therapeutic natural compounds for the treatment of Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155822. [PMID: 38909512 DOI: 10.1016/j.phymed.2024.155822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complicated neurodegenerative disease with cognitive impairment occurring in the older people, in which extracellular accumulation of β-amyloid and intracellular aggregation of hyperphosphorylated tau are regarded as the prevailing theories. However, the exact AD mechanism has not been determined. Moreover, there is no effective treatment available in phase III trials to eradicate AD, which is imperative to explore novel treatments. PURPOSE A number of up-to-date pre-clinical studies on cognitive impairment is beneficial to clarify the pathology of AD. This review recapitulates several advances in AD pathobiology and discusses the neuroprotective effects of natural compounds, such as phenolic compounds, natural polysaccharides and oligosaccharides, peptide, and lipids, underscoring the therapeutic potential for AD. METHODS Electronic databases involving PubMed, Web of Science, and Google Scholar were searched up to October 2023. Articles were conducted using the keywords like Alzheimer's disease, pathogenic mechanisms, natural compounds, and neuroprotection. RESULT This review summarized several AD pathologies and the neuroprotective effects of natural compounds such as natural polysaccharides and oligosaccharides, peptide, and lipids. CONCLUSION We have discussed the pathogenic mechanisms of AD and the effect natural products on neurodegenerative diseases particularly in treating AD. Specifically, we investigated the molecular pathways and links between natural compounds and Alzheimer's disease such as through NF-κB, Nrf2, and mTOR pathway. Further investigation is necessary in exploring the bioactivity and effectiveness of natural compounds in clinical trials, which may provide a promising treatment for AD patients.
Collapse
Affiliation(s)
- Qihui Huang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, 36310 Vigo, Spain
| | - Weihao Wu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yuxi Wen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, 36310 Vigo, Spain
| | - Suyue Lu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
16
|
Huang L, Li Z, Lv Y, Zhang X, Li Y, Li Y, Yu C. Unveiling disulfidptosis-related biomarkers and predicting drugs in Alzheimer's disease. Sci Rep 2024; 14:20185. [PMID: 39215110 PMCID: PMC11364544 DOI: 10.1038/s41598-024-70893-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease is the predominant form of dementia, and disulfidptosis is the latest reported mode of cell death that impacts various disease processes. This study used bioinformatics to analyze genes associated with disulfidptosis in Alzheimer's disease comprehensively. Based on the public datasets, the differentially expressed genes associated with disulfidptosis were identified, and immune cell infiltration was investigated through correlation analysis. Subsequently, hub genes were determined by a randomforest model. A prediction model was constructed using logistic regression. In addition, the drug-target affinity was predicted by a graph neural network model, and the results were validated by molecular docking. Five hub genes (PPEF1, NEUROD6, VIP, NUPR1, and GEM) were identified. The gene set showed significant enrichment for AD-related pathways. The logistic regression model demonstrated an AUC of 0.952, with AUC values of 0.916 and 0.864 in validated datasets. The immune infiltration analysis revealed significant heterogeneity between the Alzheimer's disease and control groups. High-affinity drugs for hub genes were identified. Through our study, a disease prediction model was constructed using potential biomarkers, and drugs targeting the genes were predicted. These results contribute to further understanding of the molecular mechanisms underlying Alzheimer's disease.
Collapse
Affiliation(s)
- Lei Huang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhengtai Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yitong Lv
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | | | - Yifan Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yingji Li
- ICE Bioscience Inc., Beijing, 100176, China.
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
17
|
Liu X, Yang Y, Lu Q, Yang J, Yuan J, Hu J, Tu Y. Association between systemic immune-inflammation index and serum neurofilament light chain: a population-based study from the NHANES (2013-2014). Front Neurol 2024; 15:1432401. [PMID: 39239395 PMCID: PMC11374650 DOI: 10.3389/fneur.2024.1432401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Background The systemic immune-inflammation index (SII) is a novel inflammatory marker used to assess the immune-inflammatory status of the human body. The systemic immune inflammation has an interplay and mutual relationship with neurological disorders. Serum neurofilament light chain (sNfL) is widely regarded as a potential biomarker for various neurological diseases. The study aimed to examine the association between SII and sNfL. Methods This cross-sectional investigation was conducted in a population with complete data on SII and sNfL from the 2013-2014 National Health and Nutrition Examination Survey (NHANES). The SII was calculated by dividing the product of platelet count and neutrophil count by the lymphocyte count. Multivariate linear regression models and smooth curves were used to explore the linear connection between SII and sNfL. Sensitivity analyses, interaction tests, and diabetes subgroup smoothing curve fitting were also performed. Results A total of 2,025 participants were included in our present research. SII showed a significant positive association with the natural logarithm-transformed sNfL (ln-sNfL) in crude model [0.17 (0.07, 0.28)], partially adjusted model [0.13 (0.03, 0.22)], and fully adjusted model [0.12 (0.02, 0.22)]. In all participants, the positive association between SII and ln-sNfL served as a linear relationship, as indicated by a smooth curve. Interaction tests showed that age, gender, BMI, hypertension, and diabetes did not have a significant impact on this positive association (p for interaction >0.05). The subgroup analysis of diabetes was conducted using smooth curve fitting. It was found that compared to the group without diabetes and the group in a pre-diabetic state, the effect was more pronounced in the group with diabetes. Conclusion Our findings suggest that there is a positive association between SII and sNfL. Furthermore, in comparison to individuals without diabetes and those in a pre-diabetic state, the positive association between SII and sNfL was more pronounced in individuals with diabetes. Further large-scale prospective studies are needed to confirm the association between SII and sNfL.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Traditional Chinese Medicine Rehabilitation, Acupuncture, Moxibustion and Massage College, Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Yang
- Department of Big Data Management and Application, Health Economics and Management College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiutong Lu
- Department of Chinese Medicine, The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianshu Yang
- Department of Acupuncture, Moxibustion and Massage, Acupuncture, Moxibustion and Massage College, Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Yuan
- Department of Traditional Chinese Medicine Rehabilitation, Acupuncture, Moxibustion and Massage College, Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Hu
- Department of Traditional Chinese Medicine Health Preservation, Acupuncture, Moxibustion and Massage College, Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Tu
- Department of Traditional Chinese Medicine Health Preservation, Acupuncture, Moxibustion and Massage College, Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
18
|
Godos J, Micek A, Currenti W, Franchi C, Poli A, Battino M, Dolci A, Ricci C, Ungvari Z, Grosso G. Fish consumption, cognitive impairment and dementia: an updated dose-response meta-analysis of observational studies. Aging Clin Exp Res 2024; 36:171. [PMID: 39162889 PMCID: PMC11335789 DOI: 10.1007/s40520-024-02823-6] [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: 06/21/2024] [Accepted: 07/28/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Cognitive impairment is projected to affect a preponderant proportion of the aging population. Lifelong dietary habits have been hypothesized to play a role in preventing cognitive decline. Among the most studied dietary components, fish consumptionhas been extensively studied for its potential effects on the human brain. AIMS To perform a meta-analysis of observational studies exploring the association between fish intake and cognitive impairment/decline and all types of dementia. METHODS A systematic search of electronic databases was performed to identify observational studies providing quantitative data on fish consumption and outcomes of interest. Random effects models for meta-analyses using only extreme exposure categories, subgroup analyses, and dose-response analyses were performed to estimate cumulative risk ratios (RRs) and 95% confidence intervals (CIs). RESULTS The meta-analysis comprised 35 studies. Individuals reporting the highest vs. the lowest fish consumption were associated with a lower likelihood of cognitive impairment/decline (RR = 0.82, 95% CI: 0.75, 0.90, I2 = 61.1%), dementia (RR = 0.82, 95% CI: 0.73, 0.93, I2 = 38.7%), and Alzheimer's disease (RR = 0.80, 95% CI: 0.67, 0.96, I2 = 20.3%). The dose-response relation revealed a significantly decreased risk of cognitive impairment/decline and all cognitive outcomes across higher levels of fish intake up to 30% for 150 g/d (RR = 0.70, 95% CI: 0.52, 0.95). The results of this relation based on APOE ε4 allele status was mixed based on the outcome investigated. CONCLUSIONS Current findings suggest fish consumption is associated with a lower risk of cognitive impairment/decline in a dose-response manner, while for dementia and Alzheimer's disease there is a need for further studies to improve the strength of evidence.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Agnieszka Micek
- Statistical Laboratory, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, 31-501, Poland
| | - Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Carlotta Franchi
- Laboratory of Pharmacoepidemiology and Human Nutrition, Department of Health Policy, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, 20156, Italy
- Italian Institute for Planetary Health (IIPH), Milan, 20124, Italy
| | - Andrea Poli
- Nutrition Foundation of Italy (NFI), Milan, 20124, Italy
| | - Maurizio Battino
- Department of Clinical Sciences, Università Politecnica Delle Marche, Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, Santander, 39011, Spain
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Alberto Dolci
- Sustainable Development Department, Bolton Food SpA, Milan, 20124, Italy
| | - Cristian Ricci
- Africa Unit for Transdisciplinary Health Research (AUTHeR), North-West University, Potchefstroom, 2531, South Africa
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral College, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy.
| |
Collapse
|
19
|
Xiaojuan L, Hongmei L, Zhuxin W, Xiaoqin L, Lanbing D, Dan L, Yi Z. Exploration of the pharmacological components and therapeutic mechanisms in treatment of Alzheimer's disease with Polygonati Rhizoma and its processed product using combined analysis of metabolomics, network pharmacology, and gut microbiota. Heliyon 2024; 10:e35394. [PMID: 39170207 PMCID: PMC11336570 DOI: 10.1016/j.heliyon.2024.e35394] [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: 04/08/2024] [Revised: 06/20/2024] [Accepted: 07/28/2024] [Indexed: 08/23/2024] Open
Abstract
Polygonati Rhizoma (PR, Huangjing in Chinese) and its processed product (PRP), which are used in Traditional Chinese medicine (TCM) for cognitive enhancement and treatment of Alzheimer's disease (AD), have not been fully explored in terms of the different mechanisms underlying their anti-AD effects. Therefore, we used APP/PS1 mice as an AD model to assess the effects of PR and PRP on anxiety-like behaviors, cognitive function, memory performance, and pathological changes in the murine brain. UPLC-HRMS was applied to identify the components of PR and PRP that entered into the blood and brain. Network pharmacology was used to elucidate potential mechanisms underlying the improvement of AD. Differences in the intestinal flora composition between mice treated with PR and PRP were investigated using 16S rRNA sequencing, establishing a correlation between pharmacological components and distinct flora profiles. The results revealed that both PR and PRP interventions ameliorated cognitive deficits and attenuated Amyloid β (Aβ) plaque deposition in the brains of AD mice. Seven specific blood-entering components, namely glutamic acid, Phe-Phe, and uridine, etc., were associated with PR intervention, whereas ten specific blood-entering components including (2R,3S)-3-isopropylmalate, 3-methylhexahydropyrrolo[1,2-a]pyrazine-1,4-dione, and 3-methoxytyrosine were related to PRP intervention. Uridine was identified as a common brain-penetrating component in both PR and PRP interventions. Network pharmacology analysis suggested that the NOD-like receptor signaling pathway, Calcium signaling pathway and Alzheimer's disease were specific pathways targeted in AD treatment using PR intervention. Moreover, the apoptosis pathway was specifically linked to AD treatment during PRP intervention. Furthermore, the administration of both PR and PRP enhanced the abundance and diversity of the intestinal flora in APP/PS1 mice. Western blotting confirmed that PR excels in regulates inflammation, whereas PRP balances autophagy and apoptosis to alleviate the progression of AD. This study offers valuable insights and establishes a robust foundation for further comprehensive exploration of the intrinsic correlation between TCM and AD.
Collapse
Affiliation(s)
- Liao Xiaojuan
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, China
| | - Liu Hongmei
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, China
| | - Wang Zhuxin
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, China
| | - Liu Xiaoqin
- College of Pharmacy, Shandong Modern University, Jinan, 250104, China
| | - Deng Lanbing
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, China
| | - Luo Dan
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, China
| | - Zhou Yi
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, China
| |
Collapse
|
20
|
Ma Y, Liu S, Zhou Q, Li Z, Zhang Z, Yu B. Approved drugs and natural products at clinical stages for treating Alzheimer's disease. Chin J Nat Med 2024; 22:699-710. [PMID: 39197961 DOI: 10.1016/s1875-5364(24)60606-0] [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: 03/23/2024] [Indexed: 09/01/2024]
Abstract
Alzheimer's disease (AD) remains the foremost cause of dementia and represents a significant unmet healthcare need globally. The complex pathogenesis of AD, characterized by various pathological and physiological events, has historically challenged the development of anti-AD drugs. However, recent breakthroughs in AD drug development, including the approvals of aducanumab, lecanemab, and sodium oligomannate (GV-971), have ended a nearly two-decade hiatus in the introduction of new AD drugs. These developments have addressed long-standing challenges in AD drug development, marking a substantial shift in the therapeutic landscape of AD. Moreover, natural products (NPs) have shown promise in AD drug research, with several currently under clinical investigation. Their distinct properties and mechanisms of action offer new avenues to complement and enhance existing AD treatment approaches. This review article aims to provide an overview of the recent advancements and prospects in AD therapeutics, focusing on both NPs and approved drugs.
Collapse
Affiliation(s)
- Yajing Ma
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China
| | - Sufang Liu
- Department of Biomedical Sciences, College of Dentistry, Texas A & M University, Dallas 75246, USA
| | - Qingfeng Zhou
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China
| | - Zhonghua Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhijian Zhang
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China.
| | - Bin Yu
- College of Chemistry, Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
21
|
He S, Jiang W, Jiang B, Yu C, Zhao G, Li Y, Qi L, Zhang J, Wang D. Potential Roles of Nr4a3-Mediated Inflammation in Immunological and Neurological Diseases. Mol Neurobiol 2024; 61:5958-5973. [PMID: 38261254 DOI: 10.1007/s12035-024-03945-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
As a protein of the orphan nuclear receptor Nr4a family, Nr4a3 has no identified natural ligands. However, its biological activity can be mediated by inducing conformational changes through interactions with specific certain small molecules and receptors. Nr4a3 is activated as an early stress factor under various pathological conditions and plays a regulatory role in various tissues and cells, participating in processes such as cell differentiation, apoptosis, metabolism, and homeostasis. At present, research on the role of Nr4a3 in the pathophysiology of inflammation is considerably limited, especially with respect to its role in the central nervous system (CNS). In this review, we discuss the role of Nr4a3 in multiple sclerosis, Alzheimer's disease, retinopathy, Parkinson's disease, and other CNS diseases. This review shows that Nr4a3 has considerable potential as a therapeutic target in the treatment of CNS diseases. We provide a theoretical basis for the targeted therapy of CNS diseases and neuroinflammation, among other conditions.
Collapse
Affiliation(s)
- Siqi He
- Department of Pathology, Beihua University, Jilin, Jilin, 132000, China
- The Second Affiliated Hospital, Hengyang Medical School, University of South, Hengyang, 421200, Hunan, China
- Institute of Digestive Diseases, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Weijie Jiang
- The Second Affiliated Hospital, Hengyang Medical School, University of South, Hengyang, 421200, Hunan, China
| | - Baoyi Jiang
- Institute of Digestive Diseases, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Chunyan Yu
- Department of Pathology, Beihua University, Jilin, Jilin, 132000, China
| | - Guifang Zhao
- Institute of Digestive Diseases, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yifei Li
- Department of Pathology, Beihua University, Jilin, Jilin, 132000, China
| | - Ling Qi
- Institute of Digestive Diseases, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| | - Jia Zhang
- The Second Affiliated Hospital, Hengyang Medical School, University of South, Hengyang, 421200, Hunan, China.
| | - Dan Wang
- Department of Pathology, Beihua University, Jilin, Jilin, 132000, China.
| |
Collapse
|
22
|
Liang Y, Liu C, Cheng M, Geng L, Li J, Du W, Song M, Chen N, Yeleen TAN, Song L, Wang X, Han Y, Sheng C. The link between gut microbiome and Alzheimer's disease: From the perspective of new revised criteria for diagnosis and staging of Alzheimer's disease. Alzheimers Dement 2024; 20:5771-5788. [PMID: 38940631 PMCID: PMC11350031 DOI: 10.1002/alz.14057] [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: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
Over the past decades, accumulating evidence suggests that the gut microbiome exerts a key role in Alzheimer's disease (AD). The Alzheimer's Association Workgroup is updating the diagnostic criteria for AD, which changed the profiles and categorization of biomarkers from "AT(N)" to "ATNIVS." Previously, most of studies focus on the correlation between the gut microbiome and amyloid beta deposition ("A"), the initial AD pathological feature triggering the "downstream" tauopathy and neurodegeneration. However, limited research investigated the interactions between the gut microbiome and other AD pathogenesis ("TNIVS"). In this review, we summarize current findings of the gut microbial characteristics in the whole spectrum of AD. Then, we describe the association of the gut microbiome with updated biomarker categories of AD pathogenesis. In addition, we outline the gut microbiome-related therapeutic strategies for AD. Finally, we discuss current key issues of the gut microbiome research in the AD field and future research directions. HIGHLIGHTS: The new revised criteria for Alzheimer's disease (AD) proposed by the Alzheimer's Association Workgroup have updated the profiles and categorization of biomarkers from "AT(N)" to "ATNIVS." The associations of the gut microbiome with updated biomarker categories of AD pathogenesis are described. Current findings of the gut microbial characteristics in the whole spectrum of AD are summarized. Therapeutic strategies for AD based on the gut microbiome are proposed.
Collapse
Affiliation(s)
- Yuan Liang
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Congcong Liu
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Manman Cheng
- Department of Respiratory MedicineThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Lijie Geng
- Department of RadiologyThe People's Hospital of YanzhouJiningChina
| | - Jing Li
- Department of EmergencyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Wenying Du
- Department of NeurologyChina‐Japan Friendship HospitalBeijingChina
| | - Minfang Song
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Nian Chen
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | | | - Li Song
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Xiaoni Wang
- Department of NeurologySir Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- Key Laboratory of Biomedical Engineering of Hainan ProvinceSchool of Biomedical EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
| | - Can Sheng
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| |
Collapse
|
23
|
Deng Q, Wu C, Parker E, Liu TCY, Duan R, Yang L. Microglia and Astrocytes in Alzheimer's Disease: Significance and Summary of Recent Advances. Aging Dis 2024; 15:1537-1564. [PMID: 37815901 PMCID: PMC11272214 DOI: 10.14336/ad.2023.0907] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Alzheimer's disease, one of the most common forms of dementia, is characterized by a slow progression of cognitive impairment and neuronal loss. Currently, approved treatments for AD are hindered by various side effects and limited efficacy. Despite considerable research, practical treatments for AD have not been developed. Increasing evidence shows that glial cells, especially microglia and astrocytes, are essential in the initiation and progression of AD. During AD progression, activated resident microglia increases the ability of resting astrocytes to transform into reactive astrocytes, promoting neurodegeneration. Extensive clinical and molecular studies show the involvement of microglia and astrocyte-mediated neuroinflammation in AD pathology, indicating that microglia and astrocytes may be potential therapeutic targets for AD. This review will summarize the significant and recent advances of microglia and astrocytes in the pathogenesis of AD in three parts. First, we will review the typical pathological changes of AD and discuss microglia and astrocytes in terms of function and phenotypic changes. Second, we will describe microglia and astrocytes' physiological and pathological role in AD. These roles include the inflammatory response, "eat me" and "don't eat me" signals, Aβ seeding, propagation, clearance, synapse loss, synaptic pruning, remyelination, and demyelination. Last, we will review the pharmacological and non-pharmacological therapies targeting microglia and astrocytes in AD. We conclude that microglia and astrocytes are essential in the initiation and development of AD. Therefore, understanding the new role of microglia and astrocytes in AD progression is critical for future AD studies and clinical trials. Moreover, pharmacological, and non-pharmacological therapies targeting microglia and astrocytes, with specific studies investigating microglia and astrocyte-mediated neuronal damage and repair, may be a promising research direction for future studies regarding AD treatment and prevention.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Emily Parker
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
24
|
Liu M, Li T, Liang H, Zhong P. Herbal medicines in Alzheimer's disease and the involvement of gut microbiota. Front Pharmacol 2024; 15:1416502. [PMID: 39081953 PMCID: PMC11286407 DOI: 10.3389/fphar.2024.1416502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by memory loss and cognitive impairment. It severely affects the quality of life of victims. The prevalence of AD has been increasing in recent years. Therefore, it is of great importance to elucidate the pathogenic mechanism of AD and search for effective therapeutic approaches. Gut microbiota dysbiosis, an altered state of gut microbiota, has been well known for its involvement in the pathogenesis of AD. Much effort has been made in searching for approaches capable of modulating the composition of gut microbiota in recent years. Herbal medicines have attracted extensive attention in recent decades for the prevention and treatment of AD. Here, we gave an overview of the recent research progress on the modulatory effects of herbal medicines and herbal formulae on gut microbiota as well as the possible beneficial effects on AD, which may provide new insights into the discovery of anti-AD agents and their therapeutic potential for AD through modulating the composition of gut microbiota.
Collapse
Affiliation(s)
- Mingli Liu
- Department of Neurology, Yangpu District Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Tuming Li
- Department of Neurology, Yangpu District Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Huazheng Liang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Monash Suzhou Research Institute, Suzhou, China
| | - Ping Zhong
- Department of Neurology, Yangpu District Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
25
|
Yu M, Shen Z, Zhang S, Zhang Y, Zhao H, Zhang L. The active components of Erzhi wan and their anti-Alzheimer's disease mechanisms determined by an integrative approach of network pharmacology, bioinformatics, molecular docking, and molecular dynamics simulation. Heliyon 2024; 10:e33761. [PMID: 39027618 PMCID: PMC11255520 DOI: 10.1016/j.heliyon.2024.e33761] [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: 09/08/2023] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Erzhi Wan (EZW), a classic Traditional Chinese Medicine formula, has shown promise as a potential therapeutic option for Alzheimer's disease (AD), yet its mechanism remains elusive. Herein, we employed an integrative in-silico approach to investigate the active components and their mechanisms against AD. We screened four active components with blood-brain barrier permeabilities from TCMSP, along with 307 corresponding targets predicted by SwissTargetPrediction, PharmMapper, and TCMbank websites. Then, we retrieved 2260 AD-related targets from Genecards, OMIM, and NCBI databases. Furthermore, we constructed the protein-protein interaction (PPI) network of the intersected targets via the STRING database and performed the GO and KEGG enrichment analyses using the "clusterProfiler" R package. The results showed that the intersected targets were intimately related to the p53/PI3K/Akt signaling pathway, serotonergic synapse, and response to oxygen level. Subsequently, 25 core targets were found differentially expressed in brain regions by bioinformatics analyses of GEO datasets of clinical samples from the Alzdata database. The binding sites and stabilities between the active components and the core targets were investigated by the molecular docking approach using Autodock 4.2.6 software, followed by pocket detection and druggability assessment via the DoGSiteScorer server. The results showed that acacetin, β-sitosterol, and 3-O-acetyldammarenediol-II strongly interacted with the druggable pockets of AR, CASP8, POLB, and PREP. Eventually, the docking results were further cross-referenced with the literature research and validated by 100 ns of molecular dynamics simulations using GROMACS software. Binding free energies were calculated via MM/PBSA strategy combined with interaction entropy. The simulation results indicated stable bindings between four docking pairs including acacetin-AR, acacetin-CASP8, β-sitosterol-POLB, and 3-O-acetyldammarenediol-II-PREP. Overall, our study demonstrated a theoretical basis for how three active components of EZW confer efficacy against AD. It provides a promising reference for subsequent research regarding drug discoveries and clinical applications.
Collapse
Affiliation(s)
- Meng Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Zhongqi Shen
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Shaozhi Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Hongwei Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Longfei Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| |
Collapse
|
26
|
Li WB, Xu LL, Wang SL, Wang YY, Pan YC, Shi LQ, Guo DS. Co-Assembled Nanoparticles toward Multi-Target Combinational Therapy of Alzheimer's Disease by Making Full Use of Molecular Recognition and Self-Assembly. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401918. [PMID: 38662940 DOI: 10.1002/adma.202401918] [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: 02/05/2024] [Revised: 04/10/2024] [Indexed: 05/07/2024]
Abstract
The complex pathologies in Alzheimer's disease (AD) severely limit the effectiveness of single-target pharmic interventions, thus necessitating multi-pronged therapeutic strategies. While flexibility is essentially demanded in constructing such multi-target systems, for achieving optimal synergies and also accommodating the inherent heterogeneity within AD. Utilizing the dynamic reversibility of supramolecular strategy for conferring sufficient tunability in component substitution and proportion adjustment, amphiphilic calixarenes are poised to be a privileged molecular tool for facilely achieving function integration. Herein, taking β-amyloid (Aβ) fibrillation and oxidative stress as model combination pattern, a supramolecular multifunctional integration is proposed by co-assembling guanidinium-modified calixarene with ascorbyl palmitate and loading dipotassium phytate within calixarene cavity. Serial pivotal events can be simultaneously addressed by this versatile system, including 1) inhibition of Aβ production and aggregation, 2) disintegration of Aβ fibrils, 3) acceleration of Aβ metabolic clearance, and 4) regulation of oxidative stress, which is verified to significantly ameliorate the cognitive impairment of 5×FAD mice, with reduced Aβ plaque content, neuroinflammation, and neuronal apoptosis. Confronted with the extremely intricate clinical realities of AD, the strategy presented here exhibits ample adaptability for necessary alterations on combinations, thereby may immensely expedite the advancement of AD combinational therapy through providing an exceptionally convenient platform.
Collapse
Affiliation(s)
- Wen-Bo Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Lin-Lin Xu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Si-Lei Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Ying-Yue Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Yu-Chen Pan
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Lin-Qi Shi
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300090, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi, 844000, China
| |
Collapse
|
27
|
Sun Y, Xia Q, Du L, Gan Y, Ren X, Liu G, Wang Y, Yan S, Li S, Zhang X, Xiao X, Jin H. Neuroprotective effects of Anshen Bunao Syrup on cognitive dysfunction in Alzheimer's disease rat models. Biomed Pharmacother 2024; 176:116754. [PMID: 38810401 DOI: 10.1016/j.biopha.2024.116754] [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: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024] Open
Abstract
Alzheimer's disease (AD) presents a significant challenge due to its prevalence and lack of cure, driving the quest for effective treatments. Anshen Bunao Syrup, a traditional Chinese medicine known for its neuroprotective properties, shows promise in addressing this need. However, understanding its precise mechanisms in AD remains elusive. This study aimed to investigate Anshen Bunao Syrup's therapeutic potential in AD treatment using a scopolamine-induced AD rat model. Assessments included novel-object recognition and Morris water maze tasks to evaluate spatial learning and memory, alongside Nissl staining and ELISA analyses for neuronal damage and biomarker levels. Results demonstrated that Anshen Bunao Syrup effectively mitigated cognitive dysfunction by inhibiting amyloid-β and phosphorylation Tau aggregation, thereby reducing neuronal damage. Metabolomics profiling of rats cortex revealed alterations in key metabolites implicated in tryptophan and fatty acid metabolism pathways, suggesting a role in the therapeutic effects of Anshen Bunao Syrup. Additionally, ELISA and correlation analyses indicated attenuation of oxidative stress and immune response through metabolic remodeling. In conclusion, this study provides compelling evidence for the neuroprotective effects of Anshen Bunao Syrup in AD models, shedding light on its potential as a therapeutic agent for AD prevention and treatment.
Collapse
Affiliation(s)
- Yuanfang Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qi Xia
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lijing Du
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yu Gan
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaopeng Ren
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Gang Liu
- Jilin Aodong Yanbian Pharmaceutical Co., Ltd., Yanbian 133700, China
| | - Yongkuan Wang
- Jilin Aodong Yanbian Pharmaceutical Co., Ltd., Yanbian 133700, China
| | - Shikai Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shasha Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiuyun Zhang
- Jilin Aodong Yanbian Pharmaceutical Co., Ltd., Yanbian 133700, China.
| | - Xue Xiao
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Huizi Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
28
|
Reda SM, Setti SE, Berthiaume AA, Wu W, Taylor RW, Johnston JL, Stein LR, Moebius HJ, Church KJ. Fosgonimeton attenuates amyloid-beta toxicity in preclinical models of Alzheimer's disease. Neurotherapeutics 2024; 21:e00350. [PMID: 38599894 PMCID: PMC11067346 DOI: 10.1016/j.neurot.2024.e00350] [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: 09/14/2023] [Revised: 03/13/2024] [Accepted: 03/16/2024] [Indexed: 04/12/2024] Open
Abstract
Positive modulation of hepatocyte growth factor (HGF) signaling may represent a promising therapeutic strategy for Alzheimer's disease (AD) based on its multimodal neurotrophic, neuroprotective, and anti-inflammatory effects addressing the complex pathophysiology of neurodegeneration. Fosgonimeton is a small-molecule positive modulator of the HGF system that has demonstrated neurotrophic and pro-cognitive effects in preclinical models of dementia. Herein, we evaluate the neuroprotective potential of fosgonimeton, or its active metabolite, fosgo-AM, in amyloid-beta (Aβ)-driven preclinical models of AD, providing mechanistic insight into its mode of action. In primary rat cortical neurons challenged with Aβ (Aβ1-42), fosgo-AM treatment significantly improved neuronal survival, protected neurite networks, and reduced tau hyperphosphorylation. Interrogation of intracellular events indicated that cortical neurons treated with fosgo-AM exhibited a significant decrease in mitochondrial oxidative stress and cytochrome c release. Following Aβ injury, fosgo-AM significantly enhanced activation of pro-survival effectors ERK and AKT, and reduced activity of GSK3β, one of the main kinases involved in tau hyperphosphorylation. Fosgo-AM also mitigated Aβ-induced deficits in Unc-like kinase 1 (ULK1) and Beclin-1, suggesting a potential effect on autophagy. Treatment with fosgo-AM protected cortical neurons from glutamate excitotoxicity, and such effects were abolished in the presence of an AKT or MEK/ERK inhibitor. In vivo, fosgonimeton administration led to functional improvement in an intracerebroventricular Aβ25-35 rat model of AD, as it significantly rescued cognitive function in the passive avoidance test. Together, our data demonstrate the ability of fosgonimeton to counteract mechanisms of Aβ-induced toxicity. Fosgonimeton is currently in clinical trials for mild-to-moderate AD (NCT04488419; NCT04886063).
Collapse
Affiliation(s)
- Sherif M Reda
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Sharay E Setti
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | | | - Wei Wu
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Robert W Taylor
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Jewel L Johnston
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Liana R Stein
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Hans J Moebius
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Kevin J Church
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA.
| |
Collapse
|
29
|
Hu XH, Yu KY, Li XX, Zhang JN, Jiao JJ, Wang ZJ, Cai HY, Wang L, He YX, Wu MN. Selective Orexin 2 Receptor Blockade Alleviates Cognitive Impairments and the Pathological Progression of Alzheimer's Disease in 3xTg-AD Mice. J Gerontol A Biol Sci Med Sci 2024; 79:glae115. [PMID: 38682858 DOI: 10.1093/gerona/glae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Indexed: 05/01/2024] Open
Abstract
The orexin system is closely related to the pathogenesis of Alzheimer's disease (AD). Orexin-A aggravates cognitive dysfunction and increases amyloid β (Aβ) deposition in AD model mice, but studies of different dual orexin receptor (OXR) antagonists in AD have shown inconsistent results. Our previous study revealed that OX1R blockade aggravates cognitive deficits and pathological progression in 3xTg-AD mice, but the effects of OX2R and its potential mechanism in AD have not been reported. In the present study, OX2R was blocked by oral administration of the selective OX2R antagonist MK-1064, and the effects of OX2R blockade on cognitive dysfunction and neuropsychiatric symptoms in 3xTg-AD mice were evaluated via behavioral tests. Then, immunohistochemistry, western blotting, and ELISA were used to detect Aβ deposition, tau phosphorylation, and neuroinflammation, and electrophysiological and wheel-running activity recording were recorded to observe hippocampal synaptic plasticity and circadian rhythm. The results showed that OX2R blockade ameliorated cognitive dysfunction, improved LTP depression, increased the expression of PSD-95, alleviated anxiety- and depression-like behaviors and circadian rhythm disturbances in 3xTg-AD mice, and reduced Aβ pathology, tau phosphorylation, and neuroinflammation in the brains of 3xTg-AD mice. These results indicated that chronic OX2R blockade exerts neuroprotective effects in 3xTg-AD mice by reducing AD pathology at least partly through improving circadian rhythm disturbance and the sleep-wake cycle and that OX2R might be a potential target for the prevention and treatment of AD; however, the potential mechanism by which OX2R exerts neuroprotective effects on AD needs to be further investigated.
Collapse
Affiliation(s)
- Xiao-Hong Hu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Kai-Yue Yu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xin-Xin Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jin-Nan Zhang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Juan-Juan Jiao
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Lei Wang
- Department of Geriatrics, Shanxi Bethune Hospital, Taiyuan, People's Republic of China
| | - Ye-Xin He
- Department of Radiology, Shanxi Provincial People's Hospital, Taiyuan, People's Republic of China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
30
|
Zhong M, Xu QQ, Hu Z, Yang W, Lin ZX, Xian YF. Tianma-Gouteng pair ameliorates the cognitive deficits on two transgenic mouse models of Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118113. [PMID: 38548119 DOI: 10.1016/j.jep.2024.118113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is a progressive neurodegenerative disease. Tianma-Gouteng Pair (TGP), commonly prescribed as a pair-herbs, can be found in many Chinese medicine formulae to treat brain diseases. However, the neuroprotective effects and molecular mechanisms of TGP remained unexplored. AIM OF THE STUDY This study investigated the difference between the TgCRND8 and 5 × FAD transgenic mice, the anti-AD effects of TGP, and underlying molecular mechanisms of TGP against AD through the two mouse models. METHODS Briefly, three-month-old TgCRND8 and 5 × FAD mice were orally administered with TGP for 4 and 6 months, respectively. Behavioral tests were carried out to determine the neuropsychological functions. Moreover, immunofluorescence and western blotting assays were undertaken to reveal the molecular mechanisms of TGP. RESULTS Although TgCRND8 and 5 × FAD mice had different beta-amyloid (Aβ) burdens, neuroinflammation status, and cognition impairments, TGP exerted neuroprotective effects against AD in the two models. In detail, behavioral tests revealed that TGP treatment markedly ameliorated the anxiety-like behavior, attenuated the recognition memory deficits, and increased the spatial learning ability as well as the reference memory of TgCRND8 and 5 × FAD mice. Moreover, TGP treatment could regulate the beta-amyloid precursor protein (APP) processing by inhibiting the Aβ production enzymes such as β- and γ-secretases and activating Aβ degrading enzyme to reduce Aβ accumulation. In addition, TGP reduced the Aβ42 level, the ratio of Aβ42/Αβ40, Aβ accumulation, and tau hyperphosphorylation in both the 5 × FAD and TgCRND8 mouse models. Furthermore, TGP ameliorated neuroinflammation by decreasing the densities of activated microglia and astrocytes, and inhibiting the production of inflammatory cytokines. TGP upregulated the SIRT1 and AMPK, and downregulated sterol response element binding protein 2 (SREBP2) in the brain of TgCRND8 mice and deactivation of the EPhA4 and c-Abl in the brain tissues of 5 × FAD mice. CONCLUSION Our experiments for the first time revealed the neuroprotective effects and molecular mechanism of TGP on 5 × FAD and TgCRND8 transgenic mouse models of different AD stages. TGP decreased the level of Aβ aggregates, improved the tauopathy, and reduced the neuroinflammation by regulation of the SIRT1/AMPK/SREBP2 axis and deactivation of EPhA4/c-Abl signaling pathway in the brains of TgCRND8 and 5 × FAD mice, respectively. All these findings unequivocally confirmed that the TGP would be promising in developing into an anti-AD therapeutic pharmaceutical.
Collapse
Affiliation(s)
- Mei Zhong
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China
| | - Qing-Qing Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China
| | - Zhen Hu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China
| | - Wen Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China; Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, Shatin, N.T., Hong Kong SAR, PR China; Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China.
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China.
| |
Collapse
|
31
|
Martín-Belmonte A, Aguado C, Alfaro-Ruiz R, Kulik A, de la Ossa L, Moreno-Martínez AE, Alberquilla S, García-Carracedo L, Fernández M, Fajardo-Serrano A, Aso E, Shigemoto R, Martín ED, Fukazawa Y, Ciruela F, Luján R. Nanoarchitecture of Ca V2.1 channels and GABA B receptors in the mouse hippocampus: Impact of APP/PS1 pathology. Brain Pathol 2024:e13279. [PMID: 38887180 DOI: 10.1111/bpa.13279] [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: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Voltage-gated CaV2.1 (P/Q-type) Ca2+ channels play a crucial role in regulating neurotransmitter release, thus contributing to synaptic plasticity and to processes such as learning and memory. Despite their recognized importance in neural function, there is limited information on their potential involvement in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we aimed to explore the impact of AD pathology on the density and nanoscale compartmentalization of CaV2.1 channels in the hippocampus in association with GABAB receptors. Histoblotting experiments showed that the density of CaV2.1 channel was significantly reduced in the hippocampus of APP/PS1 mice in a laminar-dependent manner. CaV2.1 channel was enriched in the active zone of the axon terminals and was present at a very low density over the surface of dendritic tree of the CA1 pyramidal cells, as shown by quantitative SDS-digested freeze-fracture replica labelling (SDS-FRL). In APP/PS1 mice, the density of CaV2.1 channel in the active zone was significantly reduced in the strata radiatum and lacunosum-moleculare, while it remained unaltered in the stratum oriens. The decline in Cav2.1 channel density was found to be associated with a corresponding impairment in the GABAergic synaptic function, as evidenced by electrophysiological experiments carried out in the hippocampus of APP/PS1 mice. Remarkably, double SDS-FRL showed a co-clustering of CaV2.1 channel and GABAB1 receptor in nanodomains (~40-50 nm) in wild type mice, while in APP/PS1 mice this nanoarchitecture was absent. Together, these findings suggest that the AD pathology-induced reduction in CaV2.1 channel density and CaV2.1-GABAB1 de-clustering may play a role in the synaptic transmission alterations shown in the AD hippocampus. Therefore, uncovering these layer-dependent changes in P/Q calcium currents associated with AD pathology can benefit the development of future strategies for AD management.
Collapse
Affiliation(s)
- Alejandro Martín-Belmonte
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Carolina Aguado
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| | - Rocío Alfaro-Ruiz
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| | - Akos Kulik
- Institute for Physiology II, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Luis de la Ossa
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería Informática, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Ana Esther Moreno-Martínez
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| | - Samuel Alberquilla
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Lucía García-Carracedo
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Miriam Fernández
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| | - Ana Fajardo-Serrano
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Eduardo D Martín
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, University of Fukui, Fukui, Japan
- Life Science Innovation Center, University of Fukui, Fukui, Japan
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Rafael Luján
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| |
Collapse
|
32
|
Sharma M, Tanwar AK, Purohit PK, Pal P, Kumar D, Vaidya S, Prajapati SK, Kumar A, Dhama N, Kumar S, Gupta SK. Regulatory roles of microRNAs in modulating mitochondrial dynamics, amyloid beta fibrillation, microglial activation, and cholinergic signaling: Implications for alzheimer's disease pathogenesis. Neurosci Biobehav Rev 2024; 161:105685. [PMID: 38670299 DOI: 10.1016/j.neubiorev.2024.105685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
Alzheimer's Disease (AD) remains a formidable challenge due to its complex pathology, notably involving mitochondrial dysfunction and dysregulated microRNA (miRNA) signaling. This study delves into the underexplored realm of miRNAs' impact on mitochondrial dynamics and their interplay with amyloid-beta (Aβ) aggregation and tau pathology in AD. Addressing identified gaps, our research utilizes advanced molecular techniques and AD models, alongside patient miRNA profiles, to uncover miRNAs pivotal in mitochondrial regulation. We illuminate novel miRNAs influencing mitochondrial dynamics, Aβ, and tau, offering insights into their mechanistic roles in AD progression. Our findings not only enhance understanding of AD's molecular underpinnings but also spotlight miRNAs as promising therapeutic targets. By elucidating miRNAs' roles in mitochondrial dysfunction and their interactions with hallmark AD pathologies, our work proposes innovative strategies for AD therapy, aiming to mitigate disease progression through targeted miRNA modulation. This contribution marks a significant step toward novel AD treatments, emphasizing the potential of miRNAs in addressing this complex disease.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Pharmacology, Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India.
| | - Ankur Kumar Tanwar
- Department of Pharmacy, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | | | - Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.
| | - Devendra Kumar
- Department of Pharmaceutical Chemistry, NMIMS School of Pharmacy and Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Shirpur Campus, Dhule, Maharashtra, India
| | - Sandeep Vaidya
- CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | | | - Aadesh Kumar
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Nidhi Dhama
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Sokindra Kumar
- Department of Pharmacology, Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Sukesh Kumar Gupta
- Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
33
|
Han T, Xu Y, Sun L, Hashimoto M, Wei J. Microglial response to aging and neuroinflammation in the development of neurodegenerative diseases. Neural Regen Res 2024; 19:1241-1248. [PMID: 37905870 PMCID: PMC11467914 DOI: 10.4103/1673-5374.385845] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cellular senescence and chronic inflammation in response to aging are considered to be indicators of brain aging; they have a great impact on the aging process and are the main risk factors for neurodegeneration. Reviewing the microglial response to aging and neuroinflammation in neurodegenerative diseases will help understand the importance of microglia in neurodegenerative diseases. This review describes the origin and function of microglia and focuses on the role of different states of the microglial response to aging and chronic inflammation on the occurrence and development of neurodegenerative diseases, including Alzheimer's disease, Huntington's chorea, and Parkinson's disease. This review also describes the potential benefits of treating neurodegenerative diseases by modulating changes in microglial states. Therefore, inducing a shift from the neurotoxic to neuroprotective microglial state in neurodegenerative diseases induced by aging and chronic inflammation holds promise for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Tingting Han
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yuxiang Xu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lin Sun
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, Henan Province, China
| | - Makoto Hashimoto
- Department of Basic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| |
Collapse
|
34
|
Yu X, Tao J, Xiao T, Duan X. P-hydroxybenzaldehyde protects Caenorhabditis elegans from oxidative stress and β-amyloid toxicity. Front Aging Neurosci 2024; 16:1414956. [PMID: 38841104 PMCID: PMC11150654 DOI: 10.3389/fnagi.2024.1414956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Gastrodia elata is the dried tuber of the orchid Gastrodia elata Bl. It is considered a food consisting of a source of precious medicinal herbs, whose chemical composition is relatively rich. Gastrodia elata and its extracted fractions have been shown to have neuroprotective effects. P-hydroxybenzaldehyde (p-HBA), as one of the main active components of Gastrodia elata, has anti-inflammatory, antioxidative stress, and cerebral protective effects, which has potential for the treatment of Alzheimer's disease (AD). The aim of this study was to verify the role of p-HBA in AD treatment and to investigate its mechanism of action in depth based using the Caenorhabditis elegans (C. elegans) model. Methods In this study, we used paralysis, lifespan, behavioral and antistress experiments to investigate the effects of p-HBA on AD and aging. Furthermore, we performed reactive oxygen species (ROS) assay, thioflavin S staining, RNA-seq analysis, qPCR validation, PCR Array, and GFP reporter gene worm experiment to determine the anti-AD effects of p-HBA, as well as in-depth studies on its mechanisms. Results p-HBA was able to delay paralysis, improve mobility and resistance to stress, and delay aging in the AD nematode model. Further mechanistic studies showed that ROS and lipofuscin levels, Aβ aggregation, and toxicity were reduced after p-HBA treatment, suggesting that p-HBA ameliorated Aβ-induced toxicity by enhancing antioxidant and anti-aging activity and inhibiting Aβ aggregation. p-HBA had a therapeutic effect on AD by improving stress resistance, as indicated by the down-regulation of NLP-29 and UCR-11 expression and up-regulation of PQN-75 and LYS-3 expression. In addition, the gene microarray showed that p-HBA treatment played a positive role in genes related to AD, anti-aging, ribosomal protein pathway, and glucose metabolism, which were collectively involved in the anti-AD mechanism of p-HBA. Finally, we also found that p-HBA promoted nuclear localization of DAF-16 and increased the expression of SKN-1, SOD-3, and GST-4, which contributed significantly to inhibition of Aβ toxicity and enhancement of antioxidative stress. Conclusion Our work suggests that p-HBA has some antioxidant and anti-aging activities. It may be a viable candidate for the treatment and prevention of Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
35
|
Li J, Haj Ebrahimi A, Ali AB. Advances in Therapeutics to Alleviate Cognitive Decline and Neuropsychiatric Symptoms of Alzheimer's Disease. Int J Mol Sci 2024; 25:5169. [PMID: 38791206 PMCID: PMC11121252 DOI: 10.3390/ijms25105169] [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: 04/09/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Dementia exists as a 'progressive clinical syndrome of deteriorating mental function significant enough to interfere with activities of daily living', with the most prevalent type of dementia being Alzheimer's disease (AD), accounting for about 80% of diagnosed cases. AD is associated with an increased risk of comorbidity with other clinical conditions such as hypertension, diabetes, and neuropsychiatric symptoms (NPS) including, agitation, anxiety, and depression as well as increased mortality in late life. For example, up to 70% of patients diagnosed with AD are affected by anxiety. As aging is the major risk factor for AD, this represents a huge global burden in ageing populations. Over the last 10 years, significant efforts have been made to recognize the complexity of AD and understand the aetiology and pathophysiology of the disease as well as biomarkers for early detection. Yet, earlier treatment options, including acetylcholinesterase inhibitors and glutamate receptor regulators, have been limited as they work by targeting the symptoms, with only the more recent FDA-approved drugs being designed to target amyloid-β protein with the aim of slowing down the progression of the disease. However, these drugs may only help temporarily, cannot stop or reverse the disease, and do not act by reducing NPS associated with AD. The first-line treatment options for the management of NPS are selective serotonin reuptake inhibitors/selective noradrenaline reuptake inhibitors (SSRIs/SNRIs) targeting the monoaminergic system; however, they are not rational drug choices for the management of anxiety disorders since the GABAergic system has a prominent role in their development. Considering the overall treatment failures and side effects of currently available medication, there is an unmet clinical need for rationally designed therapies for anxiety disorders associated with AD. In this review, we summarize the current status of the therapy of AD and aim to highlight novel angles for future drug therapy in our ongoing efforts to alleviate the cognitive deficits and NPS associated with this devastating disease.
Collapse
Affiliation(s)
| | | | - Afia B. Ali
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (J.L.); (A.H.E.)
| |
Collapse
|
36
|
Li Q, Liao Q, Qi S, Huang H, He S, Lyu W, Liang J, Qin H, Cheng Z, Yu F, Dong X, Wang Z, Han L, Han Y. Opportunities and perspectives of small molecular phosphodiesterase inhibitors in neurodegenerative diseases. Eur J Med Chem 2024; 271:116386. [PMID: 38614063 DOI: 10.1016/j.ejmech.2024.116386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
Phosphodiesterase (PDE) is a superfamily of enzymes that are responsible for the hydrolysis of two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). PDE inhibition promotes the gene transcription by activating cAMP-response element binding protein (CREB), initiating gene transcription of brain-derived neurotrophic factor (BDNF). The procedure exerts neuroprotective profile, and motor and cognitive improving efficacy. From this point of view, PDE inhibition will provide a promising therapeutic strategy for treating neurodegenerative disorders. Herein, we summarized the PDE inhibitors that have entered the clinical trials or been discovered in recent five years. Well-designed clinical or preclinical investigations have confirmed the effectiveness of PDE inhibitors, such as decreasing Aβ oligomerization and tau phosphorylation, alleviating neuro-inflammation and oxidative stress, modulating neuronal plasticity and improving long-term cognitive impairment.
Collapse
Affiliation(s)
- Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| | - Qinghong Liao
- Shandong Kangqiao Biotechnology Co., Ltd, Qingdao, 266033, Shandong, PR China
| | - Shulei Qi
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - He Huang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Siyu He
- Guizhou Province Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, 550004, Guizhou, PR China
| | - Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Jinxin Liang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Huan Qin
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Zimeng Cheng
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Fan Yu
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Xue Dong
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Ziming Wang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China; School of Pharmacy, Binzhou Medical University, Yantai, 256699, Shandong, PR China
| | - Lingfei Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Yantao Han
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| |
Collapse
|
37
|
Wang J, Shi L, Zhu X, Tang Q, Wu M, Li B, Liu W, Jin Y. Entropy-driven catalysis-based lateral flow assay for sensitive detection of Alzheimer 's-associated MicroRNA. Talanta 2024; 271:125656. [PMID: 38224658 DOI: 10.1016/j.talanta.2024.125656] [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: 06/25/2023] [Revised: 11/17/2023] [Accepted: 01/08/2024] [Indexed: 01/17/2024]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the brain worldwide. Currently, there is no effective cure. But accurate and early diagnosis of AD is critical to the development of patient care and future treatments. MiRNA-16 has been considered as an effective diagnostic biomarker for AD because of its regulatory effect on key proteins of AD. Herein, a colorimetric lateral flow assay (LFA) was developed for sensitive detection of miRNA-16 based on entropy-driven catalysis (EDC) amplification strategy. MiRNA-16 triggered EDC and released more linker DNAs (LDNA) of sandwich structure. Thus, AuNPs were enriched at the T-line to enhance the colorimetric signal and improve the sensitivity of visual assay. It showed good specificity and sensitivity for detecting miRNA-16 with a detection limit of 1.01 pM. The practical detection of miRNA-16 in human serum obtained satisfactory result. Significantly, EDC achieved signal amplification in homogeneous solution without enzyme and DNA labeling, leading to a cheap and easy detection of miRNA-16. Therefore, it provided a portable and rapid assay for AD-related nucleic acid, which holds a potential for point-of-care testing (POCT) of AD.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China.
| | - Lu Shi
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Xinyu Zhu
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Qiaorong Tang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Mengmeng Wu
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Baoxin Li
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Wei Liu
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Yan Jin
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
38
|
He J, Tam KY. Dual-target inhibitors of cholinesterase and GSK-3β to modulate Alzheimer's disease. Drug Discov Today 2024; 29:103914. [PMID: 38340951 DOI: 10.1016/j.drudis.2024.103914] [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: 09/21/2023] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that affects over 55 million patients worldwide. Most of the approved small-molecule drugs for AD have been designed to tackle a single pathological hallmark, such as cholinergic dysfunction or amyloid toxicity, and thus may not fully address the multifactorial nature of the disease. Inhibition of both cholinesterase and glycogen synthase kinase-3β (GSK-3β) has emerged as a promising strategy to modulate AD. However, the dual inhibition of these two targets posts challenges in molecular design: issues related to target engagements and biopharmaceutical properties in particular must be overcome. In this review, we discuss the physiopathological roles and structures of cholinesterase and GSK-3β as well as recently reported dual-target inhibitors. We critically evaluate the current status of the discovery of dual-target inhibitors of cholinesterase and GSK-3β, and highlight further perspectives.
Collapse
Affiliation(s)
- Junqiu He
- Faculty of Health Sciences, University of Macau SAR, Avenida de Universidade, Taipa, Macau SAR, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau SAR, Avenida de Universidade, Taipa, Macau SAR, China.
| |
Collapse
|
39
|
Wang G, Zhang S, Lan H, Zheng X. Ochratoxin A (OTA) causes intestinal aging damage through the NLRP3 signaling pathway mediated by calcium overload and oxidative stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:27864-27882. [PMID: 38526719 DOI: 10.1007/s11356-024-32696-1] [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: 09/08/2023] [Accepted: 02/25/2024] [Indexed: 03/27/2024]
Abstract
Ochratoxin A (OTA) is a widespread environmental toxin that poses a serious threat to human and animal health. OTA has been shown to cause cellular and tissue damage and is a global public health problem. However, the effects of OTA on gastrointestinal aging have not been reported. The aim of this study was to investigate the effects of OTA on intestinal aging in vitro and in vivo. In vitro experiments showed that OTA induced cellular inflammation through calcium overload and oxidative stress, significantly up-regulated the expression of P16, P21, and P53 proteins, markedly increased senescence-associated β-galactosidase activity (SA-β-gal) positive cells, and obviously decreased the expression of proliferating cell nuclear antigen (PCNA) proteins, which led to intestinal cell senescence. Meanwhile, we found that treatment with β-carotene ameliorated OTA-induced intestinal cell senescence. Consistent with the results of the in vitro experiments, in vivo studies showed that the intestinal aging of mice fed OTA was significantly higher than that of the control group. In conclusion, OTA may induce intestinal aging through calcium overload, oxidative stress and inflammation. This study lays a foundation for further research on the toxicological effects of OTA.
Collapse
Affiliation(s)
- Guoxia Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Shuai Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Hainan Lan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
40
|
Liu S, Han Y, Kong L, Wang G, Ye Z. Atomic force microscopy in disease-related studies: Exploring tissue and cell mechanics. Microsc Res Tech 2024; 87:660-684. [PMID: 38063315 DOI: 10.1002/jemt.24471] [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: 07/30/2023] [Revised: 10/22/2023] [Accepted: 11/26/2023] [Indexed: 03/02/2024]
Abstract
Despite significant progress in human medicine, certain diseases remain challenging to promptly diagnose and treat. Hence, the imperative lies in the development of more exhaustive criteria and tools. Tissue and cellular mechanics exhibit distinctive traits in both normal and pathological states, suggesting that "force" represents a promising and distinctive target for disease diagnosis and treatment. Atomic force microscopy (AFM) holds great promise as a prospective clinical medical device due to its capability to concurrently assess surface morphology and mechanical characteristics of biological specimens within a physiological setting. This review presents a comprehensive examination of the operational principles of AFM and diverse mechanical models, focusing on its applications in investigating tissue and cellular mechanics associated with prevalent diseases. The findings from these studies lay a solid groundwork for potential clinical implementations of AFM. RESEARCH HIGHLIGHTS: By examining the surface morphology and assessing tissue and cellular mechanics of biological specimens in a physiological setting, AFM shows promise as a clinical device to diagnose and treat challenging diseases.
Collapse
Affiliation(s)
- Shuaiyuan Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Yibo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Central Hospital of Chongqing University, Chongqing Emergency Medical Center, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratory, Chongqing, China
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratory, Chongqing, China
| |
Collapse
|
41
|
Zhang B, Wan H, Maierwufu M, Liu Q, Li T, He Y, Wang X, Liu G, Hong X, Feng Q. STAT3 ameliorates truncated tau-induced cognitive deficits. Neural Regen Res 2024; 19:915-922. [PMID: 37843229 PMCID: PMC10664106 DOI: 10.4103/1673-5374.382253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/15/2023] [Accepted: 06/29/2023] [Indexed: 10/17/2023] Open
Abstract
Proteolytic cleavage of tau by asparagine endopeptidase (AEP) creates tau-N368 fragments, which may drive the pathophysiology associated with synaptic dysfunction and memory deterioration in the brain of Alzheimer's disease patients. Nonetheless, the molecular mechanisms of truncated tau-induced cognitive deficits remain unclear. Evidence suggests that signal transduction and activator of transcription-3 (STAT3) is associated with modulating synaptic plasticity, cell apoptosis, and cognitive function. Using luciferase reporter assays, electrophoretic mobility shift assays, western blotting, and immunofluorescence, we found that human tau-N368 accumulation inhibited STAT3 activity by suppressing STAT3 translocation into the nucleus. Overexpression of STAT3 improved tau-N368-induced synaptic deficits and reduced neuronal loss, thereby improving the cognitive deficits in tau-N368 mice. Moreover, in tau-N368 mice, activation of STAT3 increased N-methyl-D-aspartic acid receptor levels, decreased Bcl-2 levels, reversed synaptic damage and neuronal loss, and thereby alleviated cognitive deficits caused by tau-N368. Taken together, STAT3 plays a critical role in truncated tau-related neuropathological changes. This indicates a new mechanism behind the effect of tau-N368 on synapses and memory deficits. STAT3 can be used as a new molecular target to treat tau-N368-induced protein pathology.
Collapse
Affiliation(s)
- Bingge Zhang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Huali Wan
- Department of Laboratory Medicine, Guangdong Provincial, People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Maimaitijiang Maierwufu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qian Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Gongping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoyue Hong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, Hubei Province, China
| | - Qiong Feng
- Department of Pathology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
42
|
Alam J, Kalash A, Hassan MI, Rahman SZ. Agents at the Peak of US FDA Approval for the Treatment of Alzheimer's Disease. Neurol Res 2024; 46:318-325. [PMID: 38197595 DOI: 10.1080/01616412.2024.2302271] [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: 05/02/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
Where Alzheimer's disease (AD) is becoming a global health issue, the present anti-AD medications have also been exposed to produce only symptomatic outcomes. The pathological factors, like neuronal transmission impairment, amyloidal-tau constituents, oxidative damage, neuro-inflammation, synaptic dysfunction, infectious agents, and impairment of gut microbiota and vitamins' levels; all favor the disease's progression and sustainability. The researchers have investigated several drugable molecules against these factors; however, no treatment could have been discovered yet to prevent the disease's progression rather than anti-amyloidal antibodies. After a comprehensive review of the literature and the clinical registry (clinicaltrials.gov), the authors of this manuscript have explored drug molecules that are under phase-3 of clinical trials and at the peak of getting approval for the management of AD. The inclusion and exclusion criteria for clinical trials were decided by considering the basis of a drug's approval. We included only the clinical trials were found in stages of Enrolling-by-Invitation, Recruiting, Not Recruiting (But active), and Not Recruiting (Not active) while excluding Completed, Terminated, Suspended, Withdrawn, or the trials of Unknown Status. We have found many potent drug molecules reached the clinical trials in phase-3 that could be futuristic anti-AD agents. This review article aims to provide an update on the prospective potential anti-AD medicines and to reveal the therapeutic targets of great significance for designing further a possible drug development strategy against AD pathology.
Collapse
Affiliation(s)
- Jahngeer Alam
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Anushka Kalash
- Department of Pharmacology, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Syed Ziaur Rahman
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
43
|
Speers AB, Wright KM, Brandes MS, Kedjejian N, Matthews DG, Caruso M, Harris CJ, Koike S, Nguyen T, Quinn JF, Soumyanath A, Gray NE. Mode of administration influences plasma levels of active Centella asiatica compounds in 5xFAD mice while markers of neuroinflammation remain unaltered. Front Neurosci 2024; 18:1277626. [PMID: 38591068 PMCID: PMC10999680 DOI: 10.3389/fnins.2024.1277626] [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: 08/14/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction A water extract of Centella asiatica (L.) Urban [Apiaceae] (CAW) has demonstrated cognitive-enhancing effects in mouse models of Alzheimer's disease and aging, the magnitude of which is influenced by whether CAW is delivered in the drinking water or the diet. These cognitive benefits are accompanied by improvements in oxidative stress and mitochondrial function in the brain, two pathways related to the neuroinflammatory response. The effect of CAW on neuroinflammation, however, has not been directly studied. Here, we investigated the effect of CAW on neuroinflammation in 5xFAD mice and compared plasma levels of CAW's active compounds following two modes of CAW administration. Methods Eight-to-nine-month-old male and female 5xFAD mice and their wild-type littermates were administered CAW in their diet or drinking water (0 or 1,000 mg/kg/day) for five weeks. Immunohistochemistry was performed for β-amyloid (Aβ), glial fibrillary acidic protein (GFAP), and Griffonia simplicifolia lectin I (GSL I) in the cortex and hippocampus. Gene expression of inflammatory mediators (IL-6, TNFα, IL-1β, TREM2, AIF1, CX3CR1, CX3CL1, CD36, C3AR1, RAGE, CCR6, CD3E) was measured in the deep grey matter. Results CAW decreased cortical Aβ plaque burden in female 5xFAD mice administered CAW in the drinking water but had no effect on Aβ plaques in other treatment groups. CAW did not impact elevated levels of GFAP or GSL I in 5xFAD mice, regardless of sex, brain region, or mode of CAW administration. In the deep grey matter, CAW increased C3AR1 expression in 5xFAD females administered CAW in the drinking water and decreased IL-1β expression in 5xFAD males administered CAW in the diet. CAW had no effect, however, on gene expression levels of any other inflammatory mediator in the deep grey, for either sex or mode of CAW administration. Mice administered CAW in the drinking water versus the diet had significantly higher plasma levels of CAW compounds. Discussion CAW had little impact on the neuroinflammatory markers selected for evaluation in the present study, suggesting that the cognitive benefits of CAW may not be mediated by an anti-inflammatory effect or that additional molecular markers are needed to fully characterize the effect of CAW on neuroinflammation.
Collapse
Affiliation(s)
- Alex B. Speers
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Kirsten M. Wright
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Mikah S. Brandes
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nareg Kedjejian
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Christopher J. Harris
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Seiji Koike
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Thuan Nguyen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Joseph F. Quinn
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
44
|
Karagöl T, Karagöl A, Zhang S. Structural bioinformatics studies of serotonin, dopamine and norepinephrine transporters and their AlphaFold2 predicted water-soluble QTY variants and uncovering the natural mutations of L->Q, I->T, F->Y and Q->L, T->I and Y->F. PLoS One 2024; 19:e0300340. [PMID: 38517879 PMCID: PMC10959339 DOI: 10.1371/journal.pone.0300340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/26/2024] [Indexed: 03/24/2024] Open
Abstract
Monoamine transporters including transporters for serotonin, dopamine, and norepinephrine play key roles in monoaminergic synaptic signaling, involving in the molecular etiology of a wide range of neurological and physiological disorders. Despite being crucial drug targets, the study of transmembrane proteins remains challenging due to their localization within the cell membrane. To address this, we present the structural bioinformatics studies of 7 monoamine transporters and their water-soluble variants designed using the QTY code, by systematically replacing the hydrophobic amino acids leucine (L), valine (V), isoleucine (I) and phenylalanine (F) with hydrophilic amino acids (glutamine (Q), threonine (T) and tyrosine (Y). The resulting QTY variants, despite significant protein transmembrane sequence differences (44.27%-51.85%), showed similar isoelectric points (pI) and molecular weights. While their hydrophobic surfaces significantly reduced, this change resulted in a minimal structural alteration. Quantitatively, Alphafold2 predicted QTY variant structures displayed remarkable similarity with RMSD 0.492Å-1.619Å. Accompanied by the structural similarities of substituted amino acids in the context of 1.5Å electron density maps, our study revealed multiple QTY and reverse QTY variations in genomic databases. We further analyzed their phenotypical and topological characteristics. By extending evolutionary game theory to the molecular foundations of biology, we provided insights into the evolutionary dynamics of chemically distinct alpha-helices, their usage in different chemotherapeutic applications, and open possibilities of diagnostic medicine. Our study rationalizes that QTY variants of monoamine transporters may not only become distinct tools for medical, structural, and evolutionary research, but these transporters may also emerge as contemporary therapeutic targets, providing a new approach to treatment for several conditions.
Collapse
Affiliation(s)
- Taner Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Alper Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
45
|
Jangra J, Bajad NG, Singh R, Kumar A, Singh SK. Identification of novel potential cathepsin-B inhibitors through pharmacophore-based virtual screening, molecular docking, and dynamics simulation studies for the treatment of Alzheimer's disease. Mol Divers 2024:10.1007/s11030-024-10821-z. [PMID: 38517648 DOI: 10.1007/s11030-024-10821-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/03/2024] [Indexed: 03/24/2024]
Abstract
Cathepsin B is a cysteine protease lysosomal enzyme involved in several physiological functions. Overexpression of the enzyme enhances its proteolytic activity and causes the breakdown of amyloid precursor protein (APP) into neurotoxic amyloid β (Aβ), a characteristic hallmark of Alzheimer's disease (AD). Therefore, inhibition of the enzyme is a crucial therapeutic aspect for treating the disease. Combined structure and ligand-based drug design strategies were employed in the current study to identify the novel potential cathepsin B inhibitors. Five different pharmacophore models were developed and used for the screening of the ZINC-15 database. The obtained hits were analyzed for the presence of duplicates, interfering PAINS moieties, and structural similarities based on Tanimoto's coefficient. The molecular docking study was performed to screen hits with better target binding affinity. The top seven hits were selected and were further evaluated based on their predicted ADME properties. The resulting best hits, ZINC827855702, ZINC123282431, and ZINC95386847, were finally subjected to molecular dynamics simulation studies to determine the stability of the protein-ligand complex during the run. ZINC123282431 was obtained as the virtual lead compound for cathepsin B inhibition and may be a promising novel anti-Alzheimer agent.
Collapse
Affiliation(s)
- Jatin Jangra
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Nilesh Gajanan Bajad
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
46
|
Gontrum EQ, Paolillo EW, Lee S, Diaz V, Ehrenberg A, Saloner R, Mundada NS, La Joie R, Rabinovici G, Kramer JH, Casaletto KB. Neuropsychiatric Profiles and Cerebral Amyloid Burden in Adults without Dementia. Dement Geriatr Cogn Disord 2024; 53:119-127. [PMID: 38513620 PMCID: PMC11187670 DOI: 10.1159/000538376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION We comprehensively evaluated how self- and informant-reported neuropsychiatric symptoms (NPS) were differentially associated with cerebral amyloid-beta (Aβ) PET levels in older adults without dementia. METHODS Two hundred and twenty-one participants (48% female, age = 73.4 years ± 8.4, Clinical Dementia Rating = 0 [n = 184] or 0.5 [n = 37]) underwent an Aβ-PET scan (florbetapir or PIB), comprehensive neuropsychological testing, and self-reported (Geriatric Depression Scale - 30 item [GDS-30]) and informant-reported interview (Neuropsychiatric Inventory Questionnaire [NPI-Q]) of NPS. Cerebral Aβ burden was quantified using centiloids (CL). NPI-Q and GDS-30 queried the presence of NPS within 4 subdomains and 6 subscales, respectively. Regression models examined the relationship between NPS and Aβ-PET CL. RESULTS Both higher self- and informant-reported NPS were associated with higher Aβ burden. Among specific NPI-Q subdomains, informant-reported changes in depression, anxiety, and irritability were all associated with higher Aβ-PET. Similarly, self-reported (GDS-30) subscales of depression, apathy, anxiety, and cognitive concern were associated with higher Aβ-PET. When simultaneously entered, only self-reported cognitive concern was associated with Aβ-PET in the GDS-30 model, while both informant-reported anxiety and depression were associated with Aβ-PET in the NPI-Q model. Clinical status moderated the association between self-reported NPS and Aβ-PET such that the positive relationship between self-perceived NPS and Aβ burden strengthened with increasing functional difficulties. CONCLUSIONS In a cohort of older adults without dementia, both self- and informant-reported measures of global NPS, particularly patient-reported cognitive concerns and informant-reported anxiety and depression, corresponded with cerebral Aβ burden. NPS may appear early in the prodromal disease state and relate to initial AD proteinopathy burden, a relationship further exaggerated in those with greater clinical severity.
Collapse
Affiliation(s)
- Eva Q Gontrum
- UCSF, Memory and Aging Center, San Francisco, California, USA,
| | | | - Shannon Lee
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | - Valentina Diaz
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | - Alexander Ehrenberg
- UCSF, Memory and Aging Center, San Francisco, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
- Innovative Genomics Institute, University of California, Berkeley, California, USA
| | - Rowan Saloner
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | - Nidhi S Mundada
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | - Renaud La Joie
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | - Gil Rabinovici
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | - Joel H Kramer
- UCSF, Memory and Aging Center, San Francisco, California, USA
| | | |
Collapse
|
47
|
Gu X, Qi L, Qi Q, Zhou J, Chen S, Wang L. Monoclonal antibody therapy for Alzheimer's disease focusing on intracerebral targets. Biosci Trends 2024; 18:49-65. [PMID: 38382942 DOI: 10.5582/bst.2023.01288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Due to the complexity of the disorder and the presence of the blood-brain barrier (BBB), its drug discovery and development are facing enormous challenges, especially after several failures of monoclonal antibody (mAb) trials. Nevertheless, the Food and Drug Administration's approval of the mAb aducanumab has ushered in a new day. As we better understand the disease's pathogenesis and identify novel intracerebral therapeutic targets, antibody-based therapies have advanced over the past few years. The mAb drugs targeting β-amyloid or hyperphosphorylated tau protein are the focus of the current research. Massive neuronal loss and glial cell-mediated inflammation are also the vital pathological hallmarks of AD, signaling a new direction for research on mAb drugs. We have elucidated the mechanisms by which AD-specific mAbs cross the BBB to bind to targets. In order to investigate therapeutic approaches to treat AD, this review focuses on the promising mAbs targeting intracerebral dysfunction and related strategies to cross the BBB.
Collapse
Affiliation(s)
- Xiaolei Gu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Long Qi
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Qing Qi
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Song Chen
- Postdoctoral Station of Xiamen University, Fujian, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
48
|
Xing S, Tang X, Wang L, Wang J, Lv B, Wang X, Guo C, Zhao Y, Feng F, Liu W, Chen Y, Sun H. Optimizing drug-like properties of selective butyrylcholinesterase inhibitors for cognitive improvement: Enhancing aqueous solubility by disrupting molecular plane. Eur J Med Chem 2024; 268:116289. [PMID: 38452730 DOI: 10.1016/j.ejmech.2024.116289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/19/2024] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
Most recently, worldwide interest in butyrylcholinesterase (BChE) as a potential target for treating Alzheimer's disease (AD) has increased. In this study, the previously obtained selective BChE inhibitors with benzimidazole-oxadiazole scaffold were further structurally modified to increase their aqueous solubility and pharmacokinetic (PK) characteristics. S16-1029 showed improved solubility (3280 μM, upgraded by 14 times) and PK parameters, including plasma exposure (AUC0-inf = 1729.95 ng/mL*h, upgraded by 2.6 times) and oral bioavailability (Fpo = 48.18%, upgraded by 2 times). S16-1029 also displayed weak or no inhibition against Cytochrome P450 (CYP450) and human ether a-go-go related gene (hERG) potassium channel. In vivo experiments on tissue distribution revealed that S16-1029 could cross the blood-brain barrier (BBB) and reach the central nervous system (CNS). In vivo cognitive improvement efficacy and good in vitro target inhibitory activity (eqBChE IC50 = 11.35 ± 4.84 nM, hBChE IC50 = 48.1 ± 11.4 nM) were also assured. The neuroprotective effects against several AD pathology characteristics allowed S16-1029 to successfully protect the CNS of progressed AD patients. According to the findings of this study, altering molecular planarity might be a viable strategy for improving the drug-like property of CNS-treating drugs.
Collapse
Affiliation(s)
- Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xu Tang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Leyan Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Jun Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Xiaolong Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Can Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Ye Zhao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Feng Feng
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, People's Republic of China; Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
49
|
Navarro-Hortal M, Romero-Márquez JM, López-Bascón MA, Sánchez-González C, Xiao J, Sumalla-Cano S, Battino M, Forbes-Hernández TY, Quiles JL. In Vitro and In Vivo Insights into a Broccoli Byproduct as a Healthy Ingredient for the Management of Alzheimer's Disease and Aging through Redox Biology. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5197-5211. [PMID: 38477041 PMCID: PMC10941188 DOI: 10.1021/acs.jafc.3c05609] [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: 08/17/2023] [Revised: 01/15/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Broccoli has gained popularity as a highly consumed vegetable due to its nutritional and health properties. This study aimed to evaluate the composition profile and the antioxidant capacity of a hydrophilic extract derived from broccoli byproducts, as well as its influence on redox biology, Alzheimer's disease markers, and aging in the Caenorhabditis elegans model. The presence of glucosinolate was observed and antioxidant capacity was demonstrated both in vitro and in vivo. The in vitro acetylcholinesterase inhibitory capacity was quantified, and the treatment ameliorated the amyloid-β- and tau-induced proteotoxicity in transgenic strains via SOD-3 and SKN-1, respectively, and HSP-16.2 for both parameters. Furthermore, a preliminary study on aging indicated that the extract effectively reduced reactive oxygen species levels in aged worms and extended their lifespan. Utilizing broccoli byproducts for nutraceutical or functional foods could manage vegetable processing waste, enhancing productivity and sustainability while providing significant health benefits.
Collapse
Affiliation(s)
- María
D. Navarro-Hortal
- Department
of Physiology, Institute of Nutrition and
Food Technology “José Mataix Verdú”, Biomedical
Research Centre, University of Granada, 18016 Armilla, Spain
| | - Jose M. Romero-Márquez
- Department
of Physiology, Institute of Nutrition and
Food Technology “José Mataix Verdú”, Biomedical
Research Centre, University of Granada, 18016 Armilla, Spain
| | - M. Asunción López-Bascón
- Research
and Development Functional Food Centre (CIDAF), Health Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
| | - Cristina Sánchez-González
- Department
of Physiology, Institute of Nutrition and
Food Technology “José Mataix Verdú”, Biomedical
Research Centre, University of Granada, 18016 Armilla, Spain
- Sport
and Health Research Centre, University of
Granada, C/Menéndez
Pelayo 32, 18016 Granada, Spain
| | - Jianbo Xiao
- Department
of Analytical Chemistry and Food Science, Faculty of Food Science
and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain
| | - Sandra Sumalla-Cano
- Research
Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, 21, 39011 Santander, Spain
- Department
of Health, Nutrition and Sport, Iberoamerican
International University, Campeche 24560, Mexico
| | - Maurizio Battino
- Research
Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, 21, 39011 Santander, Spain
- Department
of Clinical Sciences, Polytechnic University
of Marche, 60131 Ancona, Italy
- International
Joint Research Laboratory of Intelligent Agriculture and Agri-Products
Processing, Jiangsu University, Zhenjiang 212013, China
| | - Tamara Y. Forbes-Hernández
- Department
of Physiology, Institute of Nutrition and
Food Technology “José Mataix Verdú”, Biomedical
Research Centre, University of Granada, 18016 Armilla, Spain
| | - José L. Quiles
- Department
of Physiology, Institute of Nutrition and
Food Technology “José Mataix Verdú”, Biomedical
Research Centre, University of Granada, 18016 Armilla, Spain
- Research
and Development Functional Food Centre (CIDAF), Health Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
- Research
Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, 21, 39011 Santander, Spain
| |
Collapse
|
50
|
Wang J, Zhen Y, Yang J, Yang S, Zhu G. Recognizing Alzheimer's disease from perspective of oligodendrocytes: Phenomena or pathogenesis? CNS Neurosci Ther 2024; 30:e14688. [PMID: 38516808 PMCID: PMC10958408 DOI: 10.1111/cns.14688] [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: 12/11/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Accumulation of amyloid beta, tau hyperphosphorylation, and microglia activation are the three highly acknowledged pathological factors of Alzheimer's disease (AD). However, oligodendrocytes (OLs) were also widely investigated in the pathogenesis and treatment for AD. AIMS We aimed to update the regulatory targets of the differentiation and maturation of OLs, and emphasized the key role of OLs in the occurrence and treatment of AD. METHODS This review first concluded the targets of OL differentiation and maturation with AD pathogenesis, and then advanced the key role of OLs in the pathogenesis of AD based on both clinic and basic experiments. Later, we extensively discussed the possible application of the current progress in the diagnosis and treatment of this complex disease. RESULTS Molecules involving in OLs' differentiation or maturation, including various transcriptional factors, cholesterol homeostasis regulators, and microRNAs could also participate in the pathogenesis of AD. Clinical data point towards the impairment of OLs in AD patients. Basic research further supports the central role of OLs in the regulation of AD pathologies. Additionally, classic drugs, including donepezil, edaravone, fluoxetine, and clemastine demonstrate their potential in remedying OL impairment in AD models, and new therapeutics from the perspective of OLs is constantly being developed. CONCLUSIONS We believe that OL dysfunction is one important pathogenesis of AD. Factors regulating OLs might be biomarkers for early diagnosis and agents stimulating OLs warrant the development of anti-AD drugs.
Collapse
Affiliation(s)
- Jingji Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases)Anhui University of Chinese MedicineHefeiChina
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui ProvinceThe Second Affiliation Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Yilan Zhen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases)Anhui University of Chinese MedicineHefeiChina
| | - Jun Yang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases)Anhui University of Chinese MedicineHefeiChina
- The First Affiliation Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Shaojie Yang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases)Anhui University of Chinese MedicineHefeiChina
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases)Anhui University of Chinese MedicineHefeiChina
| |
Collapse
|