1
|
Silva T, Alves C, Evtyugina MG, Dias AS, Pereira GM, de Castro Vasconcellos P, de Fátima Andrade M, Oliveira H, Duarte IF. Cytotoxic and metabolic effects of organic extracts from airborne fine particulate matter (PM 2.5) in neuronal cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 116:104707. [PMID: 40311788 DOI: 10.1016/j.etap.2025.104707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Airborne fine particulate matter (PM2.5) has been linked to neurological diseases, but its cellular and metabolic effects remain incompletely understood. This study assessed the cytotoxic and metabolic impact of PM2.5 samples from São Paulo, Brazil, on SH-SY5Y neuroblastoma cells. Even at low toxicity levels (IC10-IC30), PM2.5 organic extracts induced apoptosis, increased TNF-α secretion, and triggered moderate oxidative responses. Metabolomic analyses revealed a downregulation of energy-producing pathways, including glycolysis and the TCA cycle, along with decreased ATP and phosphocreatine levels. Compensatory adaptations were evident, such as increased proline oxidation, lipid accumulation, and activation of the creatine-phosphocreatine system. One-carbon metabolism was also affected, with changes suggesting suppression of the folate and methionine cycles. Elevated glutathione levels indicated an enhanced antioxidant response. These findings highlight how PM2.5 disrupts neuronal energy homeostasis and redox balance, offering new insights into the cellular mechanisms of air pollution-related neurotoxicity.
Collapse
Affiliation(s)
- Tatiana Silva
- Department of Chemistry, CICECO - Aveiro Institute of Materials and LAQV-REQUIMTE, University of Aveiro, Aveiro 3810-193, Portugal; Department of Biology, CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Aveiro 3810-193, Portugal
| | - Célia Alves
- Department of Environment and Planning, CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Aveiro 3810-193, Portugal
| | - Margarita G Evtyugina
- Department of Environment and Planning, CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana S Dias
- Department of Chemistry, CICECO - Aveiro Institute of Materials and LAQV-REQUIMTE, University of Aveiro, Aveiro 3810-193, Portugal
| | - Guilherme Martins Pereira
- Institute of Chemistry, University of São Paulo, São Paulo, SP 05508-000, Brazil; Institute of Astronomy, Geophysics and Atmospheric Sciences, University of São Paulo, São Paulo, SP 05508-090, Brazil
| | | | - Maria de Fátima Andrade
- Institute of Astronomy, Geophysics and Atmospheric Sciences, University of São Paulo, São Paulo, SP 05508-090, Brazil
| | - Helena Oliveira
- Department of Biology, CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Aveiro 3810-193, Portugal
| | - Iola F Duarte
- Department of Chemistry, CICECO - Aveiro Institute of Materials and LAQV-REQUIMTE, University of Aveiro, Aveiro 3810-193, Portugal.
| |
Collapse
|
2
|
Liu Y, Luo J, Liu Y, Liu W, Yu G, Huang Y, Yang Y, Chen X, Chen T. Brain-Targeted Biomimetic Nanodecoys with Neuroprotective Effects for Precise Therapy of Parkinson's Disease. ACS CENTRAL SCIENCE 2022; 8:1336-1349. [PMID: 36188350 PMCID: PMC9523773 DOI: 10.1021/acscentsci.2c00741] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Indexed: 06/16/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the gradual loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein aggregates called Lewy bodies. Here, nanodecoys were designed from a rabies virus polypeptide with a 29 amino acid (RVG29)-modified red blood cell membrane (RBCm) to encapsulate curcumin nanocrystals (Cur-NCs), which could effectively protect dopaminergic neurons. The RVG29-RBCm/Cur-NCs nanodecoys effectively escaped from reticuloendothelial system (RES) uptake, enabled prolonged blood circulation, and enhanced blood-brain barrier (BBB) crossing after systemic administration. Cur-NCs loaded inside the nanodecoys exhibited the recovery of dopamine levels, inhibition of α-synuclein aggregation, and reversal of mitochondrial dysfunction in PD mice. These findings indicate the promising potential of biomimetic nanodecoys in treating PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yao Liu
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| | - Jingshan Luo
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| | - Yujing Liu
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| | - Wen Liu
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University
of Macau, Macau 999078, China
| | - Guangtao Yu
- Stomatological
Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuting Huang
- Institute
of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yang
- Institute
of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaojia Chen
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University
of Macau, Macau 999078, China
| | - Tongkai Chen
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
3
|
Comparative Metabolomics Study of the Impact of Articaine and Lidocaine on the Metabolism of SH-SY5Y Neuronal Cells. Metabolites 2022; 12:metabo12070581. [PMID: 35888705 PMCID: PMC9323911 DOI: 10.3390/metabo12070581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Articaine (ATC) and lidocaine (LDC) are the local anesthetics (LAs) currently most employed in dentistry. Cases of paresthesia, reported more frequently for ATC, have raised concerns about their potential neurotoxicity, calling for further investigation of their biological effects in neuronal cells. In this work, the impact of ATC and LDC on the metabolism of SH-SY5Y cells was investigated through 1H NMR metabolomics. For each LA, in vitro cultured cells were exposed to concentrations causing 10 and 50% reductions in cell viability, and their metabolic intracellular and extracellular profiles were characterized. Most effects were common to ATC and LDC, although with varying magnitudes. The metabolic variations elicited by the two LAs suggested (i) downregulation of glycolysis and of glucose-dependent pathways (e.g., one-carbon metabolism and hexosamine biosynthetic pathway), (ii) disturbance of branched chain amino acids (BCAA) catabolism, (iii) downregulation of TCA cycle anaplerotic fueling and activation of alternative energy producing pathways, (iv) interference with choline metabolism and (v) lipid droplet build-up. Interestingly, LDC had a greater impact on membrane phospholipid turnover, as suggested by higher phosphatidylcholine to phosphocholine conversion. Moreover, LDC elicited an increase in triglycerides, whereas cholesteryl esters accumulated in ATC-exposed cells, suggesting a different composition and handling of lipid droplets.
Collapse
|
4
|
Wang W, Zhu G, Wang Y, Li W, Yi S, Wang K, Fan L, Tang J, Chen R. Multi-Omics Integration in Mice With Parkinson’s Disease and the Intervention Effect of Cyanidin-3-O-Glucoside. Front Aging Neurosci 2022; 14:877078. [PMID: 35572129 PMCID: PMC9099026 DOI: 10.3389/fnagi.2022.877078] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Background Parkinson’s disease (PD) is a multifactorial degenerative disease of the central nervous system, which affects mostly older adults. To date, research has focused on the progression of PD. Simultaneously, it was confirmed that the imbalances in gut microbiota are associated with the onset and progression of PD. Accurate diagnosis and precise treatment of PD are currently deficient due to the absence of effective biomarkers. Methods In this study, the pharmacodynamic study of cyanidin-3-O-glucoside in PD mice was used. It intends to use the “imbalance” and “balance” of intestinal microecology as the starting point to investigate the “gut-to-brain” hypothesis using metabolomic-combined 16S rRNA gene sequencing methods. Simultaneously, metabolomic analysis was implemented to acquire differential metabolites, and microbiome analysis was performed to analyze the composition and filter the remarkably altered gut microbiota at the phylum/genera level. Afterward, metabolic pathway and functional prediction analysis of the screened differential metabolites and gut microbiota were applied using the MetaboAnalyst database. In addition, Pearson’s correlation analysis was used for the differential metabolites and gut microbiota. We found that cyanidin-3-O-glucoside could protect 1-methyl-4-phenyl-1,2,3,6− tetrahydropy ridine (MPTP)-induced PD mice. Results Metabolomic analysis showed that MPTP-induced dysbiosis of the gut microbiota significantly altered sixty-seven metabolites. The present studies have also shown that MPTP-induced PD is related to lipid metabolism, amino acid metabolism, and so on. The 16S rRNA sequencing analysis indicated that 5 phyla and 22 genera were significantly altered. Furthermore, the differential gut microbiota was interrelated with amino acid metabolism, and so on. The metabolites and gut microbiota network diagram revealed significant correlations between 11 genera and 8 differential metabolites. Conclusion In combination, this study offers potential molecular biomarkers that should be validated for future translation into clinical applications for more accurately diagnosing PD. Simultaneously, the results of this study lay a basis for further study of the association between host metabolisms, gut microbiota, and PD.
Collapse
Affiliation(s)
- Wang Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoxue Zhu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuwen Wang
- The Sixth Outpatient Department, Jinling Hospital, Nanjing, China
| | - Wei Li
- College of Traditional Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shilin Yi
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Kai Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Fan
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Lu Fan,
| | - Juanjuan Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Juanjuan Tang,
| | - Ruini Chen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Ruini Chen,
| |
Collapse
|
5
|
Mack N, Mazzio E, Badisa R, Soliman KFA. Metabolic Response to the Mitochondrial Toxin 1-Methyl-4-phenylpyridinium (MPP+) in LDH-A/B Double-knockout LS174T Colon Cancer Cells. Cancer Genomics Proteomics 2021; 18:385-405. [PMID: 33994363 DOI: 10.21873/cgp.20267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/15/2021] [Accepted: 04/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Rapid glycolytic substrate-level phosphorylation (SLP) and accumulation of lactic acid are characteristics of diverse cancers. Recent advances in drug discovery have included the use of glycolytic inhibitors with mitochondrial targeting drugs to attempt to invoke an energy crisis in aggressive metabolically active chemo-resistant cancers. In this work, we examine the consequences of inhibiting mitochondrial oxidative phosphorylation (OXPHOS) with 1-methyl-4-phenylpyridinium (MPP+) in LS14T colon cancer cells containing a genetic double knock out (DKO) of lactic acid dehydrogenase (LDHA and LDHB). MATERIALS AND METHODS Several metabolic parameters were evaluated concomitant to whole transcriptomic (WT) mRNA, microRNA, and long intergenic non-coding RNAs using Affymetrix 2.1 human ST arrays. RESULTS MPP+ effectively blocked OXPHOS where a compensatory shift toward anaerobic SLP was only observed in the control vector (CV), and not observed in the LDH-A/B DKOs (lacking the ability to produce lactic acid). Despite this, there was an unexpected resilience to MPP+ in the latter in terms of energy, which displayed significantly higher resting baseline respiratory OXPHOS capacity relative to controls. At the transcriptome level, MPP+ invoked 1738 differential expressed genes (DEGs) out of 48,226; LDH-A/B DKO resulted in 855 DEGs while 349 DEGs were found to be overlapping in both groups versus respective controls, including loss of mitochondrial complex I (subunits 3 and 6), cell cycle transcripts and fluctuations in epigenetic chromatin remodeling systems. In terms of energy, the effects of MPP+ in the CV transcripts reflect the funneling of carbon intermediates toward glycolysis. The LDH-A/B DKO transcripts reflect a flow of carbons away from glycolysis toward the production of acetyl-CoA. CONCLUSION The findings from this study suggest a metabolic resilience to MPP+ in cancer cells devoid of LDH-A/B, explainable in-part by higher baseline OXPHOS respiratory ATP production, necessitating more toxin to suppress the electron transport chain.
Collapse
Affiliation(s)
- Nzinga Mack
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Elizabeth Mazzio
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Ramesh Badisa
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F A Soliman
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
6
|
Qi Z, Wang Q, Wang H, Tan M. Metallothionein Attenuated Arsenic-Induced Cytotoxicity: The Underlying Mechanism Reflected by Metabolomics and Lipidomics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5372-5380. [PMID: 33939412 DOI: 10.1021/acs.jafc.1c00724] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Arsenic ions (As3+) have been recognized as a hazard that threatens the health of humans. Metallothionein (MT) rich in cysteine may provide favorable binding sites for chelation of As3+. However, the influence of MT on As3+-induced toxicity and the underlying mechanism are poorly understood, especially at the metabolic level. Herein, the effects of MT on As3+-induced toxicity were evaluated. Cell viability analysis suggested that MT alleviated As3+-induced cytotoxicity. The metabolic response of PC12 cells to As3+ investigated by lipidomics and metabolomics indicated that the presence of As3+ disrupted phospholipids metabolism and induced cell membrane damage. Moreover, energy and amino acid metabolism were perturbed by As3+. The perturbation of As3+ on metabolism was further illustrated by the decrease of the mitochondrial membrane potential and the rise of cellular reactive oxygen species (ROS). On the contrary, MT rescued As3+-induced metabolic disorder and suppressed ROS accumulation. In addition, the binding process between As3+ and MT was characterized. The results proved that the As3+-MT complex was formed and chelated As3+-scavenged ROS, thus alleviating the toxic effects of As3+. These results revealed that MT would be a potential agent to reduce As3+-induced cytotoxicity.
Collapse
Affiliation(s)
- Zihe Qi
- Academy of Food Interdisciplinary Sciecne, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan 1, Ganjingzi District, Dalian, 116034 Liaoning, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
| | - Qinghong Wang
- Academy of Food Interdisciplinary Sciecne, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan 1, Ganjingzi District, Dalian, 116034 Liaoning, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
| | - Haitao Wang
- Academy of Food Interdisciplinary Sciecne, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan 1, Ganjingzi District, Dalian, 116034 Liaoning, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Sciecne, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan 1, Ganjingzi District, Dalian, 116034 Liaoning, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 Liaoning, China
| |
Collapse
|
7
|
Zhang Y, Li J, Zhang X, Song D, Tian T. Advances of Mechanisms-Related Metabolomics in Parkinson's Disease. Front Neurosci 2021; 15:614251. [PMID: 33613180 PMCID: PMC7887307 DOI: 10.3389/fnins.2021.614251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial disorder characterized by progressively debilitating dopaminergic neurodegeneration in the substantia nigra and the striatum, along with various metabolic dysfunctions and molecular abnormalities. Metabolomics is an emerging study and has been demonstrated to play important roles in describing complex human diseases by integrating endogenous and exogenous sources of alterations. Recently, an increasing amount of research has shown that metabolomics profiling holds great promise in providing unique insights into molecular pathogenesis and could be helpful in identifying candidate biomarkers for clinical detection and therapies of PD. In this review, we briefly summarize recent findings and analyze the application of molecular metabolomics in familial and sporadic PD from genetic mutations, mitochondrial dysfunction, and dysbacteriosis. We also review metabolic biomarkers to assess the functional stage and improve therapeutic strategies to postpone or hinder the disease progression.
Collapse
Affiliation(s)
| | | | | | | | - Tian Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Amo T, Oji Y, Saiki S, Hattori N. Metabolomic analysis data of MPP +-exposed SH-SY5Y cells using CE-TOFMS. Data Brief 2021; 34:106707. [PMID: 33457479 PMCID: PMC7797368 DOI: 10.1016/j.dib.2020.106707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 11/15/2022] Open
Abstract
1-Methyl-4-phenylpyridinium (MPP+)-treated human neuroblastoma SH-SY5Y cells have been generally accepted as a cellular model for Parkinson's disease. This article contains metabolic analysis data of not only cell lysate but also culture supernatants to understand comprehensive metabolic disturbances in this model. Metabolic analysis employed by capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS). Data obtained by CE-TOFMS were processed to extract peak information including m/z, peak area, and migration time. The data provided in this manuscript have been analyzed and discussed in the research article entitled “Metabolomic analysis revealed mitochondrial dysfunction and aberrant choline metabolism in MPP+-exposed SH-SY5Y cells” [1].
Collapse
Affiliation(s)
- Taku Amo
- Department of Applied Chemistry, National Defense Academy, 1-10-20 Hashirimizu, Yokosuka 239-8686, Japan
| | - Yutaka Oji
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Corresponding authors.
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Corresponding authors.
| |
Collapse
|
9
|
Molecular Mechanism of Platelet-Derived Growth Factor (PDGF)-BB-Mediated Protection Against MPP+ Toxicity in SH-SY5Y Cells. J Mol Neurosci 2020; 71:1131-1143. [DOI: 10.1007/s12031-020-01735-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/02/2020] [Indexed: 12/21/2022]
|
10
|
Egunlusi AO, Malan SF, Omoruyi SI, Ekpo OE, Joubert J. 4-Oxatricyclo[5.2.1.0 2,6]dec-8-ene-3,5-dione Derivatives as NMDA Receptor- and VGCC Blockers with Neuroprotective Potential. Molecules 2020; 25:E4552. [PMID: 33027964 PMCID: PMC7582567 DOI: 10.3390/molecules25194552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 11/30/2022] Open
Abstract
The impact of excitotoxicity mediated by N-methyl-D-aspartate (NMDA) receptor overactivation and voltage gated calcium channel (VGCC) depolarization is prominent among the postulated processes involved in the development of neurodegenerative disorders. NGP1-01, a polycyclic amine, has been shown to be neuroprotective through modulation of the NMDA receptor and VGCC, and attenuation of MPP+-induced neurotoxicity. Recently, we reported on the calcium modulating effects of tricycloundecene derivatives, structurally similar to NGP1-01, on the NMDA receptor and VGCC of synaptoneurosomes. In the present study, we investigated novel 4-oxatricyclo[5.2.1.02,6]dec-8-ene-3,5-dione derivatives for their cytotoxicity, neuroprotective effects via attenuation of MPP+-induced neurotoxicity and calcium influx inhibition abilities through the NMDA receptor and VGCC using neuroblastoma SH-SY5Y cells. All compounds, in general, showed low or no toxicity against neuroblastoma cells at 10-50 µM concentrations. At 10 µM, all compounds significantly attenuated MPP+-induced neurotoxicity as evident by the enhancement in cell viability between 23.05 ± 3.45% to 53.56 ± 9.29%. In comparison to known active compounds, the derivatives demonstrated mono or dual calcium modulating effect on the NMDA receptor and/or VGCC. Molecular docking studies using the NMDA receptor protein structure indicated that the compounds are able to bind in a comparable manner to the crystallographic pose of MK-801 inside the NMDA ion channel. The biological characteristics, together with results from in silico studies, suggest that these compounds could act as neuroprotective agents for the purpose of halting or slowing down the degenerative processes in neuronal cells.
Collapse
Affiliation(s)
- Ayodeji O. Egunlusi
- Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (A.O.E.); (S.F.M.)
| | - Sarel F. Malan
- Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (A.O.E.); (S.F.M.)
| | - Sylvester I. Omoruyi
- Department of Medical Biosciences, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (S.I.O.); (O.E.E.)
| | - Okobi E. Ekpo
- Department of Medical Biosciences, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (S.I.O.); (O.E.E.)
| | - Jacques Joubert
- Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (A.O.E.); (S.F.M.)
| |
Collapse
|
11
|
Open and rearranged norbornane derived polycyclic cage molecules as potential neuroprotective agents through attenuation of MPP +- and calcium overload-induced excitotoxicity in neuroblastoma SH-SY5Y cells. Eur J Med Chem 2020; 204:112617. [PMID: 32717484 DOI: 10.1016/j.ejmech.2020.112617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
Abstract
The neuroprotective effects of closed polycyclic cage molecules such as NGP1-01, memantine and amantadine have been extensively explored. These effects are mostly linked to the antagonism of the N-methyl-d-aspartate (NMDA) receptor- and the blockage of voltage gated calcium channels (VGCC). The synthesis of structurally related open and rearranged cage derivatives has been studied in depth. However, very little is known on their neuroprotective effects. In this study, a series of open and rearranged polycyclic cage molecules containing a norbornane derived scaffold were synthesised and evaluated for cytotoxicity, neuroprotection and calcium blocking effects via the NMDA receptor and VGCC on neuroblastoma cells at a 10 μM concentration. All compounds showed negligible cytotoxicity and were able to significantly attenuate MPP+-induced neurotoxicity between 26.07 ± 12.50% to 48.42 ± 0.76%, with compound 14 showing the best neuroprotective effect. In comparison to known NMDA receptor antagonists, all compounds demonstrated moderate to excellent calcium blocking effects of 26.50 ± 2.28 to 72.95 ± 3.38%. Docking studies suggest that these compounds are able to show significant NMDA receptor channel blocking ability since they bind in a comparable manner to the crystallographic pose of MK-801 inside the NMDAR ion channel. Some compounds were also able to attenuate calcium influx through VGCC channels between 21.28 ± 3.69% to 50.34 ± 7.67%. Compound 4 and 15 showed the highest inhibition of calcium influx at the VGCC and NMDA receptor, respectively. The compounds exhibiting good cytotoxicity-, neuroprotective- and calcium blocking profiles could potentially act as neuroprotective agents to clinically benefit people suffering from neurodegenerative disorders.
Collapse
|
12
|
Chen T, Liu W, Xiong S, Li D, Fang S, Wu Z, Wang Q, Chen X. Nanoparticles Mediating the Sustained Puerarin Release Facilitate Improved Brain Delivery to Treat Parkinson's Disease. ACS APPLIED MATERIALS & INTERFACES 2019; 11:45276-45289. [PMID: 31638771 DOI: 10.1021/acsami.9b16047] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Recent work has highlighted the potential of puerarin (PU) as a valuable compound to treat Parkinson's disease (PD), but its undesirable water solubility and bioavailability have constrained its utility. In this study, we sought to develop nanoparticles (NPs) that could be used to encapsulate PU, thereby extending its in vivo half-life and improving its bioavailability and accumulation in the brain to treat the symptoms of PD. We prepared spherical NPs (88.36 ± 1.67 nm) from six-armed star-shaped poly(lactide-co-glycolide) (6-s-PLGA) NPs that were used to encapsulate PU (PU-NPs) with 89.52 ± 1.74% encapsulation efficiency, 42.97 ± 1.58% drug loading, and a 48 h sustained drug release. NP formation and drug loading were largely mediated by hydrophobic interactions, while changes in the external environment led these NPs to become increasingly hydrophilic, thereby leading to drug release. Relative to PU alone, PU-NPs exhibited significantly improved cellular internalization, permeation, and neuroprotective effects. Upon the basis of Förster resonance energy transfer (FRET) of NPs-administered zebrafish, we were able to determine that these NPs were rapidly absorbed into circulation whereupon they were able to access the brain. We further conducted oral PU-NPs administration to rats, revealing significant improvements in PU accumulation within the plasma and brain relative to rats administered free PU. In MPTP-mediated neurotoxicity in mice, we found that PU-NPs treatment improved disease-associated behavioral deficits and depletion of dopamine and its metabolites. These findings indicated that PU-NPs represent a potentially viable approach to enhancing PU oral absorption, thus improving its delivery to the brain wherein it can aid in the treatment of PD.
Collapse
Affiliation(s)
- Tongkai Chen
- Science and Technology Innovation Center , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
- Institute of Clinical Pharmacology , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
| | - Wei Liu
- Science and Technology Innovation Center , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
- Institute of Clinical Pharmacology , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
| | - Sha Xiong
- Science and Technology Innovation Center , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
- Institute of Clinical Pharmacology , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
| | - Dongli Li
- Science and Technology Innovation Center , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
- Institute of Clinical Pharmacology , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
| | - Shuhuan Fang
- Science and Technology Innovation Center , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
- Institute of Clinical Pharmacology , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education , Jiangxi University of Traditional Chinese Medicine , Nanchang 330004 , China
| | - Qi Wang
- Science and Technology Innovation Center , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
- Institute of Clinical Pharmacology , Guangzhou University of Chinese Medicine , Guangzhou 510405 , China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences , University of Macau , Macau 999078 , China
| |
Collapse
|