1
|
Abdelaziz AM. Alpha-Synuclein drives NURR1 and NLRP3 Inflammasome dysregulation in Parkinson's disease: From pathogenesis to potential therapeutic strategies. Int Immunopharmacol 2025; 156:114692. [PMID: 40267723 DOI: 10.1016/j.intimp.2025.114692] [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: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD), a progressive neurodegenerative disorder, is characterized by the loss of dopaminergic neurons and pathological aggregation of α-synuclein (α-Syn). Emerging evidence highlights the interplay between genetic susceptibility, neuroinflammation, and transcriptional dysregulation in driving PD pathogenesis. This review brings together the latest information on three important players: α-Syn, the transcription factor Orphan nuclear receptor (NURR1), and the NOD-like receptor 3 (NLRP3) inflammasome. Pathogenic α-syn aggregates cause damage to neurons by disrupting mitochondria and lysosomes and spreading in a way similar to prion proteins. They also turn on the NLRP3 inflammasome, which is a key player in neuroinflammation. NLRP3-driven release of pro-inflammatory cytokines exacerbates neurodegeneration and creates a self-sustaining inflammatory milieu. Meanwhile, reduced NURR1 activity, a pivotal modulator of dopaminergic neuron survival and development, exposes neurons to oxidative stress, neuroinflammation, and α-Syn toxicity, hence exacerbating disease progression. So, targeting this trio exhibits transformative potential against PD pathogenesis.
Collapse
Affiliation(s)
- Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| |
Collapse
|
2
|
Yao J, Liu Y, Lin H, Shao C, Jin X, Peng T, Liu Y. Caffeic acid activates Nrf2 enzymes, providing protection against oxidative damage induced by ionizing radiation. Brain Res Bull 2025; 224:111325. [PMID: 40174789 DOI: 10.1016/j.brainresbull.2025.111325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/15/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Caffeic acid (CA) is a prevalent polyphenolic compound commonly found in various plant-derived foods. Due to its diverse pharmacological properties, including antioxidant activity, cardiovascular protection, and immune regulation, CA has garnered significant attention. Ionizing radiation (IR) is extensively utilized across industrial sectors, agriculture, defense applications, scientific research, and clinical medicine; however, the detrimental effects of radiation on human health cannot be ignored. IR can directly damage the DNA, proteins, and lipids within macromolecules or ionize water molecules to generate substantial quantities of free radicals that indirectly harm cells, especially those in the brain which are highly susceptible to radiation exposure. Consequently, effective strategies for preventing and treating IR-induced neurological damage represent an urgent medical challenge that necessitates resolution. Our study aims to investigate the protective effects of CA against IR-induced neuronal cell damage along with elucidating its potential mechanisms of action. The results indicate that CA can covalently modify active cysteine residues on Keap1 protein altering its conformation; this modification disrupts the interaction between Keap1 and Nrf2 while facilitating Nrf2's translocation into the nucleus where it activates downstream expression of cellular protective factors such as heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidoreductase 1 (NQO1), Thioredoxin Reductase-1 (TrxR1) and other cellular protective factors to play a role in countering radiation-induced neurological damage. In conclusion, CA emerges as an effective radioprotective agent capable of exerting antiradiation effects. Our findings provide valuable insights for developing novel therapeutic agents aimed at preventing and treating IR-induced neurological impairment.
Collapse
Affiliation(s)
- Juan Yao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| | - Yuanyuan Liu
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Huanhuan Lin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Changxin Shao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiaojie Jin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Ting Peng
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yongqi Liu
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| |
Collapse
|
3
|
Kong RY, Zhang JB, Miao X, Yao XY, Pan MH, Yin X, Yao RQ, Ren C. HDCA alleviates Parkinson's disease symptoms by promoting autophagic degradation of α-synuclein in enteric neurons. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156749. [PMID: 40252434 DOI: 10.1016/j.phymed.2025.156749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION Bile acids (BAs) are emerging as key modulators of Parkinson's disease (PD) through gut-brain interactions, yet their therapeutic potential remains underutilized. While BA imbalances contribute to PD pathogenesis, the specific subspecies regulating α-synuclein (α-syn) homeostasis and their mechanisms in enteric neurons-critical sites for PD initiation-require systematic investigation. OBJECTIVE To investigate whether hyodeoxycholic acid (HDCA), a secondary BA with documented neuroprotective properties but unproven efficacy in synucleinopathy, modulates α-syn clearance through enteric neuronal autophagy to mitigate PD progression. METHODS A53T transgenic mice underwent behavioral assessments for PD phenotyping. State-of-the-art UPLC/MS-based metabolomics quantified BA profiles. Pharmacological interventions using target-specific inhibitors (Gly-MCA, T0070907, VER-155,008) dissected the FXR-PPARγ-HSPA8 pathway. Multiscale analyses spanning immunofluorescence, western blotting, and LC3B autophagy flux reporter assays elucidated α-syn aggregation and autophagic dynamics in primary enteric neurons. RESULTS HDCA decline correlated with PD severity, positioning it as a novel biomarker for gut-brain axis dysfunction in PD. HDCA supplementation not only alleviated motor/non-motor deficits but also conferred dual neuroprotection-reducing colonic α-syn oligomers and preserving nigral dopaminergic neurons. Mechanistic decoding revealed HDCA's unparalleled capacity to activate enteric neuronal autophagy via FXR-PPARγ-HSPA8 signaling, a pathway previously unrecognized in PD therapeutics. CONCLUSION Our study reveals a novel gut-brain axis where HDCA depletion drives PD pathogenesis via FXR-PPARγ-HSPA8-mediated autophagic dysfunction in enteric neurons. PD-associated HDCA deficiency directly impairs α-syn clearance, identifying HDCA as both a gut-derived synucleinopathy biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Ren-Yu Kong
- Xuzhou Key Laboratory of Neurobiology, Department of Cell Biology and Neurobiology, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jin-Bao Zhang
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China
| | - Xu Miao
- Xuzhou Key Laboratory of Neurobiology, Department of Cell Biology and Neurobiology, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiao-Yu Yao
- Xuzhou Key Laboratory of Neurobiology, Department of Cell Biology and Neurobiology, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mei-Hua Pan
- Xuzhou Key Laboratory of Neurobiology, Department of Cell Biology and Neurobiology, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xin Yin
- Genetic Medicine Center, Xuzhou Maternal and child Health Hospital, Xuzhou, Jiangsu 221004, China
| | - Rui-Qin Yao
- Xuzhou Key Laboratory of Neurobiology, Department of Cell Biology and Neurobiology, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Chao Ren
- Department of Neurology and Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China.
| |
Collapse
|
4
|
Mu C, Shao K, Su M, Guo Y, Qiu Y, Sun R, Sun S, Sun Y, Liu C, Wang W, Qin X, Tang C. Lysophosphatidylcholine promoting α-Synuclein aggregation in Parkinson's disease: disrupting GCase glycosylation and lysosomal α-Synuclein degradation. NPJ Parkinsons Dis 2025; 11:47. [PMID: 40089519 PMCID: PMC11910603 DOI: 10.1038/s41531-025-00902-7] [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: 10/01/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
In Parkinson's Disease (PD), elevated serum lysophosphatidylcholine (LPC) levels correlate with disease progression. However, the mechanisms by which abnormal LPC elevation contributes to PD-related neurotoxicity remain poorly understood. This study aims to investigate the pathogenic role of LPC in dopaminergic neuronal damage and elucidates its underlying mechanisms. Our results showed LPC induces α-synuclein aggregation, exacerbating cognitive dysfunction. LPC activates Cleaved-Caspase3 via the orphan receptor GPR35-ERK signaling pathway, inhibits GRASP65 expression, and disrupts the polarized structure of the Golgi apparatus. This disruption impairs glycosylation and function of glucocerebrosidase (GCase), preventing its transport to lysosomes and leading to glucosylceramide (GlcCer) accumulation, a scaffold for α-synuclein aggregation. LPC also disrupts the autophagolysosomal pathway and lysosomal acidification, exacerbating toxic α-synuclein accumulation. Restoring GCase glycosylation, limiting GlcCer synthesis, or blocking ERK signaling mitigates these effects. This study highlights LPC's role in promoting α-synuclein aggregation and autophagolysosomal dysfunction, advancing our understanding of PD pathology.
Collapse
Affiliation(s)
- Chunyan Mu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kaiquan Shao
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mingyu Su
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yurong Guo
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuxiang Qiu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ruiao Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Sihan Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yaoyu Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chenkai Liu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wei Wang
- The Second School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiaoling Qin
- Department of Neurology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, China.
| | - Chuanxi Tang
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- The Research and Engineering Center of Xuzhou neurodegenerative disease diagnosis and treatment biologics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
5
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Erustes AG, Abílio VC, Bincoletto C, Piacentini M, Pereira GJS, Smaili SS. Cannabidiol induces autophagy via CB 1 receptor and reduces α-synuclein cytosolic levels. Brain Res 2025; 1850:149414. [PMID: 39710053 DOI: 10.1016/j.brainres.2024.149414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Numerous studies have explored the role of cannabinoids in neurological conditions, chronic pain and neurodegenerative diseases. Restoring autophagy has been proposed as a potential target for the treatment of neurodegenerative diseases. In our study, we used a neuroblastoma cell line that overexpresses wild-type α-synuclein to investigate the effects of cannabidiol on autophagy modulation and reduction in the level of cytosolic α-synuclein. Our results demonstrated that cannabidiol enhances the accumulation of LC3-II- and GFP-LC3-positive vesicles, which indicates an increase in autophagic flux. In addition, cannabidiol-treated cells showed a reduction in cytosolic α-synuclein levels. These effects were inhibited when the cells were treated with a CB1 receptor-selective antagonist, which indicates that the biological effects of cannabidiol are mediated via its interaction with CB1 receptor. Additionally, we also observed that cannabinoid compounds induce autophagy and α-synuclein degradation after they interact with the CB1 receptor. In summary, our data suggest that cannabidiol induces autophagy and reduces cytosolic α-synuclein levels. These biological effects are mediated preferentially through the interaction of cannabidiol with CB1 receptors, and therefore, cannabinoid compounds that act selectively on this receptor could represent a new approach for autophagy modulation and degradation of protein aggregates.
Collapse
Affiliation(s)
- Adolfo G Erustes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil.
| | - Vanessa C Abílio
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil; National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil
| | - Claudia Bincoletto
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Gustavo J S Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Soraya S Smaili
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Cheng G, Liu Z, Yan Z, Wu J, Li Z, Gao S, Zheng C, Guo S, Pan Y, Chen X, Lin G, Zhou J, Chen T. Minocycline nanoplatform penetrates the BBB and enables the targeted treatment of Parkinson's disease with cognitive impairment. J Control Release 2025; 377:591-605. [PMID: 39608455 DOI: 10.1016/j.jconrel.2024.11.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Parkinson's disease (PD)-induced motor dysfunction and cognitive impairment are becoming increasingly common due to global population aging. However, efficient treatment strategies for these conditions are still lacking. Recent studies indicated that neuroinflammation and neuronal apoptosis could greatly worsen the symptoms of PD. Therefore, anti-apoptotic and anti-inflammatory drugs could be useful in the management of PD. In the present study, minocycline (MIN)-loaded Fe3O4 nanoparticles (Fe3O4-MIN NPs) were prepared for the targeted treatment of PD. Owing to their near-infrared (NIR) irradiation-induced photothermal effects, the Fe3O4-MIN NPs could cross the blood-brain barrier (BBB), thus enhancing the delivery of Fe3O4-MIN NPs to the brain parenchyma. Subsequently, the Fe3O4-MIN NPs exerted strong anti-inflammatory effects and alleviated neuroinflammation in the brain. Furthermore, they exerted anti-oxidative effects, scavenging excessive reactive oxygen species in the brain parenchyma and thus protecting both dopaminergic and hippocampal neurons from neuroinflammation and apoptosis. Consequently, Fe3O4-MIN NPs + NIR treatment attenuated the motor dysfunction and cognitive impairment observed in PD mice. Notably, the Fe3O4-MIN NPs also showed high biocompatibility. Hence, these BBB-penetrating MIN-loaded Fe3O4 NPs demonstrate great therapeutic potential for PD accompanied by cognitive impairment.
Collapse
Affiliation(s)
- Guowang Cheng
- School of Pharmaceutical Sciences, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhiwen Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; School of Life Sciences and Biopharmaceutics, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhao Yan
- School of Pharmaceutical Sciences, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiaxin Wu
- School of Pharmaceutical Sciences, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zilin Li
- School of Pharmaceutical Sciences, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Sijia Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Chunye Zheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Shuanshuan Guo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China.
| | - Guanghui Lin
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Jianhua Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Tongkai Chen
- School of Pharmaceutical Sciences, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
8
|
Khodadadi M, Javadi B. A Review of the Protective Effects of Alkaloids against Alpha-synuclein Toxicity in Parkinson's Disease. Mini Rev Med Chem 2025; 25:112-127. [PMID: 38874050 DOI: 10.2174/0113895575306884240604065754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Alpha-synuclein (α-syn) aggregation products may cause neural injury and several neurodegenerative disorders (NDs) known as α-synucleinopathies. Alkaloids are secondary metabolites present in a variety of plant species and may positively affect human health, particularly α-synucleinopathy-associated NDs. AIM To summarize the latest scientific data on the inhibitory properties of alkaloids in α- synucleinopathies, especially in Parkinson's disease. METHODS Literature search was performed using web-based databases including Web of Science, PubMed, and Scopus up to January 2024, in the English language. RESULTS Harmala alkaloids, caffein, lycorine, piperin, acetylcorynoline, berberin, papaverine, squalamine, trodusquemine and nicotin have been found to be the most active natural alkaloids against synucleinopathy. The underlying mechanisms that contribute to this effect would be the inhibition of α-syn aggregation; elimination of formed aggregates; improvement in autophagy activation; promotion of the activity and expression of antioxidative enzymes; and prevention of oxidative injury and apoptosis in dopaminergic neurons. CONCLUSION The findings of the present study highlight the inhibitory activities of alkaloids against synucleinopathy. However, no clinical data supports the reported activities in humans, which calls attention to the need for conducting clinical trials to elucidate the efficacy, safety, proper dosage, unwanted effects and pharmacokinetics aspects of alkaloids in humans.
Collapse
Affiliation(s)
- Mahdi Khodadadi
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behjat Javadi
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Xu J, Dai P, Zhang C, Dong N, Li C, Tang C, Jin Z, Lin S, Ye L, Sun T, Jin Y, Wu F, Luo L, Wu P, Li S, Li X, Hsu S, Jiang D, Wang Z. Injectable Hierarchical Bioactive Hydrogels with Fibroblast Growth Factor 21/Edaravone/Caffeic Acid Asynchronous Delivery for Treating Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412020. [PMID: 39630931 PMCID: PMC11775539 DOI: 10.1002/advs.202412020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Parkinson's disease (PD) is one of the most common long-term neurodegenerative disorders, with multiple comorbid psychiatric and behavioral abnormalities. The combination of clinical drugs targeting different symptoms with smart hydrogels to achieve asynchronous releases is highly translational and challenging. Here, a hierarchical bioactive hydrogel (OACDP) is designed with asynchronous release based on PD pathology. The hydrogel with caffeic acid-grafted polymer main chain is crosslinked using a micellar nanocrosslinker, with sufficient modulus (≈167 Pa), antioxidant activity (> 50%), injectability (30-gauge syringe needle), and shape-adaptability. Each of the three drugs (caffeic acid, fibroblast growth factor 21, and Edaravone) is separately engaged in different micro- or nanostructures of the hydrogel and released with asynchronous kinetics of first-order release, zero-order release, or matching Korsmeyer-Peppas model. The triple-loaded hydrogel is injected into the brains of PD rats, showing behavioral improvement. Histological analysis revealed that the triple-loaded OACDP hydrogels are effective in achieving immediate neuroprotection, i.e., reduction the loss of tyrosine hydroxylase in substantia nigra compacta and striatum (retained ≈10-fold versus control), decreasing oxidative stress, reducing astrocyte and microglia activation, and stimulating the AMPK/PGC-1α axis to regulate the mitochondrial function, providing a multi-dimensional PD therapy. The asynchronous release of OACDP hydrogel provides a new conception for PD treatment and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Junpeng Xu
- Affiliated Cixi HospitalWenzhou Medical UniversityNingboZhejiang315300China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Peng Dai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Chen Zhang
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhouZhejiang324025China
| | - Na Dong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Caiyan Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Chonghui Tang
- Affiliated Cixi HospitalWenzhou Medical UniversityNingboZhejiang315300China
| | - Zhihao Jin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Shih‐Ho Lin
- Institute of Polymer Science and EngineeringNational Taiwan UniversityTaipeiTaiwan106319Republic of China
| | - Luyang Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Tianmiao Sun
- Affiliated Cixi HospitalWenzhou Medical UniversityNingboZhejiang315300China
| | - Yukai Jin
- Affiliated Cixi HospitalWenzhou Medical UniversityNingboZhejiang315300China
| | - Fenzan Wu
- Affiliated Cixi HospitalWenzhou Medical UniversityNingboZhejiang315300China
| | - Lihua Luo
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhouZhejiang324025China
| | - Ping Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Shengcun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- Rehabilitation Medicine CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiang325000China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Shan‐hui Hsu
- Institute of Polymer Science and EngineeringNational Taiwan UniversityTaipeiTaiwan106319Republic of China
- Institute of Cellular and System MedicineNational Health Research InstitutesMiaoliTaiwan350401Republic of China
| | - Dawei Jiang
- Affiliated Cixi HospitalWenzhou Medical UniversityNingboZhejiang315300China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Zhouguang Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
- State Key Laboratory of Macromolecular Drugs and Large‐scale PreparationSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhouZhejiang325000China
| |
Collapse
|
10
|
Jiao F, Meng L, Du K, Li X. The autophagy-lysosome pathway: a potential target in the chemical and gene therapeutic strategies for Parkinson's disease. Neural Regen Res 2025; 20:139-158. [PMID: 38767483 PMCID: PMC11246151 DOI: 10.4103/nrr.nrr-d-23-01195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 05/22/2024] Open
Abstract
Parkinson's disease is a common neurodegenerative disease with movement disorders associated with the intracytoplasmic deposition of aggregate proteins such as α-synuclein in neurons. As one of the major intracellular degradation pathways, the autophagy-lysosome pathway plays an important role in eliminating these proteins. Accumulating evidence has shown that upregulation of the autophagy-lysosome pathway may contribute to the clearance of α-synuclein aggregates and protect against degeneration of dopaminergic neurons in Parkinson's disease. Moreover, multiple genes associated with the pathogenesis of Parkinson's disease are intimately linked to alterations in the autophagy-lysosome pathway. Thus, this pathway appears to be a promising therapeutic target for treatment of Parkinson's disease. In this review, we briefly introduce the machinery of autophagy. Then, we provide a description of the effects of Parkinson's disease-related genes on the autophagy-lysosome pathway. Finally, we highlight the potential chemical and genetic therapeutic strategies targeting the autophagy-lysosome pathway and their applications in Parkinson's disease.
Collapse
Affiliation(s)
- Fengjuan Jiao
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong Province, China
| | - Lingyan Meng
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
| | - Kang Du
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
| | - Xuezhi Li
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong Province, China
| |
Collapse
|
11
|
Gao L, Yang XN, Dong YX, Han YJ, Zhang XY, Zhou XL, Liu Y, Liu F, Fang JS, Ji JL, Gao ZR, Qin XM. The potential therapeutic strategy in combating neurodegenerative diseases: Focusing on natural products. Pharmacol Ther 2024; 264:108751. [PMID: 39522697 DOI: 10.1016/j.pharmthera.2024.108751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Huntington disease (HD), and Multiple sclerosis (MS), pose a significant global health challenge due to their intricate pathology and limited therapeutic interventions. Natural products represent invaluable reservoirs for combating these neurodegenerative diseases by targeting key pathological hallmarks such as protein aggregation, synaptic dysfunction, aberrant proteostasis, cytoskeletal abnormalities, altered energy homeostasis, inflammation, and neuronal cell death. This review provides an in-depth analysis of the mechanisms and therapeutic targets of natural products for their neuroprotective effects. Furthermore, it elucidates the current progress of clinical trials investigating the potential of natural products in delaying neurodegeneration. The objective of this review is to enhance the comprehension of natural products in the prevention and treatment of neurodegenerative diseases, offering new insights and potential avenues for future pharmaceutical research.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xi-Na Yang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Yi-Xiao Dong
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Yi-Jia Han
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xin-Yue Zhang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xin-Le Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Ying Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Fang Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Jian-Song Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jian-Long Ji
- College of Integrated Circuits, Taiyuan University of Technology, Taiyuan, China.
| | - Zheng-Run Gao
- Songjiang Research Institute, Songjiang Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China.
| |
Collapse
|
12
|
Cao T, Wang XL, Rao JY, Zhu HF, Qi HY, Tian Z. Periplaneta americana L. extract exerts neuroprotective effects by inhibiting endoplasmic reticulum stress via AKT-dependent pathway in experimental models of Parkinson's disease. Chin Med 2024; 19:157. [PMID: 39538357 PMCID: PMC11562093 DOI: 10.1186/s13020-024-01029-2] [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: 06/29/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a chronic neurodegenerative disorder that currently has no curable strategies. More and more evidence suggests that endoplasmic reticulum (ER) stress plays an essential role in PD pathogenesis. Periplaneta americana L. (P. americana) is a traditional Chinese medicine with diverse therapeutic properties. This study aims to investigate the neuroprotective effect and underlying mechanism of P. americana in in vitro and in vivo PD models. METHODS The exposure of SH-SY5Y cells to 1-methyl-4-phenyl-pyridinium (MPP+) was used as the in vitro PD model. MTT assay, Hoechst staining, Calcein AM-PI staining and flow cytometry were performed to measure the cell viability and apoptosis. DCFH-DA and JC-1 assay were used to measure the intracellular ROS and mitochondrial membrane potential (Δψm), respectively. Western-blot and immunostaining were conducted to detect the expression of key molecules related with ER stress. For the in vivo PD model induced by 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydro-pyridine (MPTP), the motor function of mice was assessed by behavioral tests, the level of TH was examined by western-blot and immunostaining, the expression of key molecules related with ER stress was measured by western-blot. RESULTS Periplaneta americana ethanol extract (PAE) concentration-dependently inhibited MPP+-induced cell loss and increased cell viability. PAE also remarkably attenuated ROS accumulation, the decline of Δψm as well as the excessive ER stress. The neuroprotective effects of PAE could be blocked by ROS inducer trimethylamine N-Oxide or ER stress activator tunicaymycin, while the antioxidant N-Acetyl-L-cysteine or ER stress inhibitor sodium 4-phenylbutyrate mimicked the effects of PAE. Furthermore, we found that PAE could activate AKT/GSK3β/β-catenin pathway. The effect of PAE on ROS production, Δψm and ER stress was blocked by AKT inhibitor MK-2206. In in vivo model, PAE significantly improved motor function, prevented dopaminergic neuronal loss and attenuated ER stress in substantia nigra and striatum of MPTP-treated mice. Similarly, the effects of PAE on MPTP-treated mice were also abolished by MK-2206. CONCLUSIONS Our results suggest that P. americana exerts neuroprotective effects through inhibiting ER stress via AKT-dependent pathway. Periplaneta americana may represent a promising therapeutic agent for PD treatment and is worthy of further being exploited.
Collapse
Affiliation(s)
- Ting Cao
- College of Pharmaceutical Sciences, Southwest University, No.1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Xue-Lian Wang
- College of Pharmaceutical Sciences, Southwest University, No.1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Jiang-Yan Rao
- College of Pharmaceutical Sciences, Southwest University, No.1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Hui-Feng Zhu
- College of Pharmaceutical Sciences, Southwest University, No.1 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Hong-Yi Qi
- College of Pharmaceutical Sciences, Southwest University, No.1 Tiansheng Road, Beibei District, Chongqing, 400715, China.
| | - Zhen Tian
- College of Pharmaceutical Sciences, Southwest University, No.1 Tiansheng Road, Beibei District, Chongqing, 400715, China.
| |
Collapse
|
13
|
Ahanger IA, Dar TA. Small molecule modulators of alpha-synuclein aggregation and toxicity: Pioneering an emerging arsenal against Parkinson's disease. Ageing Res Rev 2024; 101:102538. [PMID: 39389237 DOI: 10.1016/j.arr.2024.102538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Parkinson's disease (PD) is primarily characterized by loss of dopaminergic neurons in the substantia nigra pars compacta region of the brain and accumulation of aggregated forms of alpha-synuclein (α-Syn), an intrinsically disordered protein, in the form of Lewy Bodies and Lewy Neurites. Substantial evidences point to the aggregated/fibrillar forms of α-Syn as a central event in PD pathogenesis, underscoring the modulation of α-Syn aggregation as a promising strategy for PD treatment. Consequently, numerous anti-aggregation agents, spanning from small molecules to polymers, have been scrutinized for their potential to mitigate α-Syn aggregation and its associated toxicity. Among these, small molecule modulators like osmoprotectants, polyphenols, cellular metabolites, metals, and peptides have emerged as promising candidates with significant potential in PD management. This article offers a comprehensive overview of the effects of these small molecule modulators on the aggregation propensity and associated toxicity of α-Syn and its PD-associated mutants. It serves as a valuable resource for identifying and developing potent, non-invasive, non-toxic, and highly specific small molecule-based therapeutic arsenal for combating PD. Additionally, it raises pertinent questions aimed at guiding future research endeavours in the field of α-Syn aggregation remodelling.
Collapse
Affiliation(s)
- Ishfaq Ahmad Ahanger
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| |
Collapse
|
14
|
Rozhkova IN, Okotrub SV, Brusentsev EY, Rakhmanova TA, Lebedeva DA, Kozeneva VS, Shavshaeva NA, Khotskin NV, Amstislavsky SY. Substantia nigra alterations in mice modeling Parkinson's disease. Vavilovskii Zhurnal Genet Selektsii 2024; 28:744-751. [PMID: 39722665 PMCID: PMC11668818 DOI: 10.18699/vjgb-24-82] [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: 02/12/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 12/28/2024] Open
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative pathology of the central nervous system. The well-known abnormalities characteristic of PD are dysfunctions in the nigrostriatal system including the substantia nigra of the midbrain and the striatum. Moreover, in PD persons, alpha-synucleinopathy is associated with abnormalities in the dopaminergic brain system. To study the mechanisms of this pathology, genetic models in mice have been designed. Transgenic mice of the B6.Cg-Tg(Prnp-SNCA*A53T)23Mkle/J strain (referred to as B6.Cg-Tg further in the text) possess the A53T mutation in the human alpha-synuclein SNCA gene. The density of neurons in the prefrontal cortex, hippocampus, substantia nigra and striatum in B6.Cg-Tg mice was assessed in our previous work, but the dopaminergic system was not studied there, although it plays a key role in the development of PD. The aim of the current study was to investigate motor coordination and body balance, as well as dopaminergic neuronal density and alpha-synuclein accumulation in the substantia nigra in male B6.Cg-Tg mice at the age of six months. Wild-type mice of the same sex and age, siblings of the B6.Cg-Tg mice from the same litters, lacking the SNCA gene with the A53T mutation, but expressing murine alpha-synuclein, were used as controls (referred to as the wild type further in the text). Motor coordination and body balance were assessed with the rota-rod test; the density of dopaminergic neurons and accumulation of alpha-synuclein in the substantia nigra were evaluated by the immunohistochemical method. There was no difference between B6.Cg-Tg mice and WT siblings in motor coordination and body balance. However, accumulation of alpha-synuclein and a decrease in the number of dopaminergic neurons in the substantia nigra were found in the B6.Cg-Tg mouse strain. Thus, the mice of the B6.Cg-Tg strain at the age of six months have some symptoms of the onset of PD, such as the accumulation of mutant alpha-synuclein and a decrease in the number of dopaminergic neurons in the substantia nigra. Taken together, the results obtained in our work qualify the B6.Cg-Tg strain as a pertinent model for studying the early stage of human PD already at the age of six months.
Collapse
Affiliation(s)
- I N Rozhkova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S V Okotrub
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E Yu Brusentsev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - T A Rakhmanova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - D A Lebedeva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - V S Kozeneva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - N A Shavshaeva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - N V Khotskin
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S Ya Amstislavsky
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
15
|
Pedrão LFAT, Medeiros POS, Leandro EC, Falquetto B. Parkinson's disease models and death signaling: what do we know until now? Front Neuroanat 2024; 18:1419108. [PMID: 39533977 PMCID: PMC11555652 DOI: 10.3389/fnana.2024.1419108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/04/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson's disease (PD) is the second neurodegenerative disorder most prevalent in the world, characterized by the loss of dopaminergic neurons in the Substantia Nigra (SN). It is well known for its motor and non-motor symptoms including bradykinesia, resting tremor, psychiatric, cardiorespiratory, and other dysfunctions. Pathological apoptosis contributes to a wide variety of diseases including PD. Various insults and/or cellular phenotypes have been shown to trigger distinct signaling events leading to cell death in neurons affected by PD. The intrinsic or mitochondrial pathway, inflammatory or oxidative stress-induced extrinsic pathways are the main events associated with apoptosis in PD-related neuronal loss. Although SN is the main brain area studied so far, other brain nuclei are also affected by the disease leading to non-classical motor symptoms as well as non-motor symptoms. Among these, the respiratory symptoms are often overlooked, yet they can cause discomfort and may contribute to patients shortened lifespan after disease diagnosis. While animal and in vitro models are frequently used to investigate the mechanisms involved in the pathogenesis of PD in both the SN and other brain regions, these models provide only a limited understanding of the disease's actual progression. This review offers a comprehensive overview of some of the most studied forms of cell death, including recent research on potential treatment targets for these pathways. It highlights key findings and milestones in the field, shedding light on the potential role of understanding cell death in the prevention and treatment of the PD. Therefore, unraveling the connection between these pathways and the notable pathological mechanisms observed during PD progression could enhance our comprehension of the disease's origin and provide valuable insights into potential molecular targets for the developing therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Barbara Falquetto
- Department of Pharmacology, Instituto de Ciências Biomédica, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
16
|
Coelho VS, de Moura DG, Aguiar LL, Ribeiro LV, Silva VDM, da Veiga Correia VT, Melo AC, Silva MR, de Paula ACCFF, de Araújo RLB, Melo JOF. The Profile of Phenolic Compounds Identified in Pitaya Fruits, Health Effects, and Food Applications: An Integrative Review. PLANTS (BASEL, SWITZERLAND) 2024; 13:3020. [PMID: 39519939 PMCID: PMC11548494 DOI: 10.3390/plants13213020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE This integrative review aimed to identify the phenolic compounds present in pitayas (dragon fruit). METHODS We employed a comprehensive search strategy, encompassing full-text articles published between 2013 and 2023 in Portuguese, English, and Spanish from databases indexed in ScienceDirect, Capes Periodics, Scielo, and PubMed. The study's selection was guided by the question, "What are the main phenolic compounds found in pitaya fruits?". RESULTS After screening 601 papers, 57 met the inclusion criteria. The identified phytochemicals have been associated with a range of health benefits, including antioxidant, anti-inflammatory, and anxiolytic properties. Additionally, they exhibit promising applications in the management of cancer, diabetes, and obesity. These 57 studies encompassed various genera, including Hylocereus, Selenicereus, and Stenocereus. Notably, Hylocereus undatus and Hylocereus polyrhizus emerged as the most extensively characterized species regarding polyphenol content. Analysis revealed that flavonoids, particularly kaempferol and rutin, were the predominant phenolic class within the pulp and peel of these fruits. Additionally, hydroxycinnamic and benzoic acid derivatives, especially chlorogenic acid, caffeic, protocatechuic, synaptic, and ellagic acid, were frequently reported. Furthermore, betalains, specifically betacyanins, were identified, contributing to the characteristic purplish-red color of the pitaya peel and pulp. These betalains hold significant potential as natural colorants in the food industry. CONCLUSION Therefore, the different pitayas have promising sources for the extraction of pigments for incorporation in the food industry. We recommend further studies investigate their potential as nutraceuticals.
Collapse
Affiliation(s)
- Vinicius Serafim Coelho
- Departamento de Alimentos, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (V.S.C.); (D.G.d.M.); (L.L.A.); (V.T.d.V.C.); (R.L.B.d.A.)
| | - Daniela Gomes de Moura
- Departamento de Alimentos, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (V.S.C.); (D.G.d.M.); (L.L.A.); (V.T.d.V.C.); (R.L.B.d.A.)
| | - Lara Louzada Aguiar
- Departamento de Alimentos, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (V.S.C.); (D.G.d.M.); (L.L.A.); (V.T.d.V.C.); (R.L.B.d.A.)
| | - Lucas Victor Ribeiro
- Departamento de Ciências Exatas e Biológicas, Campus Sete Lagoas, Universidade Federal de São João del-Rei, Sete Lagoas 36307-352, MG, Brazil; (L.V.R.); (V.D.M.S.)
| | - Viviane Dias Medeiros Silva
- Departamento de Ciências Exatas e Biológicas, Campus Sete Lagoas, Universidade Federal de São João del-Rei, Sete Lagoas 36307-352, MG, Brazil; (L.V.R.); (V.D.M.S.)
| | - Vinícius Tadeu da Veiga Correia
- Departamento de Alimentos, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (V.S.C.); (D.G.d.M.); (L.L.A.); (V.T.d.V.C.); (R.L.B.d.A.)
| | - Angelita Cristine Melo
- Curso de Farmácia, Campus Centro-Oeste, Universidade Federal de São João del-Rei, Divinópolis 35501-296, MG, Brazil;
| | - Mauro Ramalho Silva
- Departamento de Bioquímica e Imunologia, Campus Pampulha, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil;
| | | | - Raquel Linhares Bello de Araújo
- Departamento de Alimentos, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (V.S.C.); (D.G.d.M.); (L.L.A.); (V.T.d.V.C.); (R.L.B.d.A.)
| | - Julio Onesio Ferreira Melo
- Departamento de Ciências Exatas e Biológicas, Campus Sete Lagoas, Universidade Federal de São João del-Rei, Sete Lagoas 36307-352, MG, Brazil; (L.V.R.); (V.D.M.S.)
| |
Collapse
|
17
|
Hu Y, Cui J, Sun J, Liu X, Gao S, Mei X, Wu C, Tian H. A novel biomimetic nanovesicle containing caffeic acid-coupled carbon quantum dots for the the treatment of Alzheimer's disease via nasal administration. J Nanobiotechnology 2024; 22:642. [PMID: 39425199 PMCID: PMC11490022 DOI: 10.1186/s12951-024-02912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by progressive cognitive and physical impairment. Neuroinflammation is related to AD, and the misfolding and aggregation of amyloid protein in the brain creates an inflammatory microenvironment. Microglia are the predominant contributors to neuroinflammation, and abnormal activation of microglia induces the release of a large amount of inflammatory factors, promotes neuronal apoptosis, and leads to cognitive impairment. In this study, we used microglial membranes containing caffeic acid-coupled carbon quantum dots to prepare a novel biomimetic nanocapsule (CDs-CA-MGs) for the treatment of AD. The application of CDs-CA-MGs via nasal administration can bypass the blood‒brain barrier (BBB) and directly target the site of inflammation. After treatment with CDs-CA-MGs, AD mice showed reduced inflammation in the brain, decreased neuronal apoptosis, and significantly improved learning and memory abilities. In addition, CDs-CA-MGs affect inflammation-related JAK-STAT and Toll-like receptor signaling pathways in AD mice. CDs-CA-MGs significantly downregulated interleukins (IL-1β and IL-6) and tumor necrosis factor (TNF-α). This finding suggested that CDs-CA-MGs may improve cognitive impairment by modulating inflammatory responses. In conclusion, the use of CDs-CA-MGs provides a possible therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jingwen Cui
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Junpeng Sun
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xiaobang Liu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Shuang Gao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xifan Mei
- Liaoning Vocational College of Medicine, Shenyang, Liaoning, 110101, China.
| | - Chao Wu
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| | - He Tian
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
18
|
Gao Q, Li X, Huang T, Gao L, Wang S, Deng Y, Wang F, Xue X, Duan R. Angiotensin-(1-7) relieves behavioral defects and α-synuclein expression through NEAT1/miR-153-3p axis in Parkinson's disease. Aging (Albany NY) 2024; 16:13304-13322. [PMID: 39422618 PMCID: PMC11719108 DOI: 10.18632/aging.206028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/05/2024] [Indexed: 10/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, whose characteristic pathology involves progressive deficiency of dopaminergic neurons and generation of Lewy bodies (LBs). Aggregated and misfolded α-synuclein (α-syn) is the major constituent of LBs. As the newly discovered pathway of renin-angiotensin system (RAS), Angiotensin-(1-7) (Ang-(1-7)) and receptor Mas have attracted increasing attentions for their correlation with PD, but underlying mechanisms remain not fully clear. Based on above, this study established PD models of mice and primary dopaminergic neurons with AAV-hα-syn(A53T), then discussed the effects of Ang-(1-7)/Mas on α-syn level and neuronal apoptosis for these models combined with downstream long non-coding RNA (lncRNA) and microRNA (miRNA). Results showed that Ang-(1-7) alleviated behavioral impairments, rescued dopaminergic neurons loss and lowered α-syn expression in substantia nigra of hα-syn(A53T) overexpressed PD mice. We also discovered that Ang-(1-7) decreased level of α-syn and apoptosis in the hα-syn(A53T) overexpressed dopaminergic neurons through lncRNA NEAT1/miR-153-3p axis. Moreover, miR-153-3p level in peripheral blood is found negatively correlated with that of α-syn. In conclusion, our work not only showed neuroprotective effect and underlying mechanisms for Ang-(1-7) on α-syn in vivo and vitro, but also brought new hope on miR-153-3p and NEAT1 for diagnosis and treatment in PD.
Collapse
Affiliation(s)
- Qing Gao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| | - Xiaoyuan Li
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| | - Ting Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| | - Li Gao
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Siyu Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| | - Yang Deng
- Department of Neurology, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210006, Jiangsu, P.R. China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| | - Xue Xue
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, P.R. China
| |
Collapse
|
19
|
Gahtani RM, Shoaib S, Hani U, Jayachithra R, Alomary MN, Chauhan W, Jahan R, Tufail S, Ansari MA. Combating Parkinson's disease with plant-derived polyphenols: Targeting oxidative stress and neuroinflammation. Neurochem Int 2024; 178:105798. [PMID: 38950626 DOI: 10.1016/j.neuint.2024.105798] [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: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
Parkinson's disease (PD) is a devastating neurodegenerative disorder predominantly affecting the elderly, characterized by the loss of dopaminergic neurons in the substantia nigra. Reactive oxygen species (ROS) generation plays a central role in the pathogenesis of PD and other neurodegenerative diseases. An imbalance between cellular antioxidant activity and ROS production leads to oxidative stress, contributing to disease progression. Dopamine metabolism, mitochondrial dysfunction, and neuroinflammation in dopaminergic neurons have been implicated in the pathogenesis of Parkinson's disease. Consequently, there is a pressing need for therapeutic interventions capable of scavenging ROS. Current pharmacological approaches, such as L-dihydroxyphenylalanine (levodopa or L-DOPA) and other drugs, provide symptomatic relief but are limited by severe side effects. Researchers worldwide have been exploring alternative compounds with less toxicity to address the multifaceted challenges associated with Parkinson's disease. In recent years, plant-derived polyphenolic compounds have gained significant attention as potential therapeutic agents. These compounds exhibit neuroprotective effects by targeting pathophysiological responses, including oxidative stress and neuroinflammation, in Parkinson's disease. The objective of this review is to summarize the current understanding of the neuroprotective effects of various polyphenols in Parkinson's disease, focusing on their antioxidant and anti-inflammatory properties, and to discuss their potential as therapeutic candidates. This review highlights the progress made in elucidating the molecular mechanisms of action of these polyphenols, identifying potential therapeutic targets, and optimizing their delivery and bioavailability. Well-designed clinical trials are necessary to establish the efficacy and safety of polyphenol-based interventions in the management of Parkinson's disease.
Collapse
Affiliation(s)
- Reem M Gahtani
- Department of clinical Laboratory Sciences, King Khalid University, Abha, Saudi Arabia
| | - Shoaib Shoaib
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, AL, 35205, USA.
| | - Umme Hani
- Department of pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - R Jayachithra
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia
| | - Waseem Chauhan
- Division of Hematology, Duke Comprehensive Sickle Cell Center, Department of Medicine, Duke University School of Medicine, Research Drive, Durham, NC, 27710, USA
| | - Roshan Jahan
- Department of Botany, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Saba Tufail
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, AL, 35205, USA
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia.
| |
Collapse
|
20
|
Lopes LE, da Silva Barroso S, Caldas JK, Vasconcelos PR, Canuto KM, Dariva C, Santos KS, Severino P, Cardoso JC, Souto EB, Gomes MZ. Neuroprotective effects of Tradescantia spathacea tea bioactives in Parkinson's disease: In vivo proof-of-concept. J Tradit Complement Med 2024; 14:435-445. [PMID: 39035688 PMCID: PMC11259708 DOI: 10.1016/j.jtcme.2024.01.003] [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: 03/15/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 07/23/2024] Open
Abstract
Background and aim Tradescantia spathacea (T. spathacea) is a traditional medicinal plant from Central America and its tea, obtained by infusion, has been recognized as a functional food. The aim of this work was to investigate the effects of dry tea containing biocompounds from T. spathacea tea on motor and emotional behavior, as well as tyrosine hydroxylase (TH) and glial fibrillary acidic protein (GFAP) expression in 6-hydroxydopamine (6-OHDA)-lesioned rats. Experimental procedure Bioactives were identified by Ultra Performance Liquid Chromatography (UPLC) and an in vivo study in male Wistar rats was run as proof of concept of neuroprotective effects of DTTS. Results and conclusion We found 15 biocompounds that had not been previously reported in T. spathacea: the UPLC-QTOF-MS/MS allowed identification five phenolic acids, one coumarin, two flavonoids, one iridoid, one phenylpropanoid glycoside, and six fatty acid derivatives. The dry tea of T. spathacea (DTTS) presented significant antioxidant activity and high contents of phenolic compounds and flavonoids. Doses of 10, 30, and 100 mg/kg of DTTS were protective against dopaminergic neurodegeneration and exhibited modulatory action on the astrocyte-mediated neuroinflammatory response. Behavioral tests showed that 30 mg/kg of DTTS counteracted motor impairment, while 100 mg/kg produced an anxiolytic effect. The DTTS could be, therefore, a promising strategy for the management of Parkinson's disease.
Collapse
Affiliation(s)
- Lorenna E.S. Lopes
- Tiradentes University (UNIT), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| | | | - Joanny K.M. Caldas
- Research and Technology Institute (ITP), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| | - Paulo R. Vasconcelos
- Embrapa Agroindústria Tropical, Rua Dra. Sara Mesquita, 2.270, Bairro Planalto do Pici, Fortaleza, CEP 60511-110, Ceará, Brazil
| | - Kirley M. Canuto
- Embrapa Agroindústria Tropical, Rua Dra. Sara Mesquita, 2.270, Bairro Planalto do Pici, Fortaleza, CEP 60511-110, Ceará, Brazil
| | - Claudio Dariva
- Tiradentes University (UNIT), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
- Research and Technology Institute (ITP), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| | - Klebson S. Santos
- Research and Technology Institute (ITP), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| | - Patricia Severino
- Tiradentes University (UNIT), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
- Research and Technology Institute (ITP), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| | - Juliana C. Cardoso
- Tiradentes University (UNIT), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
- Research and Technology Institute (ITP), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| | - Eliana B. Souto
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Porto, 4050-313, Portugal
- UCIBIO – Applied Molecular Biosciences Unit, MEDTECH, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, 4050-313, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, 4050-313, Portugal
| | - Margarete Z. Gomes
- Tiradentes University (UNIT), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
- Research and Technology Institute (ITP), Av. Murilo Dantas, 300, Aracaju, CEP 49032-490, Sergipe, Brazil
| |
Collapse
|
21
|
Ren Y, Wu X, Bai T, Yang N, Yuan Y, Xu L, Wen Y, Wen Y, Wang Z, Zhou L, Zou F, Li W. CDK5-USP30 signaling pathway regulates MAVS-mediated inflammation via suppressing mitophagy in MPTP/MPP + PD model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116446. [PMID: 38772138 DOI: 10.1016/j.ecoenv.2024.116446] [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: 12/14/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
The discovery of MPTP, an industrial chemical and contaminant of illicit narcotics, which causes parkinsonism in humans, non-human primates and rodents, has led to environmental pollutants exposure being convicted as key candidate in Parkinson's disease (PD) pathogenesis. Though MPTP-induced mitochondrial dysfunction and neuroinflammation are mainly responsible for the causative issue of MPTP neurotoxicity, the underlying mechanism involved remains unclear. Here, we reveal a novel signaling mechanism of CDK5-USP30-MAVS regulating MPTP/MPP+ induced PD. MPP+ (the toxic metabolite of MPTP) treatment not only led to the increased protein levels of USP30 but also to mitophagy inhibition, mitochondrial dysfunction, and MAVS-mediated inflammation in BV2 microglial cells. Both mitophagy stimulation (Urolithin A administration) and USP30 knockdown relieved MAVS-mediated inflammation via restoring mitophagy and mitochondrial function in MPP+-induced cell model. Notably, MPTP/MPP+-induced CDK5 activation regulated USP30 phosphorylation at serine 216 to stabilize USP30. Moreover, CDK5-USP30 pathway promoted MAVS-mediated inflammation in MPTP/MPP+-induced PD model. Inhibition of CDK5 not only had a protective effect on MPP+-induced cell model of PD via suppressing the upregulation of USP30 and the activation of MAVS inflammation pathway in vitro, but also prevented neurodegeneration in vivo and alleviated movement impairment in MPTP mouse model of PD. Overall, our study reveal that CDK5 blocks mitophagy through phosphorylating USP30 and activates MAVS inflammation pathway in MPTP/MPP+-induced PD model, which suggests that CDK5-USP30-MAVS signaling pathway represents a valuable treatment strategy for PD induced by environmental neurotoxic pollutants in relation to MPTP.
Collapse
Affiliation(s)
- Yixian Ren
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Occupational Environment and Health, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou, China
| | - Xian Wu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Tianyao Bai
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Nanfei Yang
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuyu Yuan
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lingling Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yue Wen
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ying Wen
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhi Wang
- Key Laboratory of Occupational Environment and Health, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou, China
| | - Liping Zhou
- Key Laboratory of Occupational Environment and Health, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Wenjun Li
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
22
|
Hou L, Ma J, Feng X, Chen J, Dong BH, Xiao L, Zhang X, Guo B. Caffeic acid and diabetic neuropathy: Investigating protective effects and insulin-like growth factor 1 (IGF-1)-related antioxidative and anti-inflammatory mechanisms in mice. Heliyon 2024; 10:e32623. [PMID: 38975173 PMCID: PMC11225750 DOI: 10.1016/j.heliyon.2024.e32623] [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: 10/09/2023] [Revised: 05/15/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Diabetic neuropathy (DN) represents a common and debilitating complication of diabetes, affecting a significant proportion of patients. Despite available treatments focusing on symptom management, there remains an unmet need for therapies that address the underlying pathophysiology. In pursuit of novel interventions, this study evaluated the therapeutic effects of caffeic acid-a natural phenolic compound prevalent in various foods-on diabetic neuropathy using a mouse model, particularly examining its interaction with the Insulin-like Growth Factor 1 (IGF-1) signaling pathway. Caffeic acid was administered orally at two dosages (5 mg/kg and 10 mg/kg), and a comprehensive set of outcomes including fasting blood glucose levels, body weight, sensory behavior, spinal cord oxidative stress markers, inflammatory cytokines, and components of the IGF-1 signaling cascade were assessed. Additionally, to determine the specific contribution of IGF-1 signaling to the observed benefits, IGF1R inhibitor Picropodophyllin (PPP) was co-administered with caffeic acid. Our results demonstrated that caffeic acid, at both dosages, effectively reduced hyperglycemia and alleviated sensory behavioral deficits in diabetic mice. This was accompanied by a marked decrease in oxidative stress markers and an increase in antioxidant enzyme activities within the spinal cord. Significantly lowered microglial activation and inflammatory cytokine expression highlighted the potent antioxidative and anti-inflammatory effects of caffeic acid. Moreover, increases in both serum and spinal levels of IGF-1, along with elevated phosphorylated IGF1R, implicated the IGF-1 signaling pathway as a mediator of caffeic acid's neuroprotective actions. The partial reversal of caffeic acid's benefits by PPP substantiated the pivotal engagement of IGF-1 signaling in mediating its effects. Our findings delineate the capability of caffeic acid to mitigate DN symptoms, particularly through reducing spinal oxidative stress and inflammation, and pinpoint the integral role of IGF-1 signaling in these protective mechanisms. The insights gleaned from this study not only position caffeic acid as a promising dietary adjunct for managing diabetic neuropathy but also highlight the therapeutic potential of targeting spinal IGF-1 signaling as part of a strategic treatment approach.
Collapse
Affiliation(s)
- Leina Hou
- Department of Anesthesiology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Jiaqi Ma
- Department of Radiology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Xugang Feng
- Department of General Surgery, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Jing Chen
- Department of Medical Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Bu-huai Dong
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| | - Li Xiao
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| | - Xi Zhang
- Department of Pediatric Neurology, Northwest Women and Children's Hospital, Xi'an, 710049, China
| | - Bin Guo
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| |
Collapse
|
23
|
Liu X, Qi S, Hou L, Liu Y, Wang X. Noninvasive Deep Brain Stimulation via Temporal Interference Electric Fields Enhanced Motor Performance of Mice and Its Neuroplasticity Mechanisms. Mol Neurobiol 2024; 61:3314-3329. [PMID: 37987957 DOI: 10.1007/s12035-023-03721-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
A noninvasive deep brain stimulation via temporal interference (TI) electric fields is a novel neuromodulation technology, but few advances about TI stimulation effectiveness and mechanisms have been reported. One hundred twenty-six mice were selected for the experiment by power analysis. In the present study, TI stimulation was proved to stimulate noninvasively primary motor cortex (M1) of mice, and 7-day TI stimulation with an envelope frequency of 20 Hz (∆f =20 Hz), instead of an envelope frequency of 10 Hz (∆f =10 Hz), could obviously improve mice motor performance. The mechanism of action may be related to enhancing the strength of synaptic connections, improving synaptic transmission efficiency, increasing dendritic spine density, promoting neurotransmitter release, and increasing the expression and activity of synapse-related proteins, such as brain-derived neurotrophic factor (BDNF), postsynaptic density protein-95 (PSD-95), and glutamate receptor protein. Furthermore, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway and its upstream BDNF play an important role in the enhancement of locomotor performance in mice by TI stimulation. To our knowledge, it is the first report about TI stimulation promoting multiple motor performances and describing its mechanisms. TI stimulation might serve as a novel promising approach to enhance motor performance and treat dysfunction in deep brain regions.
Collapse
Affiliation(s)
- Xiaodong Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Shuo Qi
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Lijuan Hou
- College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Yu Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Xiaohui Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
24
|
Li D, Yu SF, Lin L, Guo JR, Huang SM, Wu XL, You HL, Cheng XJ, Zhang QY, Zeng YQ, Pan XD. Deficiency of leucine-rich repeat kinase 2 aggravates thioacetamide-induced acute liver failure and hepatic encephalopathy in mice. J Neuroinflammation 2024; 21:123. [PMID: 38725082 PMCID: PMC11084037 DOI: 10.1186/s12974-024-03125-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/05/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Hepatic encephalopathy (HE) is closely associated with inflammatory responses. However, as a crucial regulator of the immune and inflammatory responses, the role of leucine-rich repeat kinase 2 (LRRK2) in the pathogenesis of HE remains unraveled. Herein, we investigated this issue in thioacetamide (TAA)-induced HE following acute liver failure (ALF). METHODS TAA-induced HE mouse models of LRRK2 wild type (WT), LRRK2 G2019S mutation (Lrrk2G2019S) and LRRK2 knockout (Lrrk2-/-) were established. A battery of neurobehavioral experiments was conducted. The biochemical indexes and pro-inflammatory cytokines were detected. The prefrontal cortex (PFC), striatum (STR), hippocampus (HIP), and liver were examined by pathology and electron microscopy. The changes of autophagy-lysosomal pathway and activity of critical Rab GTPases were analyzed. RESULTS The Lrrk2-/--HE model reported a significantly lower survival rate than the other two models (24% vs. 48%, respectively, p < 0.05), with no difference found between the WT-HE and Lrrk2G2019S-HE groups. Compared with the other groups, after the TAA injection, the Lrrk2-/- group displayed a significant increase in ammonium and pro-inflammatory cytokines, aggravated hepatic inflammation/necrosis, decreased autophagy, and abnormal phosphorylation of lysosomal Rab10. All three models reported microglial activation, neuronal loss, disordered vesicle transmission, and damaged myelin structure. The Lrrk2-/--HE mice presented no severer neuronal injury than the other genotypes. CONCLUSIONS LRRK2 deficiency may exacerbate TAA-induced ALF and HE in mice, in which inflammatory response is evident in the brain and aggravated in the liver. These novel findings indicate a need of sufficient clinical awareness of the adverse effects of LRRK2 inhibitors on the liver.
Collapse
Affiliation(s)
- Dan Li
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China.
- Fujian Clinical Research Center for Digestive System Tumors and Upper Gastrointestinal Diseases, Fujian, 350001, China.
| | - Shu-Fang Yu
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China
| | - Lin Lin
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Jie-Ru Guo
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China
| | - Si-Mei Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China
| | - Xi-Lin Wu
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
- Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Han-Lin You
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Xiao-Juan Cheng
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Qiu-Yang Zhang
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
| | - Yu-Qi Zeng
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Xiao-Dong Pan
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China.
- Clinical Research Center for Precision Diagnosis and Treatment of Neurological Diseases of Fujian Province, Fuzhou, 350001, China.
| |
Collapse
|
25
|
Zhang XY, Han PP, Zhao YN, Shen XY, Bi X. Crosstalk between autophagy and ferroptosis mediate injury in ischemic stroke by generating reactive oxygen species. Heliyon 2024; 10:e28959. [PMID: 38601542 PMCID: PMC11004216 DOI: 10.1016/j.heliyon.2024.e28959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Stroke represents a significant threat to global human health, characterized by high rates of morbidity, disability, and mortality. Predominantly, strokes are ischemic in nature. Ischemic stroke (IS) is influenced by various cell death pathways, notably autophagy and ferroptosis. Recent studies have increasingly highlighted the interplay between autophagy and ferroptosis, a process likely driven by the accumulation of reactive oxygen species (ROS). Post-IS, either the inhibition of autophagy or its excessive activation can escalate ROS levels. Concurrently, the interaction between ROS and lipids during ferroptosis further augments ROS accumulation. Elevated ROS levels can provoke endoplasmic reticulum stress-induced autophagy and, in conjunction with free iron (Fe2+), can trigger ferroptosis. Moreover, ROS contribute to protein and lipid oxidation, endothelial dysfunction, and an inflammatory response, all of which mediate secondary brain injury following IS. This review succinctly explores the mechanisms of ROS-mediated crosstalk between autophagy and ferroptosis and the detrimental impact of increased ROS on IS. It also offers novel perspectives for IS treatment strategies.
Collapse
Affiliation(s)
- Xing-Yu Zhang
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping-Ping Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yi-Ning Zhao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
26
|
Hack W, Gladen-Kolarsky N, Chatterjee S, Liang Q, Maitra U, Ciesla L, Gray NE. Gardenin A treatment attenuates inflammatory markers, synuclein pathology and deficits in tyrosine hydroxylase expression and improves cognitive and motor function in A53T-α-syn mice. Biomed Pharmacother 2024; 173:116370. [PMID: 38458012 PMCID: PMC11017674 DOI: 10.1016/j.biopha.2024.116370] [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: 11/28/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/10/2024] Open
Abstract
Oxidative stress and neuroinflammation are widespread in the Parkinson's disease (PD) brain and contribute to the synaptic degradation and dopaminergic cell loss that result in cognitive impairment and motor dysfunction. The polymethoxyflavone Gardenin A (GA) has been shown to activate the NRF2-regulated antioxidant pathway and inhibit the NFkB-dependent pro-inflammatory pathway in a Drosophila model of PD. Here, we evaluate the effects of GA on A53T alpha-synuclein overexpressing (A53TSyn) mice. A53TSyn mice were treated orally for 4 weeks with 0, 25, or 100 mg/kg GA. In the fourth week, mice underwent behavioral testing and tissue was harvested for immunohistochemical analysis of tyrosine hydroxylase (TH) and phosphorylated alpha synuclein (pSyn) expression, and quantification of synaptic, antioxidant and inflammatory gene expression. Results were compared to vehicle-treated C57BL6J mice. Treatment with 100 mg/kg GA improved associative memory and decreased abnormalities in mobility and gait in A53TSyn mice. GA treatment also reduced pSyn levels in both the cortex and hippocampus and attenuated the reduction in TH expression in the striatum seen in A53Tsyn mice. Additionally, GA increased cortical expression of NRF2-regulated antioxidant genes and decreased expression of NFkB-dependent pro-inflammatory genes. GA was readily detectable in the brains of treated mice and modulated the lipid profile in the deep gray brain tissue of those animals. While the beneficial effects of GA on cognitive deficits, motor dysfunction and PD pathology are promising, future studies are needed to further fully elucidate the mechanism of action of GA, optimizing dosing and confirm these effects in other PD models.
Collapse
Affiliation(s)
- Wyatt Hack
- Oregon Health & Science University, Neurology, Portland, United States
| | | | | | - Qiaoli Liang
- University of Alabama, Mass spectrometry facility, Chemistry and Biochemistry, Tuscaloosa, United States
| | - Urmila Maitra
- University of Alabama, Biological Sciences, Tuscaloosa, United States
| | - Lukasz Ciesla
- University of Alabama, Biological Sciences, Tuscaloosa, United States.
| | - Nora E Gray
- Oregon Health & Science University, Neurology, Portland, United States.
| |
Collapse
|
27
|
Xiao Y, Hong CA, Liu F, Shi D, Zhu X, Yu C, Jiang N, Li S, Liu Y. Caffeic acid activates mitochondrial UPR to resist pathogen infection in Caenorhabditis elegans via the transcription factor ATFS-1. Infect Immun 2024; 92:e0049423. [PMID: 38294242 PMCID: PMC10929418 DOI: 10.1128/iai.00494-23] [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: 11/29/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
Mitochondria play roles in the resistance of Caenorhabditis elegans against pathogenic bacteria by regulating mitochondrial unfolded protein response (UPRmt). Caffeic acid (CA) (3,4-dihydroxy cinnamic acid) is a major phenolic compound present in several plant species, which exhibits biological activities such as antioxidant, anti-fibrosis, anti-inflammatory, and anti-tumor properties. However, whether caffeic acid influences the innate immune response and the underlying molecular mechanisms remains unknown. In this study, we find that 20 µM caffeic acid enhances innate immunity to resist the Gram-negative pathogen Pseudomonas aeruginosa infection in C. elegans. Meanwhile, caffeic acid also inhibits the growth of pathogenic bacteria. Furthermore, caffeic acid promotes host immune response by reducing the bacterial burden in the intestine. Through genetic screening in C. elegans, we find that caffeic acid promotes innate immunity via the transcription factor ATFS-1. In addition, caffeic acid activates the UPRmt and immune response genes for innate immune response through ATFS-1. Our work suggests that caffeic acid has the potential to protect patients from pathogen infection.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Cao-an Hong
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
- School of Forensic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Liu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dandan Shi
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xinting Zhu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Changyan Yu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nian Jiang
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Sanhua Li
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yun Liu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- School of Forensic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
28
|
Jiang M, Wu W, Xiong Z, Yu X, Ye Z, Wu Z. Targeting autophagy drug discovery: Targets, indications and development trends. Eur J Med Chem 2024; 267:116117. [PMID: 38295689 DOI: 10.1016/j.ejmech.2023.116117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/30/2023] [Accepted: 12/31/2023] [Indexed: 02/25/2024]
Abstract
Autophagy plays a vital role in sustaining cellular homeostasis and its alterations have been implicated in the etiology of many diseases. Drugs development targeting autophagy began decades ago and hundreds of agents were developed, some of which are licensed for the clinical usage. However, no existing intervention specifically aimed at modulating autophagy is available. The obstacles that prevent drug developments come from the complexity of the actual impact of autophagy regulators in disease scenarios. With the development and application of new technologies, several promising categories of compounds for autophagy-based therapy have emerged in recent years. In this paper, the autophagy-targeted drugs based on their targets at various hierarchical sites of the autophagic signaling network, e.g., the upstream and downstream of the autophagosome and the autophagic components with enzyme activities are reviewed and analyzed respectively, with special attention paid to those at preclinical or clinical trials. The drugs tailored to specific autophagy alone and combination with drugs/adjuvant therapies widely used in clinical for various diseases treatments are also emphasized. The emerging drug design and development targeting selective autophagy receptors (SARs) and their related proteins, which would be expected to arrest or reverse the progression of disease in various cancers, inflammation, neurodegeneration, and metabolic disorders, are critically reviewed. And the challenges and perspective in clinically developing autophagy-targeted drugs and possible combinations with other medicine are considered in the review.
Collapse
Affiliation(s)
- Mengjia Jiang
- Department of Pharmacology and Pharmacy, China Jiliang University, China
| | - Wayne Wu
- College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Zijie Xiong
- Department of Pharmacology and Pharmacy, China Jiliang University, China
| | - Xiaoping Yu
- Department of Biology, China Jiliang University, China
| | - Zihong Ye
- Department of Biology, China Jiliang University, China
| | - Zhiping Wu
- Department of Pharmacology and Pharmacy, China Jiliang University, China.
| |
Collapse
|
29
|
Shen XY, Zhang XY, Han PP, Zhao YN, Xu GH, Bi X. Mechanisms of intermittent theta-burst stimulation attenuating nerve injury after ischemic reperfusion in rats through endoplasmic reticulum stress and ferroptosis. Mol Biol Rep 2024; 51:377. [PMID: 38427114 PMCID: PMC10907498 DOI: 10.1007/s11033-024-09241-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) exerts neuroprotective effects early in cerebral ischemia/reperfusion (I/R) injury. Intermittent theta-brust stimulation (iTBS), a more time-efficient modality of rTMS, improves the efficiency without at least decreasing the efficacy of the therapy. iTBS elevates cortical excitability, and in recent years it has become increasingly common to apply iTBS to patients in the early post-IS period. However, little is known about the neuroprotective mechanisms of iTBS. Endoplasmic reticulum stress (ERS), and ferroptosis have been shown to be involved in the development of I/R injury. We aimed to investigate the potential regulatory mechanisms by which iTBS attenuates neurological injury after I/R in rats. METHODS Rats were randomly divided into three groups: sham-operated group, MCAO/R group, and MCAO/R + iTBS group, and were stimulated with iTBS 36 h after undergoing middle cerebral artery occlusion (MCAO) or sham-operated. The expression of ERS, ferroptosis, and apoptosis-related markers was subsequently detected by western blot assays. We also investigated the mechanism by which iTBS attenuates nerve injury after ischemic reperfusion in rats by using the modified Neurological Severity Score (mNSS) and the balance beam test to measure nerve function. RESULTS iTBS performed early in I/R injury attenuated the levels of ERS, ferroptosis, and apoptosis, and improved neurological function, including mNSS and balance beam experiments. It is suggested that this mode of stimulation reduces the cost per treatment by several times without compromising the efficacy of the treatment and could be a practical and less costly intervention.
Collapse
Affiliation(s)
- Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xing-Yu Zhang
- Graduate School of Shanghai, University of Traditional Chinese Medicine, Shanghai, China
| | - Ping-Ping Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yi-Ning Zhao
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Guo-Hui Xu
- Huadong Hospital, Affiliated to Fudan University, 221 West Yan'an Road, Jing'an District, 200040, Shanghai, China.
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.
| |
Collapse
|
30
|
Unnithan D, Sartaj A, Iqubal MK, Ali J, Baboota S. A neoteric annotation on the advances in combination therapy for Parkinson's disease: nanocarrier-based combination approach and future anticipation. Part I: exploring theoretical insights and pharmacological advances. Expert Opin Drug Deliv 2024; 21:423-435. [PMID: 38481172 DOI: 10.1080/17425247.2024.2331214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Parkinson's disease (PD) is a neurological condition defined by a substantial reduction in dopamine-containing cells in the substantia nigra. Levodopa (L-Dopa) is considered the gold standard in treatment. Recent research has clearly shown that resistance to existing therapies can develop. Moreover, the involvement of multiple pathways in the nigrostriatal dopaminergic neuronal loss suggests that modifying the treatment strategy could effectively reduce this degeneration. AREAS COVERED This review summarizes the key concerns with treating PD patients and the combinations, aimed at effectively managing PD. Part I focuses on the clinical diagnosis at every stage of the disease as well as the pharmacological treatment strategies that are applied throughout its course. It methodically elucidates the potency of multifactorial interventions in attenuating the disease trajectory, substantiating the rationale for co-administration of dual or multiple therapeutic agents. Significant emphasis is laid on evidence-based pharmacological combinations for PD management. EXPERT OPINION By utilizing multiple drugs in a combination fashion, this approach can leverage the additive or synergistic effects of these agents, amplify the spectrum of treatment, and curtail the risk of side effects by reducing the dose of each drug, demonstrating significantly greater efficacy.
Collapse
Affiliation(s)
- Devika Unnithan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Ali Sartaj
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Kashif Iqubal
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Texas A&M University, College Station, TX, USA
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
31
|
Singh K, Gupta JK, Sethi P, Mathew S, Bhatt A, Sharma MC, Saha S, Shamim, Kumar S. Recent Advances in the Synthesis of Antioxidant Derivatives: Pharmacological Insights for Neurological Disorders. Curr Top Med Chem 2024; 24:1940-1959. [PMID: 39108007 DOI: 10.2174/0115680266305736240725052825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 10/19/2024]
Abstract
Neurological disorders, characterized by oxidative stress (OS) and inflammation, have become a major global health concern. Redox reactions play a vital role in regulating the balance of the neuronal microenvironment. Specifically, the imbalance leads to a significant weakening of the organism's natural defensive mechanisms. This, in turn, causes the development of harmful oxidative stress, which plays a crucial role in the onset and progression of neurodegenerative diseases. The quest for effective therapeutic agents has led to significant advancements in the synthesis of antioxidant derivatives. This review provides a comprehensive overview of the recent developments in the use of novel antioxidant compounds with potential pharmacological applications in the management of neurological disorders. The discussed compounds encompass a diverse range of chemical structures, including polyphenols, vitamins, flavonoids, and hybrid molecules, highlighting their varied mechanisms of action. This review also focuses on the mechanism of oxidative stress in developing neurodegenerative disease. The neuroprotective effects of these antioxidant derivatives are explored in the context of specific neurological disorders, including Alzheimer's disease, Parkinson's disease, and Huntington's disease. The ultimate goal is to provide effective treatments for these debilitating conditions and improve the quality of life for patients.
Collapse
Affiliation(s)
- Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura Uttar Pradesh, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura Uttar Pradesh, India
| | - Pranshul Sethi
- Department of Pharmacology, College of Pharmacy, Shri Venkateshwara University, Gajraula, Uttar Pradesh, India
| | - Sojomon Mathew
- Department of Zoology, Government College, Kottayam, Kerala, India
| | - Alok Bhatt
- School of Pharmacy, Graphic Era Hill University, Bell Road, Clement Town, Dehradun, Uttarakhand, India
| | | | - Sunam Saha
- Department of Chemistry, Institute of Pharmaceutical Research, GLA University, Mathura Uttar Pradesh, India
| | - Shamim
- IIMT College of Medical Sciences, IIMT University, Meerut, Uttar Pradesh, India
| | - Shivendra Kumar
- Department of Pharmacology, Rajiv Academy for Pharmacy, Mathura, Uttar Pradesh, India
| |
Collapse
|
32
|
Shi Y, Sheng P, Guo M, Chen K, Zhao Y, Wang X, Wu M, Li B. Banxia Xiexin Decoction Prevents HT22 Cells from High Glucose-induced Neurotoxicity via JNK/SIRT1/Foxo3a Signaling Pathway. Curr Comput Aided Drug Des 2024; 20:911-927. [PMID: 37608672 DOI: 10.2174/1573409920666230822110258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Type 2 diabetes-associated cognitive dysfunction (DCD) is a chronic complication of diabetes that has gained international attention. The medicinal compound Banxia Xiexin Decoction (BXXXD) from traditional Chinese medicine (TCM) has shown potential in improving insulin resistance, regulating endoplasmic reticulum stress (ERS), and inhibiting cell apoptosis through various pathways. However, the specific mechanism of action and medical value of BXXXD remain unclear. METHODS We utilized TCMSP databases to screen the chemical constituents of BXXXD and identified DCD disease targets through relevant databases. By using Stitch and String databases, we imported the data into Cytoscape 3.8.0 software to construct a protein-protein interaction (PPI) network and subsequently identified core targets through network topology analysis. The core targets were subjected to Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The results were further validated through in vitro experiments. RESULTS Network pharmacology analysis revealed the screening of 1490 DCD-related targets and 190 agents present in BXXXD. The topological analysis and enrichment analysis conducted using Cytoscape software identified 34 core targets. Additionally, GO and KEGG pathway analyses yielded 104 biological targets and 97 pathways, respectively. BXXXD exhibited its potential in treating DCD by controlling synaptic plasticity and conduction, suppressing apoptosis, reducing inflammation, and acting as an antioxidant. In a high glucose (HG) environment, the expression of JNK, Foxo3a, SIRT1, ATG7, Lamp2, and LC3 was downregulated. BXXXD intervention on HT22 cells potentially involved inhibiting excessive oxidative stress, promoting neuronal autophagy, and increasing the expression levels of JNK, SIRT1, Foxo3a, ATG7, Lamp2, and LC3. Furthermore, the neuroprotective effect of BXXXD was partially blocked by SP600125, while quercetin enhanced the favorable role of BXXXD in the HG environment. CONCLUSION BXXXD exerts its effects on DCD through multiple components, targets, levels, and pathways. It modulates the JNK/SIRT1/Foxo3a signaling pathway to mitigate autophagy inhibition and apoptotic damage in HT22 cells induced by HG. These findings provide valuable perspectives and concepts for future clinical trials and fundamental research.
Collapse
Affiliation(s)
- Yinli Shi
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pei Sheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Guo
- Southeast University, Zhongda Hospital Southeast University, Nanjing, China
| | - Kai Chen
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Zhao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xu Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mianhua Wu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
33
|
Li J, Yu J, Guo J, Liu J, Wan G, Wei X, Yang X, Shi J. Nardostachys jatamansi and levodopa combination alleviates Parkinson's disease symptoms in rats through activation of Nrf2 and inhibition of NLRP3 signaling pathways. PHARMACEUTICAL BIOLOGY 2023; 61:1175-1185. [PMID: 37559448 PMCID: PMC10416743 DOI: 10.1080/13880209.2023.2244176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/10/2023] [Accepted: 07/29/2023] [Indexed: 08/11/2023]
Abstract
CONTEXT Levodopa combined with traditional Chinese medicine has a synergistic effect on Parkinson's disease (PD). Recently, we demonstrated that Nardostachys jatamansi (D. Don) DC. [syn. Patrinia jatamansi D.Don, N. grandiflora DC.] (Valerianaceae) (NJ) can alleviate PD. OBJECTIVE To explore the synergistic effect of NJ combined with levodopa against PD. MATERIALS AND METHODS The PD model was established by injecting rotenone. Eighty-four Sprague-Dawley rats were randomly divided into seven groups: sham, model, different doses of NJ (0.31, 0.62, or 1.24 g/kg) combined with levodopa (25 mg/kg), and levodopa alone (25 and 50 mg/kg) groups. The synergistic effect of the combination was investigated by pharmacodynamic investigation and detection of expression of nuclear factor erythro2-related factor 2 (Nrf2) and NLR family proteins containing Pyrin-related domain 3 (NLRP3) pathways. RESULTS Compared with the model group, NJ + levodopa (1.24 g/kg + 25 mg/kg) increased the moving distance of PD rats in the open field (2395.34 ± 668.73 vs. 1501.41 ± 870.23, p < 0.01), enhanced the stay time on the rotating rod (84.86 ± 18.15 vs. 71.36 ± 17.53, p < 0.01) and the combination was superior to other treatments. The synergistic effects were related to NJ + levodopa (1.24 g/kg + 25 mg/kg) increasing the neurotransmitter levels by 38.80%-88.67% in PD rats, and inhibiting oxidative stress and NLRP3 pathway by activating Nrf2 pathway. DISCUSSION AND CONCLUSIONS NJ combined with levodopa is a promising therapeutic candidate for PD, which provides a scientific basis for the subsequent clinical combination therapy of levodopa to enhance the anti-PD effect.
Collapse
Affiliation(s)
- Jiayuan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiahe Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jianyou Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Jinfeng Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guohui Wan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojia Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jinli Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
34
|
Huang Q, Yang P, Liu Y, Ding J, Lu M, Hu G. The interplay between α-Synuclein and NLRP3 inflammasome in Parkinson's disease. Biomed Pharmacother 2023; 168:115735. [PMID: 37852103 DOI: 10.1016/j.biopha.2023.115735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/20/2023] Open
Abstract
α-Synuclein is a member of a protein of synucleins, which is a presynaptic neuron protein. It is usually highly expressed in the brain and participates in the formation and transmission of nerve synapses. It has been reported that abnormal aggregation of α-Syn can induce the activation of NLRP3 inflammasome in microglia, increase the production of IL-1β, and aggravate neuroinflammation. Therefore, it is recognized as one of the important factors leading to neuroinflammation in Parkinson's disease. In this paper, we aimed to explore the influence of post-translational modification of α-Syn on its pathological aggregation and summarize various pathways that activate NLRP3 triggered by α-Syn and targeted therapeutic strategies, which provided new insights for further exploring the origin and targeted therapy of Parkinson's disease.
Collapse
Affiliation(s)
- Qianhui Huang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Pei Yang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Liu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianhua Ding
- Department of Pharmacology, Nanjing Medical University, Jiangsu 211166, China
| | - Ming Lu
- Department of Pharmacology, Nanjing Medical University, Jiangsu 211166, China.
| | - Gang Hu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Department of Pharmacology, Nanjing Medical University, Jiangsu 211166, China.
| |
Collapse
|
35
|
Hack W, Gladen-Kolarsky N, Chatterjee S, Liang Q, Maitra U, Ciesla L, Gray NE. Gardenin A improves cognitive and motor function in A53T-α-syn mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564401. [PMID: 37961574 PMCID: PMC10634905 DOI: 10.1101/2023.10.27.564401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Oxidative stress and neuroinflammation are widespread in the Parkinson's disease (PD) brain and contribute to the synaptic degradation and dopaminergic cell loss that result in cognitive impairment and motor dysfunction. The polymethoxyflavone Gardenin A (GA) has been shown to activate the NRF2-regulated antioxidant pathway and inhibit the NFkB-dependent pro-inflammatory pathway in a Drosophila model of PD. Here, we evaluate the effects of GA on A53T alpha-synuclein overexpressing (A53TSyn) mice. A53TSyn mice were treated orally for 4 weeks with 0, 25, or 100 mg/kg GA. In the fourth week, mice underwent behavioral testing and tissue was harvested for immunohistochemical analysis of tyrosine hydroxylase (TH) and phosphorylated alpha synuclein (pSyn) expression, and quantification of synaptic, antioxidant and inflammatory gene expression. Results were compared to vehicle-treated C57BL6 mice. Treatment with 100 mg/kg GA improved associative memory and decreased abnormalities in mobility and gait in A53TSyn mice. GA treatment also reduced cortical and hippocampal levels of pSyn and attenuated the reduction in TH expression in the striatum. Additionally, GA increased cortical expression of NRF2-regulated antioxidant genes and decreased expression of NFkB-dependent pro-inflammatory genes. GA was readily detectable in the brains of treated mice and modulated the lipid profile in the deep gray brain tissue of those animals. While the beneficial effects of GA on cognitive deficits, motor dysfunction and PD pathology are promising, future studies are needed to further fully elucidate the mechanism of action of GA, optimizing dosing and confirm these effects in other PD models. Significance Statement The polymethoxyflavone Gardenin A can improve cognitive and motor function and attenuate both increases in phosphorylated alpha synuclein and reductions in tyrosine hydroxylase expression in A53T alpha synuclein overexpressing mice. These effects may be related to activation of the NRF2-regulated antioxidant response and downregulation of NFkB-dependent inflammatory response by Gardenin A in treated animals. The study also showed excellent brain bioavailability of Gardenin A and modifications of the lipid profile, possibly through interactions between Gardenin A with the lipid bilayer, following oral administration. The study confirms neuroprotective activity of Gardenin A previously reported in toxin induced Drosophila model of Parkinson's disease.
Collapse
|
36
|
Wang X, Hu W, Qu L, Wang J, Wu A, Lo HH, Ng JPL, Tang Y, Yun X, Wu J, Wong VKW, Chung SK, Wang L, Luo W, Ji X, Law BYK. Tricin promoted ATG-7 dependent autophagic degradation of α-synuclein and dopamine release for improving cognitive and motor deficits in Parkinson's disease. Pharmacol Res 2023; 196:106874. [PMID: 37586619 DOI: 10.1016/j.phrs.2023.106874] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023]
Abstract
Tricin, a natural nontoxic flavonoid distributed in grasses and euphorbia plants, has been reported to scavenge free radicals, possess anti-inflammatory and antioxidative effects. However, its autophagic effect on Parkinson's disease (PD) has not been elucidated. By adopting cellular and C. elegans models of PD, the autophagic effect of tricin was identified based on the level of autophagy markers (LC3-II and p62). Besides, the pharmacological effects on neurotransmitters (dopamine), inflammatory cytokines (IFN γ, TNFα, MCP-1, IL-10, IL-6 and IL-17A), histology (hematoxylin & eosin and Nissl staining) and behavioural pathology (open-field test, hindlimb clasping, Y-maze, Morris water-maze and nest building test) were also confirmed in the A53T-α-synuclein transgenic PD mouse model. Further experiments demonstrated that tricin induced autophagic flux and lowered the level of α-synuclein through AMPK-p70s6K- and ATG7-dependent mechanism. Compared to the existing clinical PD drugs, tricin mitigated pathogenesis and symptoms of PD with no observable side effects. In summary, tricin is proposed as a potential adjuvant remedy or nutraceutical for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Xingxia Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Hu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Liqun Qu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jian Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug ability Evaluation, Luzhou Key Laboratory of Activity Screening and Draggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hang Hong Lo
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Jerome P L Ng
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Yong Tang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Sichuan Key Medical Laboratory of New Drug Discovery and Drug ability Evaluation, Luzhou Key Laboratory of Activity Screening and Draggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyun Yun
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Jianhui Wu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Vincent Kam Wai Wong
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Sookja Kim Chung
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Faculty of Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Linna Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Weidan Luo
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Xiang Ji
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Betty Yuen Kwan Law
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China.
| |
Collapse
|
37
|
Chandrasekaran V, Hediyal TA, Anand N, Kendaganna PH, Gorantla VR, Mahalakshmi AM, Ghanekar RK, Yang J, Sakharkar MK, Chidambaram SB. Polyphenols, Autophagy and Neurodegenerative Diseases: A Review. Biomolecules 2023; 13:1196. [PMID: 37627261 PMCID: PMC10452370 DOI: 10.3390/biom13081196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Polyphenols are secondary metabolites from plant origin and are shown to possess a wide range of therapeutic benefits. They are also reported as regulators of autophagy, inflammation and neurodegeneration. The autophagy pathway is vital in degrading outdated organelles, proteins and other cellular wastes. The dysregulation of autophagy causes proteinopathies, mitochondrial dysfunction and neuroinflammation thereby contributing to neurodegeneration. Evidence reveals that polyphenols improve autophagy by clearing misfolded proteins in the neurons, suppress neuroinflammation and oxidative stress and also protect from neurodegeneration. This review is an attempt to summarize the mechanism of action of polyphenols in modulating autophagy and their involvement in pathways such as mTOR, AMPK, SIRT-1 and ERK. It is evident that polyphenols cause an increase in the levels of autophagic proteins such as beclin-1, microtubule-associated protein light chain (LC3 I and II), sirtuin 1 (SIRT1), etc. Although it is apparent that polyphenols regulate autophagy, the exact interaction of polyphenols with autophagy markers is not known. These data require further research and will be beneficial in supporting polyphenol supplementation as a potential alternative treatment for regulating autophagy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Vichitra Chandrasekaran
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (V.C.); (T.A.H.); (A.M.M.)
- Center for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India;
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (V.C.); (T.A.H.); (A.M.M.)
- Center for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India;
| | - Nikhilesh Anand
- Department of Pharmacology, College of Medicine, American University of Antigua, Saint John’s P.O. Box W-1451, Antigua and Barbuda;
| | - Pavan Heggadadevanakote Kendaganna
- Center for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India;
| | | | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (V.C.); (T.A.H.); (A.M.M.)
- Center for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India;
| | - Ruchika Kaul Ghanekar
- Symbiosis Centre for Research and Innovation (SCRI), Symbiosis International (Deemed University), Pune 412115, India;
| | - Jian Yang
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Meena Kishore Sakharkar
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (V.C.); (T.A.H.); (A.M.M.)
- Center for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India;
| |
Collapse
|
38
|
Yang L, Nao J, Dong X. The Therapeutic Potential of Hydroxycinnamic Acid Derivatives in Parkinson's Disease: Focus on In Vivo Research Advancements. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37432913 DOI: 10.1021/acs.jafc.3c02787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Hydroxycinnamic acid derivatives (HCDs) are polyphenols that are abundant in cereals, coffee, tea, wine, fruits, vegetables, and other plant-based foods. To aid in the clinical prevention and treatment of Parkinson's disease (PD), we evaluated in vivo investigations of the pharmacological properties of HCDs relevant to PD, and their pharmacokinetic and safety aspects. An extensive search of published journals was conducted using several literature databases, including PubMed, Google Scholar, and the Web of Science. The search terms included "hydroxycinnamic acid derivatives," "ferulic acid," "caffeic acid," "sinapic acid," "p-coumaric acid," "Parkinson's disease," and combinations of these keywords. As of April 2023, 455 preclinical studies were retrieved, of which 364 were in vivo studies; we included 17 of these articles on the pharmaceutics of HCDs in PD. Available evidence supports the protective effects of HCDs in PD due to their anti-inflammatory, antioxidant, as well as antiapoptotic physiological activities. Studies have identified possible molecular targets and pathways for the protective actions of HCDs in PD. However, the paucity of studies on these compounds in PD, and the risk of toxicity induced with high-dose applications, limits their use. Thus, multifaceted studies of HCDs in vitro and in vivo are needed.
Collapse
Affiliation(s)
- Lan Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China
| |
Collapse
|
39
|
Wang Q, Yao S, Yang ZX, Zhou C, Zhang Y, Zhang Y, Zhang L, Li JT, Xu ZJ, Zhu WL, Zhang NX, Ye Y, Feng LY. Pharmacological characterization of the small molecule 03A10 as an inhibitor of α-synuclein aggregation for Parkinson's disease treatment. Acta Pharmacol Sin 2023; 44:1122-1134. [PMID: 36627343 PMCID: PMC10203352 DOI: 10.1038/s41401-022-01039-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
Aggregation of α-synuclein, a component of Lewy bodies (LBs) or Lewy neurites in Parkinson's disease (PD), is strongly linked with disease development, making it an attractive therapeutic target. Inhibiting aggregation can slow or prevent the neurodegenerative process. However, the bottleneck towards achieving this goal is the lack of such inhibitors. In the current study, we established a high-throughput screening platform to identify candidate compounds for preventing the aggregation of α-synuclein among the natural products in our in-house compound library. We found that a small molecule, 03A10, i.e., (+)-desdimethylpinoresinol, which is present in the fruits of Vernicia fordii (Euphorbiaceae), modulated aggregated α-synuclein, but not monomeric α-synuclein, to prevent further elongation of α-synuclein fibrils. In α-synuclein-overexpressing cell lines, 03A10 (10 μM) efficiently prevented α-synuclein aggregation and markedly ameliorated the cellular toxicity of α-synuclein fibril seeds. In the MPTP/probenecid (MPTP/p) mouse model, oral administration of 03A10 (0.3 mg· kg-1 ·d-1, 1 mg ·kg-1 ·d-1, for 35 days) significantly alleviated behavioral deficits, tyrosine hydroxylase (TH) neuron degeneration and p-α-synuclein aggregation in the substantia nigra (SN). As the Braak hypothesis postulates that the prevailing site of early PD pathology is the gastrointestinal tract, we inoculated α-synuclein preformed fibrils (PFFs) into the mouse colon. We demonstrated that α-synuclein PFF inoculation promoted α-synuclein pathology and neuroinflammation in the gut and brain; oral administration of 03A10 (5 mg· kg-1 ·d-1, for 4 months) significantly attenuated olfactory deficits, α-synuclein accumulation and neuroinflammation in the olfactory bulb and SN. We conclude that 03A10 might be a promising drug candidate for the treatment of PD. 03A10 might be a novel drug candidate for PD treatment, as it inhibits α-synuclein aggregation by modulating aggregated α-synuclein rather than monomeric α-synuclein to prevent further elongation of α-synuclein fibrils and prevent α-synuclein toxicity in vitro, in an MPTP/p mouse model, and PFF-inoculated mice.
Collapse
Affiliation(s)
- Qing Wang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
| | - Sheng Yao
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, China
| | - Ze-Xian Yang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
| | - Chen Zhou
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Yu Zhang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Ye Zhang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
| | - Lei Zhang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Jin-Tian Li
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhi-Jian Xu
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei-Liang Zhu
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Nai-Xia Zhang
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China.
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
| | - Yang Ye
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China.
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201203, China.
| | - Lin-Yin Feng
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
40
|
Taban Akça K, Çınar Ayan İ, Çetinkaya S, Miser Salihoğlu E, Süntar İ. Autophagic mechanisms in longevity intervention: role of natural active compounds. Expert Rev Mol Med 2023; 25:e13. [PMID: 36994671 PMCID: PMC10407225 DOI: 10.1017/erm.2023.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/14/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023]
Abstract
The term 'autophagy' literally translates to 'self-eating' and alterations to autophagy have been identified as one of the several molecular changes that occur with aging in a variety of species. Autophagy and aging, have a complicated and multifaceted relationship that has recently come to light thanks to breakthroughs in our understanding of the various substrates of autophagy on tissue homoeostasis. Several studies have been conducted to reveal the relationship between autophagy and age-related diseases. The present review looks at a few new aspects of autophagy and speculates on how they might be connected to both aging and the onset and progression of disease. Additionally, we go over the most recent preclinical data supporting the use of autophagy modulators as age-related illnesses including cancer, cardiovascular and neurodegenerative diseases, and metabolic dysfunction. It is crucial to discover important targets in the autophagy pathway in order to create innovative therapies that effectively target autophagy. Natural products have pharmacological properties that can be therapeutically advantageous for the treatment of several diseases and they also serve as valuable sources of inspiration for the development of possible new small-molecule drugs. Indeed, recent scientific studies have shown that several natural products including alkaloids, terpenoids, steroids, and phenolics, have the ability to alter a number of important autophagic signalling pathways and exert therapeutic effects, thus, a wide range of potential targets in various stages of autophagy have been discovered. In this review, we summarised the naturally occurring active compounds that may control the autophagic signalling pathways.
Collapse
Affiliation(s)
- Kevser Taban Akça
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - İlknur Çınar Ayan
- Department of Medical Biology, Medical Faculty, Necmettin Erbakan University, Meram, Konya, Türkiye
| | - Sümeyra Çetinkaya
- Biotechnology Research Center of Ministry of Agriculture and Forestry, Yenimahalle, Ankara, Türkiye
| | - Ece Miser Salihoğlu
- Biochemistry Department, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - İpek Süntar
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| |
Collapse
|
41
|
Paeoniflorin protects 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease mice by inhibiting oxidative stress and neuronal apoptosis through activating the Nrf2/HO-1 signaling pathway. Neuroreport 2023; 34:255-266. [PMID: 36881748 DOI: 10.1097/wnr.0000000000001884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
OBJECTIVES This study aimed to explore the neuroprotective effects of paeoniflorin on oxidative stress and apoptosis in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) mice. METHODS The effects of paeoniflorin on motor function in mice were evaluated by behavioral test. Then substantia nigra of mice were collected and neuronal damage was assessed using Nissl staining. Positive expression of tyrosine hydroxylase (TH) was detected by immunohistochemistry. Levels of malondialdehyde, superoxide dismutase (SOD) and glutathione were measured by biochemical method. terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay was used to detect apoptosis of dopaminergic neurons. Western blotting and real-time fluorescence quantitative PCR were used to detect the protein and mRNA expressions of Nrf2, heme oxygenase-1 (HO-1), B-cell lymphoma-2(Bcl-2), Bax and cleaved caspase-3. RESULTS Paeoniflorin treatment significantly ameliorated the motor performance impairment in MPTP-induced PD mice. Moreover, it notably increased the positive expression rate of TH and reduced the damage and apoptosis of dopaminergic neurons in the substantia nigra. Furthermore, paeoniflorin increased the levels of SOD and glutathione and decreased the malondialdehyde content. It also promoted Nrf2 nuclear translocation, increased the protein and mRNA expressions of HO-1 and Bcl-2 and reduced the protein and mRNA expressions of BCL2-Associated X2 (Bax) and cleaved caspase-3. Treatment with the Nrf2 inhibitor, ML385, notably reduced the effects of paeoniflorin in MPTP-induced PD mice. CONCLUSIONS Neuroprotective effects of paeoniflorin in MPTP-induced PD mice may be mediated via inhibition of oxidative stress and apoptosis of dopaminergic neurons in substantia nigra through activation of the Nrf2/HO-1 signaling pathway.
Collapse
|
42
|
Lv QK, Tao KX, Wang XB, Yao XY, Pang MZ, Liu JY, Wang F, Liu CF. Role of α-synuclein in microglia: autophagy and phagocytosis balance neuroinflammation in Parkinson's disease. Inflamm Res 2023; 72:443-462. [PMID: 36598534 DOI: 10.1007/s00011-022-01676-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease, and is characterized by accumulation of α-synuclein (α-syn). Neuroinflammation driven by microglia is an important pathological manifestation of PD. α-Syn is a crucial marker of PD, and its accumulation leads to microglia M1-like phenotype polarization, activation of NLRP3 inflammasomes, and impaired autophagy and phagocytosis in microglia. Autophagy of microglia is related to degradation of α-syn and NLRP3 inflammasome blockage to relieve neuroinflammation. Microglial autophagy and phagocytosis of released α-syn or fragments from apoptotic neurons maintain homeostasis in the brain. A variety of PD-related genes such as LRRK2, GBA and DJ-1 also contribute to this stability process. OBJECTIVES Further studies are needed to determine how α-syn works in microglia. METHODS A keyword-based search was performed using the PubMed database for published articles. CONCLUSION In this review, we discuss the interaction between microglia and α-syn in PD pathogenesis and the possible mechanism of microglial autophagy and phagocytosis in α-syn clearance and inhibition of neuroinflammation. This may provide a novel insight into treatment of PD.
Collapse
Affiliation(s)
- Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Kang-Xin Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Bo Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Yu Yao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Meng-Zhu Pang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
43
|
Liu C, Yan L, Qian Y, Song P, Wang T, Wei M. The Extract of Acanthopanacis Cortex Relieves the Depression-Like Behavior and Modulates IL-17 Signaling in Chronic Mild Stress-Induced Depressive Mice. Dose Response 2023; 21:15593258221148817. [PMID: 36865497 PMCID: PMC9972068 DOI: 10.1177/15593258221148817] [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] [Indexed: 03/03/2023] Open
Abstract
Background Acanthopanacis Cortex (AC) is a valuable Chinese medicine, which exerts beneficial effects on anti-fatigue, anti-stress, and inflammatory modulation in the periphery. However, the central nervous system (CNS) function of AC has not been clearly illustrated. As communication between the peripheral immune system and the CNS converges, it promotes a heightened neuroinflammatory environment that contributes to depression. We investigated the effect of AC against depression through neuroinflammatory modulation. Methods Network pharmacology was used to screen for target compounds and pathways. Mice with CMS-induced depression were used to evaluate the efficacy of AC against depression. Behavioral studies and detection of neurotransmitters, neurotrophic factors, and pro-inflammatory cytokines were performed. The IL-17 signaling cascade was involved to further investigate the underlying mechanism of AC against depression. Results Twenty-five components were screened by network pharmacology and the IL-17 mediated signaling pathway was associated with the antidepressant action of AC. This herb had a beneficial effect on CMS-induced depressive mice, including improvements in depressive behavior, modulation of neurotransmitter levels, neurotrophic factors, and pro-inflammatory cytokines. Conclusions Our results revealed that AC exhibits effects on anti-depression and one of the mechanisms was mediated by neuroinflammatory modulation.
Collapse
Affiliation(s)
- Chuhan Liu
- Nanjing University of Chinese
Medicine, Nanjing, China,Jiangsu Key Laboratory for the
Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy
of Sciences, Nanjing, China
| | - Lu Yan
- Jiangsu Key Laboratory for the
Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy
of Sciences, Nanjing, China
| | - Yiyun Qian
- Jiangsu Key Laboratory for the
Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy
of Sciences, Nanjing, China
| | - Pingping Song
- Jiangsu Key Laboratory for the
Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy
of Sciences, Nanjing, China
| | - Tao Wang
- New drug screening center/Jiangsu
Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical
University, Nanjing, China
| | - Min Wei
- Nanjing University of Chinese
Medicine, Nanjing, China,Jiangsu Key Laboratory for the
Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy
of Sciences, Nanjing, China,Min Wei, Nanjing University of Chinese
Medicine, No.138 Xianlin Avenue, Qixia District, Nanjing 210028, China.
| |
Collapse
|
44
|
Galzitskaya OV, Grishin SY, Glyakina AV, Dovidchenko NV, Konstantinova AV, Kravchenko SV, Surin AK. The Strategies of Development of New Non-Toxic Inhibitors of Amyloid Formation. Int J Mol Sci 2023; 24:3781. [PMID: 36835194 PMCID: PMC9964835 DOI: 10.3390/ijms24043781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
In recent years, due to the aging of the population and the development of diagnostic medicine, the number of identified diseases associated with the accumulation of amyloid proteins has increased. Some of these proteins are known to cause a number of degenerative diseases in humans, such as amyloid-beta (Aβ) in Alzheimer's disease (AD), α-synuclein in Parkinson's disease (PD), and insulin and its analogues in insulin-derived amyloidosis. In this regard, it is important to develop strategies for the search and development of effective inhibitors of amyloid formation. Many studies have been carried out aimed at elucidating the mechanisms of amyloid aggregation of proteins and peptides. This review focuses on three amyloidogenic peptides and proteins-Aβ, α-synuclein, and insulin-for which we will consider amyloid fibril formation mechanisms and analyze existing and prospective strategies for the development of effective and non-toxic inhibitors of amyloid formation. The development of non-toxic inhibitors of amyloid will allow them to be used more effectively for the treatment of diseases associated with amyloid.
Collapse
Affiliation(s)
- Oxana V. Galzitskaya
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Sergei Y. Grishin
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, 625003 Tyumen, Russia
| | - Anna V. Glyakina
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Mathematical Problems of Biology RAS, The Branch of Keldysh Institute of Applied Mathematics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Nikita V. Dovidchenko
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Anastasiia V. Konstantinova
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Faculty of Biotechnology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Sergey V. Kravchenko
- Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, 625003 Tyumen, Russia
| | - Alexey K. Surin
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- The Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
- State Research Center for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia
| |
Collapse
|
45
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Honey and Alzheimer's Disease-Current Understanding and Future Prospects. Antioxidants (Basel) 2023; 12:427. [PMID: 36829985 PMCID: PMC9952506 DOI: 10.3390/antiox12020427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, has been a global concern. AD is associated with the involvement of the central nervous system that causes the characteristic impaired memory, cognitive deficits, and behavioral abnormalities. These abnormalities caused by AD is known to be attributed by extracellular aggregates of amyloid beta plaques and intracellular neurofibrillary tangles. Additionally, genetic factors such as abnormality in the expression of APOE, APP, BACE1, PSEN-1, and PSEN-2 play a role in the disease. As the current treatment aims to treat the symptoms and to slow the disease progression, there has been a continuous search for new nutraceutical agent or medicine to help prevent and cure AD pathology. In this quest, honey has emerged as a powerful nootropic agent. Numerous studies have demonstrated that the high flavonoids and phenolic acids content in honey exerts its antioxidant, anti-inflammatory, and neuroprotective properties. This review summarizes the effect of main flavonoid compounds found in honey on the physiological functioning of the central nervous system, and the effect of honey intake on memory and cognition in various animal model. This review provides a new insight on the potential of honey to prevent AD pathology, as well as to ameliorate the damage in the developed AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
46
|
Shuai W, Bu F, Zhu Y, Wu Y, Xiao H, Pan X, Zhang J, Sun Q, Wang G, Ouyang L. Discovery of Novel Indazole Chemotypes as Isoform-Selective JNK3 Inhibitors for the Treatment of Parkinson's Disease. J Med Chem 2023; 66:1273-1300. [PMID: 36649216 DOI: 10.1021/acs.jmedchem.2c01410] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
c-Jun N-terminal kinases (JNKs) are involved in the pathogenesis of various diseases. In particular, JNK3 and not JNK1/2 is primarily expressed in the brain and plays a key role in mediating neurodegenerative diseases like Parkinson's disease (PD). Due to the sequence similarity of JNK isoforms, developing isoform-selective JNK3 inhibitors to evaluate their biological functions and therapeutic potential in PD has become a challenge. Herein, docking-based virtual screening and structure-activity relationship studies identified 25c with excellent inhibitory activity against JNK3 (IC50 = 85.21 nM) and exhibited an over 100-fold isoform selectivity for JNK3 over JNK1/2 and remarkable kinase selectivity. 25c showed neuroprotective effects on in vitro and in vivo PD models by selectively inhibiting JNK3. Meanwhile, 25c showed an ideal blood-brain barrier permeability and low toxicity. Overall, this study provided a valuable molecular tool for investigating the role of JNK3 in PD and a solid foundation for developing JNK3-targeted drugs in PD treatment.
Collapse
Affiliation(s)
- Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Faqian Bu
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yumeng Zhu
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yongya Wu
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xiaoli Pan
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Varesi A, Campagnoli LIM, Carrara A, Pola I, Floris E, Ricevuti G, Chirumbolo S, Pascale A. Non-Enzymatic Antioxidants against Alzheimer's Disease: Prevention, Diagnosis and Therapy. Antioxidants (Basel) 2023; 12:180. [PMID: 36671042 PMCID: PMC9855271 DOI: 10.3390/antiox12010180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive memory loss and cognitive decline. Although substantial research has been conducted to elucidate the complex pathophysiology of AD, the therapeutic approach still has limited efficacy in clinical practice. Oxidative stress (OS) has been established as an early driver of several age-related diseases, including neurodegeneration. In AD, increased levels of reactive oxygen species mediate neuronal lipid, protein, and nucleic acid peroxidation, mitochondrial dysfunction, synaptic damage, and inflammation. Thus, the identification of novel antioxidant molecules capable of detecting, preventing, and counteracting AD onset and progression is of the utmost importance. However, although several studies have been published, comprehensive and up-to-date overviews of the principal anti-AD agents harboring antioxidant properties remain scarce. In this narrative review, we summarize the role of vitamins, minerals, flavonoids, non-flavonoids, mitochondria-targeting molecules, organosulfur compounds, and carotenoids as non-enzymatic antioxidants with AD diagnostic, preventative, and therapeutic potential, thereby offering insights into the relationship between OS and neurodegeneration.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | | | - Adelaide Carrara
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Ilaria Pola
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Elena Floris
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
48
|
Azlan UK, Khairul Annuar NA, Mediani A, Aizat WM, Damanhuri HA, Tong X, Yanagisawa D, Tooyama I, Wan Ngah WZ, Jantan I, Hamezah HS. An insight into the neuroprotective and anti-neuroinflammatory effects and mechanisms of Moringa oleifera. Front Pharmacol 2023; 13:1035220. [PMID: 36686668 PMCID: PMC9849397 DOI: 10.3389/fphar.2022.1035220] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Neurodegenerative diseases (NDs) are sporadic maladies that affect patients' lives with progressive neurological disabilities and reduced quality of life. Neuroinflammation and oxidative reaction are among the pivotal factors for neurodegenerative conditions, contributing to the progression of NDs, such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS) and Huntington's disease (HD). Management of NDs is still less than optimum due to its wide range of causative factors and influences, such as lifestyle, genetic variants, and environmental aspects. The neuroprotective and anti-neuroinflammatory activities of Moringa oleifera have been documented in numerous studies due to its richness of phytochemicals with antioxidant and anti-inflammatory properties. This review highlights up-to-date research findings on the anti-neuroinflammatory and neuroprotective effects of M. oleifera, including mechanisms against NDs. The information was gathered from databases, which include Scopus, Science Direct, Ovid-MEDLINE, Springer, and Elsevier. Neuroprotective effects of M. oleifera were mainly assessed by using the crude extracts in vitro and in vivo experiments. Isolated compounds from M. oleifera such as moringin, astragalin, and isoquercitrin, and identified compounds of M. oleifera such as phenolic acids and flavonoids (chlorogenic acid, gallic acid, ferulic acid, caffeic acid, kaempferol, quercetin, myricetin, (-)-epicatechin, and isoquercitrin) have been reported to have neuropharmacological activities. Therefore, these compounds may potentially contribute to the neuroprotective and anti-neuroinflammatory effects. More in-depth studies using in vivo animal models of neurological-related disorders and extensive preclinical investigations, such as pharmacokinetics, toxicity, and bioavailability studies are necessary before clinical trials can be carried out to develop M. oleifera constituents into neuroprotective agents.
Collapse
Affiliation(s)
- Ummi Kalthum Azlan
- 1Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | | | - Ahmed Mediani
- 1Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Wan Mohd Aizat
- 1Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Hanafi Ahmad Damanhuri
- 2Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Xiaohui Tong
- 3School of Life Sciences, Anhui University of Chinese Medicine, Hefei, China
| | - Daijiro Yanagisawa
- 4Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Tooyama
- 5Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Wan Zurinah Wan Ngah
- 5Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Ibrahim Jantan
- 1Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Hamizah Shahirah Hamezah
- 1Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia,*Correspondence: Hamizah Shahirah Hamezah,
| |
Collapse
|
49
|
Caffeic Acid and Diseases-Mechanisms of Action. Int J Mol Sci 2022; 24:ijms24010588. [PMID: 36614030 PMCID: PMC9820408 DOI: 10.3390/ijms24010588] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
Caffeic acid belongs to the polyphenol compounds we consume daily, often in the form of coffee. Even though it is less explored than caffeic acid phenethyl ester, it still has many positive effects on human health. Caffeic acid can affect cancer, diabetes, atherosclerosis, Alzheimer's disease, or bacterial and viral infections. This review focuses on the molecular mechanisms of how caffeic acid achieves its effects.
Collapse
|
50
|
Goncalves VC, Silva da Fonsêca V, de Paula Faria D, Izidoro MA, Berretta AA, de Almeida ACG, Affonso Fonseca FL, Scorza FA, Scorza CA. Propolis induces cardiac metabolism changes in 6-hydroxydopamine animal model: A dietary intervention as a potential cardioprotective approach in Parkinson’s disease. Front Pharmacol 2022; 13:1013703. [PMID: 36313332 PMCID: PMC9606713 DOI: 10.3389/fphar.2022.1013703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022] Open
Abstract
While there is sustained growth of the older population worldwide, ageing is a consistent risk factor for neurodegenerative diseases, such as Parkinson’s-disease (PD). Considered an emblematic movement disorder, PD comprises a miscellany of non-motor symptoms, for which effective management remains an unfulfilled need in clinical practice. Highlighted are the cardiovascular abnormalities, that cause significant burden in PD patients. Evidence suggests that key biological processes underlying PD pathophysiology can be modulated by diet-derived bioactive compounds, such as green propolis, a natural functional food with biological and pharmacological properties. The effects of propolis on cardiac affection associated to PD have received little coverage. In this study, a metabolomics approach and Positron Emission Tomography (PET) imaging were used to assess the metabolic response to diet supplementation with green propolis on heart outcomes of rats with Parkinsonism induced by 6-hydroxydopamine (6-OHDA rats). Untargeted metabolomics approach revealed four cardiac metabolites (2-hydroxybutyric acid, 3-hydroxybutyric acid, monoacylglycerol and alanine) that were significantly modified between animal groups (6-OHDA, 6-OHDA + Propolis and sham). Propolis-induced changes in the level of these cardiac metabolites suggest beneficial effects of diet intervention. From the metabolites affected, functional analysis identified changes in propanoate metabolism (a key carbohydrate metabolism related metabolic pathway), glucose-alanine cycle, protein and fatty acid biosynthesis, energy metabolism, glutathione metabolism and urea cycle. PET imaging detected higher glucose metabolism in the 17 areas of the left ventricle of all rats treated with propolis, substantially contrasting from those rats that did not consume propolis. Our results bring new insights into cardiac metabolic substrates and pathways involved in the mechanisms of the effects of propolis in experimental PD and provide potential novel targets for research in the quest for future therapeutic strategies.
Collapse
Affiliation(s)
- Valeria C. Goncalves
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- *Correspondence: Valeria C. Goncalves, ; Carla Alessandra Scorza,
| | - Victor Silva da Fonsêca
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | - Mario Augusto Izidoro
- Laboratório de Espectrometria de Massas—Associação Beneficente de Coleta de Sangue (COLSAN), São Paulo, Brazil
| | | | - Antônio-Carlos G. de Almeida
- Laboratório de Neurociências Experimental e Computacional, Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del-Rei (UFSJ), Minas Gerais, Brazil
| | - Fernando Luiz Affonso Fonseca
- Laboratório de Análises Clínicas da Faculdade de Medicina Do ABC, Santo André, São Paulo, Brazil
- Departamento de Ciencias Farmaceuticas da Universidade Federal de Sao Paulo (UNIFESP), Diadema, Brazil
| | - Fulvio Alexandre Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Carla Alessandra Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- *Correspondence: Valeria C. Goncalves, ; Carla Alessandra Scorza,
| |
Collapse
|