1
|
Wang Y, Xie D, Ma S, Wang Y, Zhang C, Chen Z. Beta-asarone alleviated cerebral ischemia/reperfusion injury by targeting PINK1/Parkin-dependent mitophagy. Eur J Pharmacol 2025; 1002:177831. [PMID: 40490171 DOI: 10.1016/j.ejphar.2025.177831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 05/19/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) describes a secondary type of brain damage that happens when blood flow is restored to brain tissue; it ranks among the primary contributors of disability and mortality. The activation of PINK1/Parkin-mediated mitophagy exerts neuroprotective effects during CIRI. Beta-asarone (β-ASA), the principal active component of traditional natural drugs such as Acori tatarinowii rhizoma and Ligusticum chuanxiong Hort, possesses anti-inflammatory, antioxidant, and autophagy-enhancing properties. However, whether β-ASA can ameliorate CIRI by regulating the PINK1/Parkin-dependent mitophagy pathway remains unclear and warrants further investigation. The purpose of this study is to explore the underlying mechanism through which β-ASA influences PINK1/Parkin-mediated mitophagy in the hippocampus following ischemia-reperfusion. In the results section, the present study examined the effects of β-ASA on middle cerebral artery occlusion/reperfusion (MCAO/R)-induced neurological deficits using the Longa test and TTC staining, rats were then treated with β-ASA (20, 40, and 80 mg/kg.). The findings demonstrate that β-ASA promotes functional recovery in post-ischemic stroke, as evidenced by improved neurological function, reduced infarct volume, decreased neuronal damage, and lowered neuronal apoptosis. Furthermore, β-ASA significantly enhanced autophagy by increasing Beclin1 expression while reducing P62 and LC3-I/LC3-II expression. Additionally, β-ASA markedly activated PINK1/Parkin-mediated mitophagy. Finally, the introduction of mitophagy inhibitors was employed to clarify the relationship between autophagy and β-ASA, indicating that β-ASA promotes autophagy by activating the PINK1/Parkin signalling pathway. In conclusion, this study elucidates that β-ASA alleviates cerebral infarction, neurological impairment, and neuronal damage by targeting PINK1/Parkin-dependent mitophagy, thereby presenting a potential therapeutic strategy for CIRI.
Collapse
Affiliation(s)
- Yujiao Wang
- The First Clinical Medical College of Anhui University of Chinese Medicine, 230036, Hefei, Anhui, China.
| | - Daojun Xie
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, 230036, Hefei, Anhui, China.
| | - Shijia Ma
- The First Clinical Medical College of Anhui University of Chinese Medicine, 230036, Hefei, Anhui, China.
| | - Yuhe Wang
- The First Clinical Medical College of Anhui University of Chinese Medicine, 230036, Hefei, Anhui, China.
| | - Chengcheng Zhang
- The First Clinical Medical College of Anhui University of Chinese Medicine, 230036, Hefei, Anhui, China.
| | - Zhuyue Chen
- The First Clinical Medical College of Anhui University of Chinese Medicine, 230036, Hefei, Anhui, China.
| |
Collapse
|
2
|
Gupta S, Gupta S, Singh M, Patel AK. Anticancer potential of exosome-like nanoparticles isolated from Acorus calamus in breast cancer. 3 Biotech 2025; 15:186. [PMID: 40421230 PMCID: PMC12103446 DOI: 10.1007/s13205-025-04349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Plant-derived exosome-like nanoparticles represent a novel class of plant-based therapeutics with potential anti-cancer applications. The present study aims to isolate, characterize, and evaluate the Acorus calamus-derived exosome-like nanoparticles (ACENPs) for their cytotoxic and apoptotic effects on breast cancer cells. Nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM) revealed that ACENPs exhibited a mean hydrodynamic diameter of 122.4 ± 5.0 nm and a particle concentration of 1.58 × 1011 particles/mL. Cellular uptake studies confirmed the efficient internalization of ACENPs in MCF-7, MDA-MB-453, and MDA-MB-231 breast cancer cells. Cytotoxicity assay demonstrated a significant reduction in cell viability by 17.6%, 25%, and 35.8% in MCF-7, MDA-MB-231, and MDA-MB-453 breast cancer cells, respectively. Apoptosis induction was validated through AO/EB staining, DAPI nuclear fragmentation assays, annexin V-FITC/PI staining, and Western blot analysis of apoptosis-related proteins. Treatment with ACENPs resulted in an increased Bax/Bcl-2 ratio, indicating apoptotic activation. Metabolomic profiling identified bioactive compounds such as arecoline, trigonelline, asarone, and gingerol, known for their anti-cancer properties. Our study findings highlight that ACENPs could be utilized as a promising therapeutic approach for breast cancer treatment. We propose to conduct future research focusing on in vivo validation and optimizing large-scale production for clinical translation. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04349-8.
Collapse
Affiliation(s)
- Sunny Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016 India
| | - Shipra Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016 India
| | - Manju Singh
- All India Institute of Ayurveda Delhi, New Delhi, 110076 India
| | - Ashok Kumar Patel
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016 India
| |
Collapse
|
3
|
Lin Z, Sun M. Phytochemical regulation of CaMKII in Alzheimer's disease: A review of molecular mechanisms and therapeutic potential. Pharmacol Res 2025; 216:107790. [PMID: 40409522 DOI: 10.1016/j.phrs.2025.107790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/08/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder that leads to cognitive decline. CaMKII is a calcium-regulated kinase that is crucial for synaptic plasticity and memory. Phytochemicals with diverse origins, safety, and biological activity have attracted considerable attention in AD research. This systematic analysis of phytochemicals targeting CaMKII reveals their neuroprotective mechanisms against AD pathogenesis, highlighting CaMKII as a promising therapeutic target that warrants further preclinical investigation and drug development. We conducted a comprehensive review of the literature of phytochemicals that target CaMKII as a protective mechanism against AD. The search was conducted across multiple databases, including PubMed, Web of Science, China National Knowledge Internet, and Google Scholar, and covered the period from January 2000 to October 2024. A total of 301 articles were retrieved, of which 22 articles were included. The results showed that flavonoid, glycoside, terpene, and polyphenol analogs positively regulated CaMKII expression, whereas alkaloid analogs negatively regulated CaMKII expression. Different components of traditional Chinese medicine played different roles in CaMKII expression. Flavonoid compounds upregulated the expression of SYN, PSD-95, MAP2, and GluR1 to exert neuroprotective effects. Alkaloid and glycoside analogs inhibited Aβ deposition and tau hyperphosphorylation. Terpene analogs upregulated the SYN, PSD-95, NMDAR, BDNF, and PI3K/Akt signaling pathways to exert neuroprotection. Polyphenol analogs upregulated PSD-95, Munc18-1, SNAP25, SYN, and BDNF to exert neuroprotective effects. Emerging evidence demonstrates that select phytochemicals and traditional Chinese medicine compounds exert neuroprotective effects in AD by modulating CaMKII activity, thereby reducing Aβ accumulation, attenuating tau hyperphosphorylation, and enhancing synaptic plasticity, suggesting promising therapeutic potential.
Collapse
Affiliation(s)
- Zhongying Lin
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Miao Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
4
|
Wang J, Yang J, Lu J, Lu J, Ling B, Song Y, Chen G. Ursonic Acid Ameliorates H 2O 2-Induced Oxidative Damage in PC12 Cells and Prolonged the Lifespan in C. elegans by Activating MAPKs/Nrf2/HO-1 Signaling Pathway. Mol Neurobiol 2025; 62:6598-6611. [PMID: 39849276 DOI: 10.1007/s12035-025-04701-2] [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: 03/18/2024] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
Growing evidence suggests that plant compounds are emerging as a tremendous source for slowing the onset and progression of Alzheimer's disease (AD). Ursonic acid (UNA) is a naturally occurring pentacyclic triterpenoid with some hypoglycemic, anticancer, and antiinflammatory activities. However, the pharmacological effects of UNA on AD are still unknown. In the present experiments, the effects and mechanisms of UNA on AD were firstly explored by H2O2-induced PC12 cells and Aβ1-42-induced AD model of Caenorhabditis elegans (C. elegans). The results showed that UNA increased the cell viability and the level of mitochondrial membrane potential and reversed apoptosis in H2O2-induced PC12 cells, and Nrf2 and HO-1 were involved in this process. UNA also decreased the phosphorylation of related proteins in the MAPK signaling pathway. Meanwhile, we found that UNA upregulated cellular catalase levels and decreased ROS production. Besides, UNA could activate the Nrf2 /HO-1 signaling pathway and upregulate its protein expression under oxidative stress conditions, whereas Nrf2 and HO-1 inhibitors partially eliminated the protective ability of UNA. The results of in vivo experiments showed that UNA prolonged the lifespan and enhanced the health parameters of AD C. elegans and reduced Aβ1-42-induced neurotoxicity and ROS levels. Moreover, UNA increased GFP-tagged LGG-1 puncta in DA2123 C. elegans. Further mechanistic studies suggested that the protection of UNA on AD C. elegans may be through modulation of the MAPKs/Nrf2/HO-1 pathway. These results highlight the neuroprotective potential of UNA and suggest that UNA may be an effective therapeutic agent for alleviating AD.
Collapse
Affiliation(s)
- Jiahui Wang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Jia Yang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Jia Lu
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Jingshi Lu
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Bai Ling
- Department of Pharmacy, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Yan Song
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.
| | - Guangtong Chen
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.
| |
Collapse
|
5
|
Bu Y, Li S, Ye T, Wang Y, Song M, Chen J. Volatile oil of Acori tatarinowii rhizoma: potential candidate drugs for mitigating dementia. Front Pharmacol 2025; 16:1552801. [PMID: 40337511 PMCID: PMC12055781 DOI: 10.3389/fphar.2025.1552801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Objective This study aims to elucidate the mitigating effects of the volatile oil of Acori tatarinowii rhizoma (ATR) on dementia, in order to provide a reference for future research and applications of the volatile oil of ATR in the field of dementia. Materials and methods A search strategy was developed using terms such as "Acori tatarinowii rhizoma," "Acorus tatarinowii Schott," "Asarone," and "Dementia." The literature search was conducted in PubMed, Web of Science, and Google Scholar, and studies not meeting the inclusion criteria were excluded. This study summarizes the main metabolites, active ingredients, toxicological properties, and pharmacokinetic characteristics of the volatile oil from ATR in mitigating dementia, with a particular focus on its potential mechanisms of action. Furthermore, the study highlights the limitations of existing research and offers insights into future research directions. Results The volatile oil of ATR mitigates dementia through multiple pathways, including reducing abnormal protein aggregation, promoting neurogenesis, inhibiting neuronal apoptosis, regulating neurotransmitters, improving synaptic function, modulating autophagy, countering cellular stress, reducing neuroinflammation, and alleviating vascular risk factors. Conclusion The multi-pathway pharmacological effects of the volatile oil of ATR are well-aligned with the complex mechanisms of dementia progression, highlighting its significant therapeutic potential for anti-dementia applications. This provides new perspectives for the development of more effective anti-dementia drugs. Nonetheless, further rigorous and high-quality preclinical and clinical investigations are required to address key issues, including the chemical characterization of the volatile oil of ATR, potential synergistic effects among active ingredients, toxicity profiles, and definitive clinical efficacy.
Collapse
Affiliation(s)
- Yifan Bu
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Songzhe Li
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ting Ye
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuqing Wang
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mingrong Song
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Chen
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Alanazi MM, Albaker AB, Alzaagi LA, Alsabhan JF, Alasmari F, Almutairi MM, Alharbi MS, Alasmari AF, Alqahtani F, Alsanea S. Oxytocin Protects PC12 Cells Against β-Amyloid-Induced Cell Injury. Pharmaceuticals (Basel) 2025; 18:390. [PMID: 40143166 PMCID: PMC11944556 DOI: 10.3390/ph18030390] [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: 01/28/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Neurodegenerative diseases, particularly Alzheimer's disease (AD), are characterized by progressive cognitive decline and non-cognitive symptoms that significantly affect health and quality of life. Beta-amyloid (Aβ) protein accumulation is a key factor in AD pathology, leading to neuronal damage. Oxytocin (OXT), a neuropeptide with neuroprotective potential, has garnered interest owing to its ability to mitigate neurotoxicity. We hypothesized that oxytocin could protect PC12 cells from Aβ-induced cytotoxicity through antioxidant effects and modulation of apoptotic pathways (i.e., mitochondrial and MAPK pathways). In this study, we aim to assess oxytocin's protective effects on cell viability, oxidative stress, mitochondrial function, and apoptotic signaling. Methods: PC12 cells were treated with Aβ25-35 and pre-treated with varying oxytocin concentrations to assess cell viability, reactive oxygen species (ROS) generation, and mitochondrial membrane potential. Western blotting was performed to analyze the effects on mitochondrial apoptosis and MAPK pathways. Results: Oxytocin treatment significantly improved cell viability in a dose-dependent manner and reduced Aβ-induced oxidative stress and mitochondrial dysfunction. Oxytocin-treated groups exhibited decreased ROS levels, increased mitochondrial membrane potential, and modulation of apoptosis-related proteins. Oxytocin upregulated phosphorylated ERK1/2 and Bcl-2 while downregulating BAX and caspase-3, reducing the BAX/Bcl-2 ratio. Conclusions: Oxytocin effectively protects PC12 cells from Aβ-induced neurotoxicity, highlighting its potential as a therapeutic agent for AD. Further research is needed to clarify oxytocin's mechanisms and clinical implications in AD treatment.
Collapse
Affiliation(s)
- Mohammed Mufadhe Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Awatif B. Albaker
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Lamia A. Alzaagi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Jawza F. Alsabhan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab S. Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
7
|
Wang Y, Zhuang Z, He G, Zalán Z, Shi H, Du M, Kan J, Cai T, Chen K. A preliminary study of combined toxicity and underlying mechanisms of imidacloprid and cadmium coexposure using a multiomics integration approach. Toxicology 2025; 511:154063. [PMID: 39842396 DOI: 10.1016/j.tox.2025.154063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Imidacloprid (IMI) and cadmium (Cd) have been shown to be harmful to mammals separately, but their combined toxicity to mammals remains largely unknown. In this study, biochemical analysis (oxidative stress and serum indicators of liver and kidney function), pathological sections and multiomics (metabolomics and transcriptomics) methods were used to investigate the changes and mechanisms of liver and kidney in mice coexposed to IMI and Cd. Biochemical analysis and pathological section results showed that oxidative stress, organ function, and cell damage were aggravated after the combination of the two methods. Omics results revealed the following mechanism: When mouse liver and kidney cells were threatened by the external environment, mitochondrial DNA was inhibited, which leads to changes in energy metabolism. In this process, lipid metabolism and amino acid metabolism were disordered, resulting in the inhibition of substances related to lipid metabolism and amino acid metabolism that protect the body from oxidative damage, and then showed more serious liver and kidney oxidative stress and liver and kidney function and cell damage. This research offers novel insights for the assessment of the safety profile associated with the concurrent exposure of the two chemicals in mammalian species.
Collapse
Affiliation(s)
- Yuankai Wang
- College of Food Science, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China; Laboratory of Quality & Safety Risk Assessment for Agro-products on Storage and Preservation (Chongqing), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, PR China
| | - Ziyue Zhuang
- College of Food Science, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China; Laboratory of Quality & Safety Risk Assessment for Agro-products on Storage and Preservation (Chongqing), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, PR China
| | - Guangyun He
- Institute of Quality Standard and Testing Technology for Agro-Products, Sichuan Academy of Agricultural Sciences, Chengdu 610066, PR China
| | - Zsolt Zalán
- Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Buda Campus, Villányi str. 29-43, Budapest 1118, Hungary
| | - Hui Shi
- College of Food Science, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China; Laboratory of Quality & Safety Risk Assessment for Agro-products on Storage and Preservation (Chongqing), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, PR China
| | - Muying Du
- College of Food Science, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China; Laboratory of Quality & Safety Risk Assessment for Agro-products on Storage and Preservation (Chongqing), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, PR China
| | - Jianquan Kan
- College of Food Science, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China; Laboratory of Quality & Safety Risk Assessment for Agro-products on Storage and Preservation (Chongqing), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, PR China
| | - Tian Cai
- School of Chemistry and Chemical Engineering, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China.
| | - Kewei Chen
- College of Food Science, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, PR China; Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, PR China; Laboratory of Quality & Safety Risk Assessment for Agro-products on Storage and Preservation (Chongqing), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Chongqing 400715, PR China; Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, PR China.
| |
Collapse
|
8
|
Zhao Y, Hu Y, Yang J, Qi Y, Miao J, Miao M. Network pharmacology and experimental validation reveal the mechanisms of sniffing essential oil of Acori Tatarinowii rhizoma in treating olfactory dysfunction. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118851. [PMID: 39326811 DOI: 10.1016/j.jep.2024.118851] [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: 06/05/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acorus tatarinowii Rhizoma, a traditional Chinese medicine known for open the orifices and transform phlegm, is used in the treatment of brain disorders. The essential oil of Acorus tatarinowii Rhizoma (EOAT) has demonstrated neuroprotective properties clinically. However, research into its effect on Olfactory Dysfunction (OD) remains limited. AIM OF THE STUDY This study aimed to investigate the effects and mechanisms of sniffing EOAT on improving olfactory function in a 3-Methylindole (3-MI)-induced OD mouse model. MATERIALS AND METHODS The research involved intraperitoneal injection of 3-MI to induce OD in mice. The effects of EOAT treatment were assessed on olfactory function, olfactory bulb (OB) pathology, inflammatory factors, olfactory marker protein (OMP), microglial activation, and related pathway proteins and mRNA. RESULTS Based on the GC-MS analysis results of EOAT and network pharmacology studies, we predicted 18 targets associated with the treatment of OD. SLC6A3, MAOB, DRD1, and PTGS2 were identified as the core targets of EOAT against OD. Molecular docking and KEGG enrichment results indicated that EOAT may exert anti-inflammatory effects by acting on the core target PTGS2, with its anti-inflammatory mechanism possibly related to the PI3K/Akt signaling pathway. Subsequent animal experiments confirmed that inhalation of EOAT significantly increased the body weight of OD model mice, shortened the foraging time, enhanced the expression of OMP in OB, reduced damage to the OB cells, and improved olfactory function. Meanwhile, EOAT significantly alleviated the inflammatory response in OB of OD model mice, inhibited the activation of microglial cells, and suppressed the expression of PI3K/Akt signaling pathway proteins and mRNA. CONCLUSION EOAT inhalation could improve olfactory function in 3-MI-induced OD model. The underlying mechanism may be related to the modulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yinan Zhao
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Yilong Hu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Jingying Yang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Yupu Qi
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Jinxin Miao
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Mingsan Miao
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
9
|
Wu J, Sun H, Zhao Y, Lian L, Bian H, Guo Y, Li D, Huang L. The spectrum-efficacy correlation of Kai-Xin-San for cognition of Aβ 42 transgenic Drosophila and verification of its active ingredients. Front Pharmacol 2025; 16:1538837. [PMID: 39936091 PMCID: PMC11811076 DOI: 10.3389/fphar.2025.1538837] [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: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction This study aims to establish the fingerprint spectra of Kai-Xin-San (KXS) and investigate its spectrum-effect relationship in treating Alzheimer's disease (AD). Methods Initially, the fingerprints of 15 batches of KXS were established and analyzed using HPLC, with the method's precision, stability, and repeatability thoroughly evaluated. Subsequently, the effects of the 15 batches of KXS were assessed in an olfactory escape memory experiment, utilizing Aβ42 transgenic drosophila as a model. Finally, the spectrum-effect relationship between the KXS fingerprint and memory improvement was analyzed, with the active ingredients subjected to validation testing. Results The results identified seventeen common peaks in the fingerprint, and eight active components were determined: polygalaxanthone III, 3-6-disinapoylsucrose, ginsenoside Rg1, ginsenoside Rb1, β-asarone, α-asarone, dehydrotumulosic acid, and dehydropachymic acid. Treatment with KXS (1%, for 4 days) significantly enhanced the performance index of Aβ42 flies in the olfactory experiment. Both spectrum-effect analysis and validation tests indicated that polygalaxanthone III, ginsenoside Rg1, ginsenoside Rb1, β-asarone, and α-asarone were positively correlated with the performance index and improved the performance index in the olfactory experiment. The HPLC fingerprint method for KXS demonstrated excellent precision, accuracy, and reproducibility, making it suitable for quality evaluation and control of KXS. Polygalaxanthone III, ginsenoside Rg1, ginsenoside Rb1, β-asarone, and α-asarone are identified as potential active ingredients of KXS for anti-AD effects. Discussion These findings provide an experimental basis for developing new drugs based on KXS and its active ingredient combinations.
Collapse
Affiliation(s)
- Jinfu Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hang Sun
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yiyang Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lian Lian
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongsheng Bian
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yong Guo
- Shineway Pharmaceutical Group Co., Ltd., Shijiazhuang, China
| | - Dan Li
- Shineway Pharmaceutical Group Co., Ltd., Shijiazhuang, China
| | - Lili Huang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
10
|
Kaur K, Narang RK, Singh S. Role of Nrf2 in Oxidative Stress, Neuroinflammation and Autophagy in Alzheimer's Disease: Regulation of Nrf2 by Different Signaling Pathways. Curr Mol Med 2025; 25:372-387. [PMID: 37493162 DOI: 10.2174/1566524023666230726145447] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023]
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disorder and the leading cause of dementia. AD is characterized by the aggregation of amyloid-ß (Aß) peptide, increased levels of tau protein, and loss of redox homeostasis responsible for mitochondrial dysfunction, oxidative stress, and neuroinflammation. Excessive accumulation of toxic Aß plaques activates microglia, which initiates neuroinflammation and consequently accelerates synaptic damage and neuronal loss. Various proinflammatory cytokines release, microglia proliferation, reactive astrocyte, and oxidative (reactive oxygen species (ROS) production, level of antioxidant enzymes, redox homeostasis, and lipid peroxidation) stress play a major role in AD. Several studies revealed that nuclear factor erythroid 2-related factor 2 (Nrf2) regulates redox homeostasis and works as an anti-inflammatory in various neurodegenerative disorders. D-Glutamate expression of transcription factor Nrf2 and its genes (glutamate-cysteine ligase catalytic subunit (GCLC), Heme oxygenase-1 (HO-1), and NADPH quinone oxidoreductase I (NQO1)) has been found in AD. Nrf2-HO-1 enhances the expression of antioxidant genes, inhibits microglia-mediated inflammation, and boosts mitochondrial function, suggesting that modulators of this protein may be useful to manage AD. This review focuses on the role of Nrf2 in AD, with a particular emphasis on the various pathways involved in the positive and negative modulation of Nrf2, namely Phosphoinositide 3-kinase (PI3K), Glycogen synthase kinase-3 (GSK-3), Nuclear factor kappa-B (NF-κB), and p38Mitogen-activated protein kinases (p38MAPK). Also, we have discussed the progress and challenges regarding the Nrf2 activators for AD treatment.
Collapse
Affiliation(s)
- Karamjeet Kaur
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India affiliated to IKG- Punjab Technical University, Jalandhar Punjab, 144603, India
| | - Raj Kumar Narang
- Nanomedicine Research Centre, Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| |
Collapse
|
11
|
Xia M, Yi M, Guo C, Xie Y, Yu W, Wang D, Dai X. β-Asarone regulates microglia polarization to alleviate TBI-induced nerve damage via Fas/FasL signaling axis. Hum Cell 2024; 38:33. [PMID: 39718669 DOI: 10.1007/s13577-024-01161-z] [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: 08/23/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Acute injury and secondary injury caused by traumatic brain injury (TBI) seriously threaten the health of patients. The purpose of this study was to investigate the role of β-Asarone in TBI-induced neuroinflammation and injury. In this work, the effects of β-Asarone on nerve injury and neuronal apoptosis were investigated in mice with TBI by controlled cortical impingement. The results of this research implied that β-Asarone dose-dependently decreased the mNSS score, brain water content and neuronal apoptosis, but increased the levels of the axonal markers Nrp-1 and Tau in TBI mice. In addition, β-Asarone caused a decrease in the levels of Fas, FasL, and inflammatory factors in cerebrospinal fluid and serum of TBI mice. Therefore, β-Asarone inhibited neuroinflammation and promoted axon regeneration in TBI mice. Besides, β-Asarone treatment inhibited M1 phenotype polarization but promoted M2 phenotype polarization in microglia of TBI mice. Overexpression of Fas and FasL reversed the above effects of β-Asarone. Thus, β-Asarone regulated microglial M1/M2 polarization balance in TBI mice by suppressing Fas/FasL signaling axis. In conclusion, β-Asarone inhibited Fas/FasL signaling pathway to promote the M1/M2 polarization balance of microglia toward M2 polarization, thus alleviating TBI-induced nerve injury.
Collapse
Affiliation(s)
- Mingyue Xia
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Chunyuan Guo
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- Jiangxi Provincial, People's Hospital, Clinical College of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yeli Xie
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
| | - Wenting Yu
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
| | - Dongsheng Wang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingping Dai
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Jiangxi Hospital, National Reginal Center for Neurological Disease, Honggutan District, No.266 Fenghe North Avenue, Nanchang, 330038, Jiangxi, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Changsha, Hunan, China.
| |
Collapse
|
12
|
Gu S, Wang R, Zhang W, Wen C, Chen C, Liu S, Lei Q, Zhang P, Zeng S. The production, function, and clinical applications of IL-33 in type 2 inflammation-related respiratory diseases. Front Immunol 2024; 15:1436437. [PMID: 39301028 PMCID: PMC11410612 DOI: 10.3389/fimmu.2024.1436437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Epithelial-derived IL-33 (Interleukin-33), as a member of alarm signals, is a chemical substance produced under harmful stimuli that can promote innate immunity and activate adaptive immune responses. Type 2 inflammation refers to inflammation primarily mediated by Type 2 helper T cells (Th2), Type 2 innate lymphoid cells (ILC2), and related cytokines. Type 2 inflammation manifests in various forms in the lungs, with diseases such as asthma and chronic obstructive pulmonary disease chronic obstructive pulmonary disease (COPD) closely associated with Type 2 inflammation. Recent research suggests that IL-33 has a promoting effect on Type 2 inflammation in the lungs and can be regarded as an alarm signal for Type 2 inflammation. This article provides an overview of the mechanisms and related targets of IL-33 in the development of lung diseases caused by Type 2 inflammation, and summarizes the associated treatment methods. Analyzing lung diseases from a new perspective through the alarm of Type 2 inflammation helps to gain a deeper understanding of the pathogenesis of these related lung diseases. This, in turn, facilitates a better understanding of the latest treatment methods and potential therapeutic targets for diseases, with the expectation that targeting lL-33 can propose new strategies for disease prevention.
Collapse
Affiliation(s)
- Shiyao Gu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruixuan Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wantian Zhang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Cen Wen
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Su Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
13
|
Chen H, Zhang CJ, Zhao ZY, Gao YY, Zhao JT, Li XX, Zhang M, Wang H. Mechanisms underlying LncRNA SNHG1 regulation of Alzheimer's disease involve DNA methylation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:428-435. [PMID: 38551404 DOI: 10.1080/15287394.2024.2334248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease associated with long non-coding RNAs and DNA methylation; however, the mechanisms underlying the role of lncRNA small nucleolar RNA host gene 1 (lncRNA SNHG1) and subsequent involvement of DNA methylation in AD development are not known. The aim of this study was to examine the regulatory mechanisms attributed to lncRNA SNHG1 gene utilizing 2 strains of senescence-accelerated mouse prone 8 (SAMP8) model of AD and compared to senescence-accelerated mouse resistant (SAMR) considered a control. Both strains of the mouse were transfected with either blank virus, psLenti-U6-SNHG1(low gene expression) virus, and psLenti-pA-SNHG1(gene overexpression) virus via a single injection into the brains for 2 weeks. At 2 weeks mice were subjected to a Morris water maze to determine any behavioral effects followed by sacrifice to extract hippocampal tissue for Western blotting to measure protein expression of p-tau, DNMT1, DNMT3A, DNMT3B, TET1, and p-Akt. No marked alterations were noted in any parameters following blank virus transfection. In SAMP8 mice, a significant decrease was noted in protein expression of DNMT1, DNMT3A, DNMT3B, and p-Akt associated with rise in p-tau and TET1. Transfection with ps-Lenti-U6-SNHG1 alone in SAMR1 mice resulted in a significant rise in DNMTs and p-Akt and a fall in p-tau and TET1. Transfection of SAMP8 with ps-Lenti-U6-SNHG1 blocked effects on overexpression noted in this mouse strain. However, knockdown of lncRNA SNHG1 yielded the opposite results as found in SAMR1 mice. In conclusion, the knockdown of lncRNA SNHG1 enhanced DNA methylation through the PI3K/Akt signaling pathway, thereby reducing the phosphorylation levels of tau in SAMP8 AD model mice with ameliorating brain damage attributed to p-tau accumulation with consequent neuroprotection.
Collapse
Affiliation(s)
- Hong Chen
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Chun-Jie Zhang
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
- Center of Collaborative Innovation in Translational Medicine, Baotou Medical College, Inner Mongolia, China
| | - Zhi-Ying Zhao
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Yang-Yang Gao
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Jian-Tian Zhao
- Institute of Public Health, Baotou Medical College, Inner Mongolia, China
| | - Xiao-Xu Li
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Ming Zhang
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - He Wang
- School of Health Sciences, University of Newcastle, Newcastle, Australia
| |
Collapse
|
14
|
Qin Z, Xie L, Li W, Wang C, Li Y. New Insights into Mechanisms Traditional Chinese Medicine for Allergic Rhinitis by Regulating Inflammatory and Oxidative Stress Pathways. J Asthma Allergy 2024; 17:97-112. [PMID: 38405022 PMCID: PMC10888064 DOI: 10.2147/jaa.s444923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Allergy rhinitis (AR) is becoming more common and has serious medical and societal consequences. Sneezing, paroxysmal nasal blockage, nasal itching, mucosal edema, coughing, and rhinorrhea are symptoms of this type I allergic immunological illness. Immunoglobulin E-mediated inflammation is the cause of it. Because AR is prone to recurrent attacks, extended medication therapy may impair its effectiveness. In addition to negatively affecting the patients' physical health, this can also negatively impact their mental health. During AR development, there are inflammatory and oxidative stress responses that are linked to problems in a number of signal transduction pathways. By using the terms "allergic rhinitis", "traditional Chinese medicine", "inflammation", and "oxidative stress", we screened for pertinent research published over the previous five years in databases like PubMed. We saw that NF-KB, TLR, IL-33/ST2, PI3K/AKT, MAPK, and Nrf2 are some of the most important inflammatory and oxidative stress pathways in AR. Studies have revealed that antioxidant and anti-inflammatory therapy reduced the risk of AR and was therapeutic; however, the impact of the therapy varies widely. The Chinese medical system places a high value on traditional Chinese medicine (TCM), which has been there for virtually all of China's 5000-year history. By influencing signaling pathways related to inflammation and oxidative stress, Chinese herbal medicine and its constituent compounds have been shown to prevent allergic rhinitis. This review will focus on this evidence and provide references for clinical treatment and scientific research applications.
Collapse
Affiliation(s)
- Zhu Qin
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Liangzhen Xie
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Wentao Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Chao Wang
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Yan Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
15
|
Avola R, Furnari AG, Graziano ACE, Russo A, Cardile V. Management of the Brain: Essential Oils as Promising Neuroinflammation Modulator in Neurodegenerative Diseases. Antioxidants (Basel) 2024; 13:178. [PMID: 38397776 PMCID: PMC10886016 DOI: 10.3390/antiox13020178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Neuroinflammation, a pivotal factor in the pathogenesis of various brain disorders, including neurodegenerative diseases, has become a focal point for therapeutic exploration. This review highlights neuroinflammatory mechanisms that hallmark neurodegenerative diseases and the potential benefits of essential oils in counteracting neuroinflammation and oxidative stress, thereby offering a novel strategy for managing and mitigating the impact of various brain disorders. Essential oils, derived from aromatic plants, have emerged as versatile compounds with a myriad of health benefits. Essential oils exhibit robust antioxidant activity, serving as scavengers of free radicals and contributing to cellular defense against oxidative stress. Furthermore, essential oils showcase anti-inflammatory properties, modulating immune responses and mitigating inflammatory processes implicated in various chronic diseases. The intricate mechanisms by which essential oils and phytomolecules exert their anti-inflammatory and antioxidant effects were explored, shedding light on their multifaceted properties. Notably, we discussed their ability to modulate diverse pathways crucial in maintaining oxidative homeostasis and suppressing inflammatory responses, and their capacity to rescue cognitive deficits observed in preclinical models of neurotoxicity and neurodegenerative diseases.
Collapse
Affiliation(s)
- Rosanna Avola
- Faculty of Medicine and Surgery, University of Enna "Kore", 94100 Enna, Italy
| | | | | | - Alessandra Russo
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|
16
|
Hu Y, Guo H, Cheng S, Sun J, Du J, Liu X, Xiong Y, Chen L, Liu C, Wu C, Tian H. Functionalized Cerium Dioxide Nanoparticles with Antioxidative Neuroprotection for Alzheimer's Disease. Int J Nanomedicine 2023; 18:6797-6812. [PMID: 38026525 PMCID: PMC10658952 DOI: 10.2147/ijn.s434873] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Background Oxidative stress induced reactive oxygen species (ROS) and aggregation of amyloid β (Aβ) in the nervous system are significant contributors to Alzheimer's disease (AD). Cerium dioxide and manganese oxide are known as to be effective and recyclable ROS scavengers with high efficiency in neuroprotection. Methods A hollow-structured manganese-doped cerium dioxide nanoparticle (LMC) was synthesized for loading Resveratrol (LMC-RES). The LMC-RES were characterized by TEM, DLS, Zeta potential, and X-ray energy spectrum analysis. We also tested the biocompatibility of LMC-RES and the ability of LMC-RES to cross the blood-brain barrier (BBB). The antioxidant effects of LMC-RES were detected by SH-SY5Y cells. Small animal live imaging was used to detect the distribution of LMC-RES in the brain tissue of AD mice. The cognitive abilities of mice were tested by water maze and nesting experiments. The effects of LMC-RES in reducing oxidative stress and protecting neurons was also explored by histological analysis. Results The results showed that LMC-RES had good sustained release effect and biocompatibility. The drug release rate of LMC-RES at 24 hours was 80.9 ± 2.25%. Meanwhile, LMC-RES could cross the BBB and enrich in neurons to exert antioxidant effects. In Aβ-induced SH-SY5Y cells, LMC-RES could inhibits oxidative stress through the Nrf-2/HO-1 signaling pathway. In AD model mice, LMC-RES was able to reduce ROS levels, inhibit Aβ-induced neurotoxicity, and protect neurons and significantly improve cognitive deficits of AD mice after drug administration. Conclusion LMC-RES can effectively across the BBB, reduce oxidative stress, inhibit Aβ aggregation, and promote the recovery of neurological function.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Hui Guo
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Shuai Cheng
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Junpeng Sun
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Jiaqun Du
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Xiaobang Liu
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Ying Xiong
- Laboratoire Catalyse et Spectrochimie (LCS), Normandie Université, ENSICAEN, UNICAEN, CNRS, Caen, 14050, France
| | - Liqing Chen
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Chang Liu
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Chao Wu
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - He Tian
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| |
Collapse
|
17
|
Aleksandrova Y, Neganova M. Deciphering the Mysterious Relationship between the Cross-Pathogenetic Mechanisms of Neurodegenerative and Oncological Diseases. Int J Mol Sci 2023; 24:14766. [PMID: 37834214 PMCID: PMC10573395 DOI: 10.3390/ijms241914766] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The relationship between oncological pathologies and neurodegenerative disorders is extremely complex and is a topic of concern among a growing number of researchers around the world. In recent years, convincing scientific evidence has accumulated that indicates the contribution of a number of etiological factors and pathophysiological processes to the pathogenesis of these two fundamentally different diseases, thus demonstrating an intriguing relationship between oncology and neurodegeneration. In this review, we establish the general links between three intersecting aspects of oncological pathologies and neurodegenerative disorders, i.e., oxidative stress, epigenetic dysregulation, and metabolic dysfunction, examining each process in detail to establish an unusual epidemiological relationship. We also focus on reviewing the current trends in the research and the clinical application of the most promising chemical structures and therapeutic platforms that have a modulating effect on the above processes. Thus, our comprehensive analysis of the set of molecular determinants that have obvious cross-functional pathways in the pathogenesis of oncological and neurodegenerative diseases can help in the creation of advanced diagnostic tools and in the development of innovative pharmacological strategies.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 420088 Kazan, Russia
| |
Collapse
|
18
|
Duan F, Ju T, Song C, Liu M, Xiong Y, Han X, Lu W. Synergetic effect of β-asarone and cannabidiol against Aβ aggregation in vitro and in vivo. Comput Struct Biotechnol J 2023; 21:3875-3884. [PMID: 37602231 PMCID: PMC10432915 DOI: 10.1016/j.csbj.2023.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Alzheimer's disease (AD) is a complex and multifactorial neurodegenerative disorder, and it is unlikely that any single drug or intervention will be very successful. The pathophysiology of Alzheimer's disease involves a range of complicated biological processes, including the accumulation of beta-amyloid protein and tau protein. Given the complexity of AD and amyloid accumulation, a combination of interventions remains to be further explored. Here, we investigated the potential of combining β-asarone and cannabidiol (CBD) as a treatment for AD. The study analyzed the combined effects of these two phytochemicals on beta-amyloid (Aβ) protein aggregation and toxicity in bulk solution, in cells as well as in C.elegans. We detailed the morphological and size changes of Aβ40 aggregates in the presence of β-asarone and cannabidiol. More importantly, the presence of both compounds synergistically inhibited apoptosis and downregulated relative gene expression in cells, and that it may also slow aging, decrease the rate of paralysis, enhance learning capacity, and boost autophagy activity in C.elegans. Our studies suggest that multiple drugs, like β-asarone and CBD, may be potentially developed as a medicinal adjunct in the treatment of AD, although further clinical trials are needed to determine the efficacy and safety of this combination treatment in humans.
Collapse
Affiliation(s)
- Fangyuan Duan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Ting Ju
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Chen Song
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Mengyao Liu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Yi Xiong
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Xue Han
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
- The Intelligent Equipment Research Center for the Development of Special Medicine and Food Resources, Chongqing Research Institute of HIT. Harbin Institute of Technology, Chongqing 401120, China
| |
Collapse
|
19
|
Park C, Cha HJ, Hwangbo H, Bang E, Hong SH, Song KS, Noh JS, Kim DH, Kim GY, Choi YH. β-Asarone Alleviates High-Glucose-Induced Oxidative Damage via Inhibition of ROS Generation and Inactivation of the NF-κB/NLRP3 Inflammasome Pathway in Human Retinal Pigment Epithelial Cells. Antioxidants (Basel) 2023; 12:1410. [PMID: 37507949 PMCID: PMC10376195 DOI: 10.3390/antiox12071410] [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: 06/01/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of vision loss and a major complication of diabetes. Hyperglycemia-induced accumulation of reactive oxygen species (ROS) is an important risk factor for DR. β-asarone, a major component of volatile oil extracted from Acori graminei Rhizoma, exerts antioxidant effects; however, its efficacy in DR remains unknown. In this study, we investigated whether β-asarone inhibits high-glucose (HG)-induced oxidative damage in human retinal pigment epithelial (RPE) ARPE-19 cells. We found that β-asarone significantly alleviated cytotoxicity, apoptosis, and DNA damage in HG-treated ARPE-19 cells via scavenging of ROS generation. β-Asarone also significantly attenuated the excessive accumulation of lactate dehydrogenase and mitochondrial ROS by increasing the manganese superoxide dismutase and glutathione activities. HG conditions markedly increased the release of interleukin (IL)-1β and IL-18 and upregulated their protein expression and activation of the nuclear factor-kappa B (NF-κB) signaling pathway, whereas β-asarone reversed these effects. Moreover, expression levels of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome multiprotein complex molecules, including thioredoxin-interacting protein, NLRP3, apoptosis-associated speck-like protein containing a caspase-recruitment domain, and cysteinyl aspartate-specific proteinase-1, were increased in ARPE-19 cells under HG conditions. However, their expression levels remained similar to those in the control group in the presence of β-asarone. Therefore, β-asarone protects RPE cells from HG-induced injury by blocking ROS generation and NF-κB/NLRP3 inflammasome activation, indicating its potential as a therapeutic agent for DR treatment.
Collapse
Affiliation(s)
- Cheol Park
- Department Division of Basic Sciences, College of Liberal Studies, Dong-eui University, Busan 47340, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University, Busan 49104, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - EunJin Bang
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47340, Republic of Korea
| | - Kyoung Seob Song
- Department of Medical Life Science, College of Medicine, Kosin University, Busan 49104, Republic of Korea
| | - Jeong Sook Noh
- Department of Food Science & Nutrition, Tongmyong University, Busan 48520, Republic of Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, College of Fisheries Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47340, Republic of Korea
| |
Collapse
|
20
|
Zhao S, Zhang P, Yan Y, Xu W, Li J, Wang L, Wang N, Huang Y. Network pharmacology-based prediction and validation of the active ingredients and potential mechanisms of the Huangxiong formula for treating ischemic stroke. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116507. [PMID: 37080367 DOI: 10.1016/j.jep.2023.116507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/30/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangxiong Formula (HXF) is composed of four herbs: Rheum palmatum L., Ligusticum striatum DC., Curcuma aromatica Salisb., and Acorus gramineus Aiton. HXF is clinically used for the treatment of ischemic stroke (IS). However, its molecular mechanism remains unclear. AIM OF THE STUDY A network pharmacology-based strategy combined with experimental study in vivo and in vitro to were used to investigate the bioactive components, potential targets, and molecular mechanisms of HXF in the treatment of IS. MATERIALS AND METHODS The components of HXF were detected by ultra-performance liquid chromatography (UPLC). The potential active ingredients of HXF were acquired from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and literature, and corresponding targets were discerned through the Swiss TargetPrediction database. IS-related targets were obtained from Genecards, Online Mendelian Inheritance in Man (OMIM), Therapeutic Target Database (TTD), and DisGeNET. The intersection of ingredient and disease targets was screened, and a herbal-compound-target network was constructed. A protein-protein interaction (PPI) network was created, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. Based on these analyses, we established a compound-target-pathway (C-T-P) network. A cerebral ischemia-reperfusion (I/R) animal model was established, and the cerebral protective effect of HXF was assessed. The accuracy of the predicted targets was verified by real-time quantitative polymerase chain reaction (RT-qPCR). Hippocampal neuronal injury cell model induced by oxygen-glucose deprivation and reperfusion (OGD/R) was used to evaluate the protective effect of α-Asarone. Furthermore, molecular docking, drug affinity responsive target stability (DARTS) assay, and cellular thermal shift assay (CETSA) were performed to verify whether α-Asarone can bind to PI3K. RESULTS A total of 44 active ingredients and 795 gene targets were identified through network pharmacology. Network analysis showed that naringenin, eupatin, kaempferol, and α-Asarone were possible drug candidates. SRC, AKT1, TP53, MAPK3, STAT3, HRAS, CTNNB1, EGFR, VEGFA, PIK3R1 could serve as potential drug targets. KEGG analysis implied that the PI3K/AKT signaling pathway might play an important role in treating IS by HXF. Moreover, HXF significantly reduced neurological impairment, cerebral infarct volume, brain index, and brain histopathological damage in I/R rats. The mRNA expression of the top 10 potential targets was verified in the brain tissue. The C-T-P network and UPLC analysis suggested that α-Asarone might be an important component of HXF and can inhibit oxidative stress and apoptosis in HT22 cells by activating the PI3K/AKT signaling pathway. Molecular docking, DARTS, and CETSA assay analysis confirmed that there were direct interactions between α-Asarone and PI3K. CONCLUSION HXF had a therapeutic effect in IS with multi-component, multi-target, and multi-approach features. α-Asarone, identified as one of the major active components of HXF, could alleviate oxidative stress and apoptosis by targeting PI3K/AKT pathway.
Collapse
Affiliation(s)
- Saihong Zhao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Pingping Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yonghuan Yan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Weifang Xu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jiacheng Li
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ning Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China; Institute for the Evaluation of the Efficacy and Safety of Chinese Medicines, Anhui Academy of Chinese Medicine, Hefei, China.
| | - Yingying Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China; Institute for the Evaluation of the Efficacy and Safety of Chinese Medicines, Anhui Academy of Chinese Medicine, Hefei, China.
| |
Collapse
|
21
|
Zou C, Su L, Pan M, Chen L, Li H, Zou C, Xie J, Huang X, Lu M, Zou D. Exploration of novel biomarkers in Alzheimer's disease based on four diagnostic models. Front Aging Neurosci 2023; 15:1079433. [PMID: 36875704 PMCID: PMC9978156 DOI: 10.3389/fnagi.2023.1079433] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/25/2023] [Indexed: 02/18/2023] Open
Abstract
Background Despite tremendous progress in diagnosis and prediction of Alzheimer's disease (AD), the absence of treatments implies the need for further research. In this study, we screened AD biomarkers by comparing expression profiles of AD and control tissue samples and used various models to identify potential biomarkers. We further explored immune cells associated with these biomarkers that are involved in the brain microenvironment. Methods By differential expression analysis, we identified differentially expressed genes (DEGs) of four datasets (GSE125583, GSE118553, GSE5281, GSE122063), and common expression direction of genes of four datasets were considered as intersecting DEGs, which were used to perform enrichment analysis. We then screened the intersecting pathways between the pathways identified by enrichment analysis. DEGs in intersecting pathways that had an area under the curve (AUC) > 0.7 constructed random forest, least absolute shrinkage and selection operator (LASSO), logistic regression, and gradient boosting machine models. Subsequently, using receiver operating characteristic curve (ROC) and decision curve analysis (DCA) to select an optimal diagnostic model, we obtained the feature genes. Feature genes that were regulated by differentially expressed miRNAs (AUC > 0.85) were explored further. Furthermore, using single-sample GSEA to calculate infiltration of immune cells in AD patients. Results Screened 1855 intersecting DEGs that were involved in RAS and AMPK signaling. The LASSO model performed best among the four models. Thus, it was used as the optimal diagnostic model for ROC and DCA analyses. This obtained eight feature genes, including ATP2B3, BDNF, DVL2, ITGA10, SLC6A12, SMAD4, SST, and TPI1. SLC6A12 is regulated by miR-3176. Finally, the results of ssGSEA indicated dendritic cells and plasmacytoid dendritic cells were highly infiltrated in AD patients. Conclusion The LASSO model is the optimal diagnostic model for identifying feature genes as potential AD biomarkers, which can supply new strategies for the treatment of patients with AD.
Collapse
Affiliation(s)
- Cuihua Zou
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Li Su
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liechun Chen
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hepeng Li
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chun Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaohua Huang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Mengru Lu
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China.,Clinical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
22
|
Kim CJ, Kwak TY, Bae MH, Shin HK, Choi BT. Therapeutic Potential of Active Components from Acorus gramineus and Acorus tatarinowii in Neurological Disorders and Their Application in Korean Medicine. J Pharmacopuncture 2022; 25:326-343. [PMID: 36628348 PMCID: PMC9806153 DOI: 10.3831/kpi.2022.25.4.326] [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: 08/26/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 12/30/2022] Open
Abstract
Neurological disorders represent a substantial healthcare burden worldwide due to population aging. Acorus gramineus Solander (AG) and Acorus tatarinowii Schott (AT), whose major component is asarone, have been shown to be effective in neurological disorders. This review summarized current information from preclinical and clinical studies regarding the effects of extracts and active components of AG and AT (e.g., α-asarone and β-asarone) on neurological disorders and biomedical targets, as well as the mechanisms involved. Databases, including PubMed, Embase, and RISS, were searched using the following keywords: asarone, AG, AT, and neurological disorders, including Alzheimer's disease, Parkinson's disease, depression and anxiety, epilepsy, and stroke. Meta-analyses and reviews were excluded. A total of 873 studies were collected. A total of 89 studies were selected after eliminating studies that did not meet the inclusion criteria. Research on neurological disorders widely reported that extracts or active components of AG and AT showed therapeutic efficacy in treating neurological disorders. These components also possessed a wide array of neuroprotective effects, including reduction of pathogenic protein aggregates, antiapoptotic activity, modulation of autophagy, anti-inflammatory and antioxidant activities, regulation of neurotransmitters, activation of neurogenesis, and stimulation of neurotrophic factors. Most of the included studies were preclinical studies that used in vitro and in vivo models, and only a few clinical studies have been performed. Therefore, this review summarizes the current knowledge on AG and AT therapeutic effects as a basis for further clinical studies, and clinical trials are required before these findings can be applied to human neurological disorders.
Collapse
Affiliation(s)
- Cheol Ju Kim
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Tae Young Kwak
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Min Hyeok Bae
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea,Graduate Training Program of Korean Medical Therapeutics for Healthy Aging, Pusan National University, Yangsan, Republic of Korea,Corresponding Author Hwa Kyoung Shin, Department of Korean Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea, Tel: +82-51-510-8476, E-mail:, Byung Tae Choi, Department of Korean Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea, Tel: +82-51-510-8475, E-mail:
| | - Byung Tae Choi
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea,Graduate Training Program of Korean Medical Therapeutics for Healthy Aging, Pusan National University, Yangsan, Republic of Korea,Corresponding Author Hwa Kyoung Shin, Department of Korean Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea, Tel: +82-51-510-8476, E-mail:, Byung Tae Choi, Department of Korean Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea, Tel: +82-51-510-8475, E-mail:
| |
Collapse
|
23
|
Tan W, Qi L, Hu X, Tan Z. Research progress in traditional Chinese medicine in the treatment of Alzheimer's disease and related dementias. Front Pharmacol 2022; 13:921794. [PMID: 36506569 PMCID: PMC9729772 DOI: 10.3389/fphar.2022.921794] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is the world's leading cause of dementia and has become a huge economic burden on nations and families. However, the exact etiology of AD is still unknown, and there are no efficient medicines or methods to prevent the deterioration of cognition. Traditional Chinese medicine (TCM) has made important contributions in the battle against AD based on the characteristics of multiple targets of TCM. This study reviewed the treatment strategies and new discoveries of traditional Chinese medicine in current research, which may be beneficial to new drug researchers.
Collapse
Affiliation(s)
- Wanying Tan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lingjun Qi
- Sichuan Academy of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenghuai Tan
- Sichuan Academy of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
24
|
Tan L, Yang Y, Peng J, Zhang Y, Wu B, He B, Jia Y, Yan T. Schisandra chinensis (Turcz.) Baill. essential oil exhibits antidepressant-like effects and against brain oxidative stress through Nrf2/HO-1 pathway activation. Metab Brain Dis 2022; 37:2261-2275. [PMID: 35666395 PMCID: PMC9168360 DOI: 10.1007/s11011-022-01019-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/26/2022] [Indexed: 11/28/2022]
Abstract
The present study aimed to evaluate the antidepressant-like effect of essential oils from Schisandra chinensis (Turcz.) Baill. (SEO) and its possible mechanisms of SEO. The behavioral despair mouse model in vivo and H2O2-induced PC12 cells model in vitro were employed. And the potential effective components were identified by the spectrum-effect relationships analysis. SEO significantly decreased the immobility time in the forced swimming test and tail suspension test, which indicated a promising antidepressant-like effect of SEO in depressed mice. The decreased levels of SOD, GSH, and CAT, and increased levels of MDA were significantly reversed by SEO treatment, which showed good antioxidant activities both in vitro and in vivo. Besides, SEO significantly promoted the nuclear translocation of Nrf2 and the expression of HO-1 in depressed mice and H2O2-induced PC12 cells. The histopathological examination results showed a potential neuronal protective effect of SEO in the hippocampus and cortex. Furthermore, the upregulation of PI3K/AKT/GSK3β signaling was observed after SEO treatment in the H2O2-induced PC12 cells. Additionally, based on the spectrum-effect relationship analysis, 9 peaks were identified as positively correlated with the antioxidant activity of SEO. These results suggested that SEO promoted Nrf2/HO-1 pathway to improve the oxidative stress status and exerted the antidepressant-like effects.
Collapse
Affiliation(s)
- Liang Tan
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resource, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810001, China
| | - Yunfang Yang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Jing Peng
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Yue Zhang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Bo Wu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Bosai He
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Ying Jia
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Tingxu Yan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China.
| |
Collapse
|
25
|
Wu Q, Li X, Jiang XW, Yao D, Zhou LJ, Xu ZH, Wang N, Zhao QC, Zhang Z. Yuan-Zhi decoction in the treatment of Alzheimer’s disease: An integrated approach based on chemical profiling, network pharmacology, molecular docking and experimental evaluation. Front Pharmacol 2022; 13:893244. [PMID: 36091836 PMCID: PMC9451491 DOI: 10.3389/fphar.2022.893244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
Yuan-Zhi Decoction (YZD) is a traditional Chinese medical formulation with demonstrated clinical benefits in Alzheimer’s disease (AD). We used liquid chromatography coupled with mass spectrometry to identify 27 unique chemical components of YZD. Analyzing these using network pharmacology and molecular docking models identified 34 potential interacting molecular targets involved in 26 biochemical pathways. When tested in an animal model of AD, the APP/PS1 transgenic mice showed measurable improvements in spatial orientation and memory after the administration of YZD. These improvements coincided with significantly reduced deposition of Aβ plaques and tau protein in the hippocampi in the treated animals. In addition, a decreased BACE1 and beta-amyloid levels, a downregulation of the p-GSK-3β/GSK-3β, and an upregulation of the PI3K and p-AKT/AKT pathway was seen in YZD treated animals. These in vivo changes validated the involvement of molecular targets and pathways predicted in silico analysis of the chemical components of YZD. This study provides scientific support for the clinical use of YZD and justifies further investigations into its effects in AD. Furthermore, it demonstrates the utility of network pharmacology in elucidating the biochemical mechanisms underlying the beneficial effects of traditional Chinese medicines (TCM).
Collapse
Affiliation(s)
- Qiong Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiao-Wen Jiang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Dong Yao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Li-Jun Zhou
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Zi-Hua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Nan Wang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Qing-Chun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
- *Correspondence: Zhou Zhang, ; Qing-Chun Zhao,
| | - Zhou Zhang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- *Correspondence: Zhou Zhang, ; Qing-Chun Zhao,
| |
Collapse
|
26
|
Urati A, Dey M, Gautam AS, Singh RK. Iron-induced cellular in vitro neurotoxic responses in rat C6 cell line. ENVIRONMENTAL TOXICOLOGY 2022; 37:1968-1978. [PMID: 35446454 DOI: 10.1002/tox.23543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/03/2022] [Accepted: 04/10/2022] [Indexed: 06/14/2023]
Abstract
Iron is an essential metal critical for normal cellular and biochemical function and it is used as a cofactor in many vital biological pathways within the brain. However, accumulation of excess iron in brain is commonly associated with several neurodegenerative and neurotoxic adverse effects. Chronic exposure of iron leads to an increased risk for several neurodegenerative diseases. The exact mechanism of iron-induced neurotoxicity is still unclear. Therefore, our study aimed to investigate the mechanism of neurotoxic and neurodegenerative effects through in vitro exposure of ferrous sulphate in rat C6 cell line. The findings of our study have indicated that ferrous sulphate exposure may lead to induction of molecular markers of neuronal inflammation, apoptotic neuronal cell death, amyloid-beta and hyperphosphorylated tau levels. This study provides a basic mechanistic understanding of signaling pathway and biomarkers involved during iron-induced neurotoxicity.
Collapse
Affiliation(s)
- Anuradha Urati
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, Uttar Pradesh, India
| | - Mangaldeep Dey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, Uttar Pradesh, India
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, Uttar Pradesh, India
| |
Collapse
|
27
|
Cystathionine β-Synthase Regulates the Proliferation, Migration, and Invasion of Thyroid Carcinoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8678363. [PMID: 35795862 PMCID: PMC9252770 DOI: 10.1155/2022/8678363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/17/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022]
Abstract
Thyroid cancer is considered to be one of the most common endocrine tumors worldwide. Cystathionine β-synthase (CBS) plays a crucial role in the occurrence of several types of malignancies. And yet, the mechanism of action of CBS in the growth of thyroid carcinoma cells is still unrevealed. We found that CBS level in thyroid carcinoma tissue was higher than that in adjacent normal tissue. The overexpression of CBS enhanced the proliferation, migration, and invasion of thyroid cancer cells, while the downregulation of CBS exerted reverse effects. CBS overexpression reduced the levels of cleaved caspase-3 and cleaved poly ADP-ribose polymerase in thyroid cancer cells, whereas CBS knockdown showed reverse trends. CBS overexpression decreased reactive oxygen species (ROS) levels but increased the levels of Wnt3a and phosphorylations of phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB/AKT), mammalian target of rapamycin (mTOR), β-catenin, and glycogen synthase kinase-3 beta, while CBS knockdown exerted opposite effects. In addition, CBS overexpression promoted the growth of xenografted thyroid carcinoma, whereas CBS knockdown decreased the tumor growth by modulating angiogenesis, cell cycle, and apoptosis. Furthermore, aminooxyacetic acid (an inhibitor of CBS) dose-dependently inhibited thyroid carcinoma cell growth. CBS can regulate the proliferation, migration, and invasion of human thyroid cancer cells via ROS-mediated PI3K/AKT/mTOR and Wnt/β-catenin pathways. CBS can be a potential biomarker for diagnosing or prognosing thyroid carcinoma. Novel donors that inhibit the expression of CBS can be developed in the treatment of thyroid carcinoma.
Collapse
|
28
|
You Y, He M. Simvastatin Alleviates Vascular Cognitive Impairment Caused by Lacunar Cerebral Infarction Through Protein Kinase B/Nuclear Factor Erythroid 2–Related Factor 2 (AKT/Nrf2) Signaling Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Lacunar cerebral infarction (LACI) is one of the main causes of vascular cognitive impairment (VCI). Herein, this study explored the potential effect of Simvastatin (Sim) on VCI secondary to LACI and Akt/Nrf2 signaling transduction and apoptosis. We established a rat model of VCI and
the animals were administered with Sim (40 mg/kg and 80 mg/kg) every day for 28 days. After that, the cognition and memory abilities of rats were assessed together with analysis of morphological changes of hippocampal neurons by immunohistochemistry staining and level of anti-apoptotic related
proteins and Akt and Nrf2 signaling proteins by western blot. Compared with normal saline (control group), Sim administration significantly improved the capacity spatial learning and relieved the memory impairment with an improvement in morphological defects. Importantly, Sim treatment restored
the p-Akt, t-Nrf2, n-Nrf2 and HO-1 expression along with up-regulation of Bcl-2 and down-regulation of Bax. In conclusion, Sim improves cognitive and morphological disorders induced by LACI possibly through regulating Akt/Nrf2 signaling pathway. These evidence might promote the development
of Sim-based treatment for VCI and LACI.
Collapse
Affiliation(s)
- Yiping You
- Department of Neurology, People’s Hospital, Wuxi 214000, Jiangsu, China
| | - Min He
- Department of Nail Breast, The Second People’s Hospital, Wuxi 214000, Jiangsu, China
| |
Collapse
|
29
|
Molecular Mechanisms and Therapeutic Potential of α- and β-Asarone in the Treatment of Neurological Disorders. Antioxidants (Basel) 2022; 11:antiox11020281. [PMID: 35204164 PMCID: PMC8868500 DOI: 10.3390/antiox11020281] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 12/03/2022] Open
Abstract
Neurological disorders are important causes of morbidity and mortality around the world. The increasing prevalence of neurological disorders, associated with an aging population, has intensified the societal burden associated with these diseases, for which no effective treatment strategies currently exist. Therefore, the identification and development of novel therapeutic approaches, able to halt or reverse neuronal loss by targeting the underlying causal factors that lead to neurodegeneration and neuronal cell death, are urgently necessary. Plants and other natural products have been explored as sources of safe, naturally occurring secondary metabolites with potential neuroprotective properties. The secondary metabolites α- and β-asarone can be found in high levels in the rhizomes of the medicinal plant Acorus calamus (L.). α- and β-asarone exhibit multiple pharmacological properties including antioxidant, anti-inflammatory, antiapoptotic, anticancer, and neuroprotective effects. This paper aims to provide an overview of the current research on the therapeutic potential of α- and β-asarone in the treatment of neurological disorders, particularly neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), as well as cerebral ischemic disease, and epilepsy. Current research indicates that α- and β-asarone exert neuroprotective effects by mitigating oxidative stress, abnormal protein accumulation, neuroinflammation, neurotrophic factor deficit, and promoting neuronal cell survival, as well as activating various neuroprotective signalling pathways. Although the beneficial effects exerted by α- and β-asarone have been demonstrated through in vitro and in vivo animal studies, additional research is required to translate laboratory results into safe and effective therapies for patients with AD, PD, and other neurological and neurodegenerative diseases.
Collapse
|
30
|
Raut S, Patel R, Pervaiz I, Al-Ahmad AJ. Abeta Peptides Disrupt the Barrier Integrity and Glucose Metabolism of Human Induced Pluripotent Stem Cell-Derived Brain Microvascular Endothelial Cells. Neurotoxicology 2022; 89:110-120. [DOI: 10.1016/j.neuro.2022.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022]
|
31
|
Oxidative Stress and Beta Amyloid in Alzheimer's Disease. Which Comes First: The Chicken or the Egg? Antioxidants (Basel) 2021; 10:antiox10091479. [PMID: 34573112 PMCID: PMC8468973 DOI: 10.3390/antiox10091479] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of Alzheimer's disease involves β amyloid (Aβ) accumulation known to induce synaptic dysfunction and neurodegeneration. The brain's vulnerability to oxidative stress (OS) is considered a crucial detrimental factor in Alzheimer's disease. OS and Aβ are linked to each other because Aβ induces OS, and OS increases the Aβ deposition. Thus, the answer to the question "which comes first: the chicken or the egg?" remains extremely difficult. In any case, the evidence for the primary occurrence of oxidative stress in AD is attractive. Thus, evidence indicates that a long period of gradual oxidative damage accumulation precedes and results in the appearance of clinical and pathological AD symptoms, including Aβ deposition, neurofibrillary tangle formation, metabolic dysfunction, and cognitive decline. Moreover, oxidative stress plays a crucial role in the pathogenesis of many risk factors for AD. Alzheimer's disease begins many years before its symptoms, and antioxidant treatment can be an important therapeutic target for attacking the disease.
Collapse
|