1
|
Ortasoz AM, Ozdemir E, Taskıran AS, Ozturk A. Sinapic acid alleviates glutamate-induced excitotoxicity by inhibiting neuroinflammation and endoplasmic reticulum stress pathway in C6 glioma cells. Toxicol In Vitro 2025; 103:105977. [PMID: 39615638 DOI: 10.1016/j.tiv.2024.105977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Sinapic acid (SA) is a polyphenol compound derived from hydroxycinnamic acid found in various foods such as cereals and vegetables and has antioxidant, anti-inflammatory and neuroprotective properties. However, its effects on glutamate-induced excitotoxicity, which is important in neurodegenerative diseases, have not been fully elucidated. This study aimed to investigate the effect of SA on glutamate excitotoxicity and the possible role of proinflammatory cytokines and the endoplasmic reticulum (ER) stress pathway. In the study, C6 rat glioma cell line was used and the cells were divided into 4 groups: control, glutamate, SA and glutamate+SA. Cells were treated with 10 mM glutamate for 24 h to induce excitotoxicity. Additionally, SA was applied to cells at concentrations of 12.5 to 100 μM to examine its effects on glutamate excitotoxicity. XTT test was used for cell viability, and apoptotic cells were determined by immunofluorescence and flow cytometry methods. Proinflammatory cytokines (tumor necrosis factor-alpha, TNF-α and interleukin-beta, IL-1β), ER stress markers (glucose regulatory protein 78, GRP78; C/EBP homologous protein, CHOP and activating transcription factor-4, ATF-4) and caspase-3 was used to measure ELISA method. Findings indicated that SA (50 μM) significantly increased cell viability against glutamate-induced excitotoxicity (p < 0.05). Also, SA caused a significant decrease in TNF-α, IL-1β, GRP78, CHOP, ATF-4 and caspase-3 levels in glutamate-treated cells (p < 0.05). Flow cytometry and immunofluorescence staining results showed that SA reduced apoptosis in C6 glioma cells. In conclusion, our findings suggested that SA attenuated glutamate-induced excitotoxicity by preventing apoptosis through reducing proinflammatory cytokines and ER stress protein levels.
Collapse
Affiliation(s)
- Ahmet Mahmut Ortasoz
- Sivas Cumhuriyet University, Medicine Faculty, Department of Physiology, Sivas, Turkey
| | - Ercan Ozdemir
- Sivas Cumhuriyet University, Medicine Faculty, Department of Physiology, Sivas, Turkey.
| | - Ahmet Sevki Taskıran
- Sivas Cumhuriyet University, Medicine Faculty, Department of Physiology, Sivas, Turkey
| | - Aysegul Ozturk
- Sivas Cumhuriyet University, Vocational School of Health Services, Deparment of Therapy and Rehabilitation, Sivas, Turkey
| |
Collapse
|
2
|
Tekin V, Altintas F, Oymak B, Unal EB, Tunc-Ata M, Elmas L, Kucukatay V. S-Sulfocysteine's toxic effects on HT-22 cells are not triggered by glutamate receptors, nor do they involve apoptotic or genotoxicity mechanisms. Cytotechnology 2025; 77:32. [PMID: 39749013 PMCID: PMC11688261 DOI: 10.1007/s10616-024-00697-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
S-Sulfocysteine (SSC) is a metabolite derived from the metabolism of sulfur-containing amino acids. It has been implicated in neurotoxicity observed in children with sulfite oxidase deficiency. The aim of our study was to confirm the neurotoxic effects of SSC using a mouse hippocampal cell line (HT-22) and to investigate the role of apoptosis in these effects, especially in terms of caspase-3 activation and genotoxicity. Based on the viability graph obtained following increasing concentrations of SSC, we determined the LC50 dose of SSC to be 125 µM by probit analysis. The cytotoxic effects of SSC were not reversed by glutamate receptor blocker administration. However, SSC treatment did not induce caspase-3 activation or induce DNA damage. Our results showed that SSC has a cytotoxic effect on neurons like glutamate, but glutamate receptor blockers reversed glutamate-induced toxicity, while these blockers did not protect neurons from SSC toxicity. The absence of caspase-3 activation and DNA fragmentation, which are indicative of apoptosis, in SSC-induced cell death suggests that alternative cell death pathways, such as necrosis and oxytosis may be implicated. Further research is necessary to fully elucidate SSC-induced cell death. The aim of our study was to confirm the neurotoxic effects of SSC using a mouse hippocampal cell line (HT-22) and to investigate the role of apoptosis in these effects, especially in terms of caspase-3 activation and genotoxicity.
Collapse
Affiliation(s)
- Volkan Tekin
- Department of Physiology, Gülhane Faculty of Medicine, University of Health Sciences, Ankara, Türkiye
| | - Fatih Altintas
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Burak Oymak
- Department of Physiology, Prof. Dr. Cemil Taşcıoğlu City Hospital, Istanbul, Türkiye
| | - Egem Burcu Unal
- Department of Physiology, Adana City Education and Research Hospital, Adana, Türkiye
| | - Melek Tunc-Ata
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Levent Elmas
- Department of Medical Biology, Faculty of Medicine, Bakırçay University, İzmir, Türkiye
| | - Vural Kucukatay
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| |
Collapse
|
3
|
do Rêgo AGDO, D'Amico F, D'Angelo V, Cardarelli S, Cutuli D, Decandia D, Landolfo E, Petrosini L, Pellegrini M, D'Amelio M, Mercuri NB, Giorgi M, Sancesario G. Haploinsufficiency of PDE2A causes in mice increased exploratory behavior associated with upregulation of neural nitric oxide synthase in the striatum. Neurobiol Dis 2025; 205:106781. [PMID: 39733958 DOI: 10.1016/j.nbd.2024.106781] [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: 07/30/2024] [Revised: 12/21/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024] Open
Abstract
Phosphodiesterase 2 A (PDE2A) function is stimulated by cGMP to catabolize cAMP. However, neurological and neurochemical effects of PDE2A deficiency are poorly understood. To address this gap, we studied behavioral characteristics and cerebral morpho-chemical changes of adult male heterozygous C57BL/6-PDE2A+/- (HET), and wild type C57BL/6-PDE2A+/+ (WT) mice. Behavioral functions of mice were evaluated by a wide test battery. HET mice exhibited greater tendency to explore novel environments in comparison to WT mice, but spatial working memory, anxiety, and sociability were similar in adult HET and WT mice. In HET mice, PDE2A mRNA, PDE2A protein expression, and cGMP hydrolyzing enzymatic activity were consistently reduced by about 50 %. Consequently, the cyclic nucleotide levels were significantly increased in HET mice, but unexpectedly the mean percentage variation was higher for cGMP equal to 153.23 %, and lower for cAMP equal to 16.41 %. Therefore, to try to explain the preponderant increase of cGMP to cAMP we evaluated other PDE enzymes functionally related to PDE2A. Surprisingly, results were quite contradictory: in HET mice protein levels of the other dual-specificity enzyme PDE3A and PDE10A were reduced, whereas the expressions of PDE5A and PDE9A that selectively hydrolyze cGMP were increased. Therefore, we investigated the involvement of neuronal nitric oxide synthase (nNOS) expression, as determinant of a possible increased synthesis of NO/cGMP signaling. Interestingly, in HET mice the expression level of brain nNOS, measured by western blot and immune-histochemistry was significantly increased, particularly in interneurons from the striatum. In conclusion, the deficiency of PDE2A could be compensated in the striatum by upregulating nNOS/NO/cGMP pathway, which in turn likely upregulates PDE2A-dependent cAMP hydrolysis. The neuroanatomical correlation between striatal nNOS upregulation and the behavioral phenotype of increased exploratory behavior in HET mice is advanced.
Collapse
Affiliation(s)
| | - Francesca D'Amico
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, 00185 Rome, Italy
| | - Vincenza D'Angelo
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Silvia Cardarelli
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, 00185 Rome, Italy
| | - Debora Cutuli
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy; Department of Psychology, Sapienza University, 00185 Rome, Italy
| | - Davide Decandia
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy; Department of Psychology, Sapienza University, 00185 Rome, Italy
| | | | | | - Manuela Pellegrini
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015 Monterotondo Scalo, Rome, Italy
| | - Marcello D'Amelio
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy; Department of Medicine, Campus Biomedico University, 00128 Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Mauro Giorgi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, 00185 Rome, Italy.
| | | |
Collapse
|
4
|
Feng Y, Wang H, Hu Y, Zhang X, Miao X, Li Z, Jia J. Hederagenin ameliorates ferroptosis-induced damage by regulating PPARα/Nrf2/GPX4 signaling pathway in HT22 cells: An in vitro and in silico study. Bioorg Chem 2025; 155:108119. [PMID: 39755103 DOI: 10.1016/j.bioorg.2024.108119] [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: 11/06/2024] [Revised: 12/16/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Hederagenin (HG), derived from ivy seeds, is known to offer protection against Alzheimer's disease (AD). However, the specific molecular pathways through which it counters ferroptosis-induced neurotoxicity are not fully elucidated. This investigation seeks to delineate the processes by which HG mitigates neurotoxic effects in HT22 cells subjected to glutamate (Glu)-induced ferroptosis. METHODS HT22 cell ferroptosis was prompted by Glu exposure. Cell viability was assessed using CCK-8 and LDH assays, while Fe2+ fluorescence and assays of iron-related proteins served to gauge intracellular Fe2+ concentrations. Evaluations of mitochondrial structure and functionality employed JC-1 staining and transmission electron microscopy. Assessments of ROS, lipid peroxidation, MDA, 4-HNE, and the GSSG/GSH ratio were conducted to ascertain HG's antioxidative efficacy. The expression of proteins within the PPARα/Nrf2/GPX4 pathway was quantified via western blotting, with molecular docking (MD), and molecular dynamics simulations (MDS) used to explore protein interactions. RESULTS HG diminished the cellular toxicity triggered by Glu in HT22 cells, lowered Fe2+ within cells, and rejuvenated mitochondrial morphology and performance. Concurrently, it modulated proteins critical to Fe2+ metabolism, diminished ROS and lipid peroxidation, and elevated GSH/GSSG ratios. Enhanced PPARα/Nrf2/GPX4 protein levels were corroborated by western blot results. Furthermore, molecular docking revealed favorable binding of HG to the proteins PPARα, Nrf2, and GPX4, with binding energies of -7.751, -7.535, and -7.414 kcal/mol, respectively. MDS confirmed robust interactions between HG and these pivotal targets. CONCLUSION The evidence suggests that HG effectively mitigates Glu-induced ferroptosis in HT22 cells by activating the PPARα/Nrf2/GPX4 signaling pathway. These findings endorse HG's potential as a nutritional adjunct for AD management.
Collapse
Affiliation(s)
- Yuxin Feng
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - Heran Wang
- Institute of Geriatrics, The 2nd Medical Center, China National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yazhuo Hu
- Institute of Geriatrics, The 2nd Medical Center, China National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoxue Zhang
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - XiuLing Miao
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - Zihan Li
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - JianJun Jia
- Institute of Geriatrics, The 2nd Medical Center, China National Clinical Research Center for Geriatric Disease, Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
5
|
Sedlacek J, Smahelova Z, Adamek M, Subova D, Svobodova L, Kadlecova A, Majer P, Machara A, Grantz Saskova K. Small-molecule activators of NRF1 transcriptional activity prevent protein aggregation. Biomed Pharmacother 2025; 183:117864. [PMID: 39884031 DOI: 10.1016/j.biopha.2025.117864] [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: 10/31/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/01/2025] Open
Abstract
Intracellular protein aggregation causes proteotoxic stress, underlying highly debilitating neurodegenerative disorders in parallel with decreased proteasome activity. Nevertheless, under such stress conditions, the expression of proteasome subunits is upregulated by Nuclear Factor Erythroid 2-related factor 1 (NRF1), a transcription factor that is encoded by NFE2L1. Activating the NRF1 pathway could accordingly delay the onset of neurodegenerative and other disorders with impaired cell proteostasis. Here, we present a series of small-molecule compounds based on bis(phenylmethylen)cycloalkanones and their heterocyclic analogues, identified via targeted library screening, that can induce NRF1-dependent downstream events, such as proteasome synthesis, heat shock response, and autophagy, in both model cell lines and Caenorhabditis elegans strains. These compounds increase proteasome activity and decrease the size and number of protein aggregates without causing any cellular stress or inhibiting the ubiquitin-proteasome system (UPS). Therefore, our compounds represent a new promising therapeutic approach for various protein conformational diseases, including the most debilitating neurodegenerative diseases.
Collapse
Affiliation(s)
- Jindrich Sedlacek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic; Department of Genetics and Microbiology, Charles University and Research Center BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Zuzana Smahelova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic; Department of Genetics and Microbiology, Charles University and Research Center BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Michael Adamek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic; Department of Genetics and Microbiology, Charles University and Research Center BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Dominika Subova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic; Department of Genetics and Microbiology, Charles University and Research Center BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic; First Faculty of Medicine & General University Hospital, Charles University, U Nemocnice 2, Prague 2 12808, Czech Republic
| | - Lucie Svobodova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic; Department of Organic Chemistry, Charles University, Hlavova 2030/8, Prague 2 12843, Czech Republic
| | - Alena Kadlecova
- Department of Experimental Biology, Palacky University, Slechtitelu 27, Olomouc 78371, Czech Republic
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic
| | - Ales Machara
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic.
| | - Klara Grantz Saskova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, Prague 16610, Czech Republic; Department of Genetics and Microbiology, Charles University and Research Center BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic.
| |
Collapse
|
6
|
Zhang L, Yan H, Rahman MS, Mol-van der Veen CHJ, Covarrubias AM, Rafie K, Pendin D, Schmidt M, Dolga AM. Regulation of calcium signaling prevents neuronal death mediated by NIST DEP in xenoferroptotic cell death conditions. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137374. [PMID: 39892142 DOI: 10.1016/j.jhazmat.2025.137374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/03/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Increased calcium levels are associated with the ferroptosis pathway in neurodegenerative conditions. Recent evidence showed that exposure to particulate matter (PM) could accelerate the pathology of neurodegenerative diseases. However, the molecular mechanisms of how PM could affect brain cell pathology is not fully understood. We hypothesized that diesel exhaust particles (NIST DEP) could alter the ferroptosis pathway through calcium signaling, and therefore accelerate the cell death pathway. In this study, we used mouse hippocampal neuronal-like HT22 cells to evaluate whether exposure to NIST DEP could accelerate RSL-3-induced ferroptosis by increasing calcium deregulation, mitochondrial dysfunction and reactive oxygen species (ROS). MTT assay results showed that NIST DEP (25, 50, 75, and 100 μg/mL) did not significantly reduce the survival rate of HT22 cells, while NIST DEP significantly increased RSL-3-induced ferroptotic cell death in a concentration-dependent manner. Based on fluorescence image analysis, co-exposure to NIST DEP and RSL-3 disrupted HT22 cell mitochondrial morphology, intracellular and mitochondrial calcium levels. Combined exposure resulted in an increase in ER-mitochondria contact sites measured by proximity ligation assay (PLA) compared to control solvent group. Additionally, lipid peroxidation, mitochondrial ROS and malondialdehyde content, were increased significantly by combined exposure to NIST DEP and RSL-3. Interestingly, the calcium regulators of the mitochondrial calcium uniporter MCUi4 and positive modulation of small conductance calcium-activated potassium channels by CyPPA significantly preserved cellular metabolic activity, restored calcium homeostasis, and alleviated fragmentation of mitochondria. Consequently, targeting calcium signaling may be promising therapeutic option for xenoferroptotic conditions in which PM affect cell survival.
Collapse
Affiliation(s)
- Leshan Zhang
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Mohammad Saidur Rahman
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Christina Ht J Mol-van der Veen
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Ana Manzano Covarrubias
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Karim Rafie
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Diana Pendin
- Neuroscience Institute, Padua Section, National Research Council, Padua, Italy; Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Instiute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands; Depatment of Pathology and Medical Biology, University Medical Center of Groningen, Groningen, The Netherlands.
| |
Collapse
|
7
|
Banoei MM, Hutchison J, Panenka W, Wong A, Wishart DS, Winston BW. Metabolomic in severe traumatic brain injury: exploring primary, secondary injuries, diagnosis, and severity. Crit Care 2025; 29:26. [PMID: 39815318 PMCID: PMC11737060 DOI: 10.1186/s13054-025-05258-1] [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: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major public health concern worldwide, contributing to high rates of injury-related death and disability. Severe traumatic brain injury (sTBI), although it accounts for only 10% of all TBI cases, results in a mortality rate of 30-40% and a significant burden of disability in those that survive. This study explored the potential of metabolomics in the diagnosis of sTBI and explored the potential of metabolomics to examine probable primary and secondary brain injury in sTBI. METHODS Serum samples from 59 adult patients with sTBI and 35 age- and sex-matched orthopedic injury controls were subjected to quantitative metabolomics, including proton nuclear magnetic resonance (1H-NMR) and direct infusion/liquid chromatography-tandem mass spectrometry (DI/LC-MS/MS), to identify and quantify metabolites on days 1 and 4 post-injury. In addition, we used advanced analytical methods to discover metabo-patterns associated with sTBI diagnosis and those related to probable primary and secondary brain injury. RESULTS Our results showed different serum metabolic profiles between sTBI and orthopedic injury (OI) controls, with significant changes in measured metabolites on day 1 and day 4 post-brain injury. The number of altered metabolites and the extent of their change were more pronounced on day 4 as compared to day 1 post-injury, suggesting an evolution of mechanisms from primary to secondary brain injury. Data showed high sensitivity and specificity in separating sTBI from OI controls for diagnosis. Energy-related metabolites such as glucose, pyruvate, lactate, mannose, and polyamine metabolism metabolites (spermine and putrescine), as well as increased acylcarnitines and sphingomyelins, occurred mainly on day 1 post-injury. Metabolites of neurotransmission, catecholamine, and excitotoxicity mechanisms such as glutamate, phenylalanine, tyrosine, and branched-chain amino acids (BCAAs) increased to a greater degree on day 4. Further, there was an association of multiple metabolites, including acylcarnitines (ACs), lysophosphatidylcholines (LysoPCs), glutamate, and phenylalanine, with injury severity at day 4, while lactate, glucose, and pyruvate correlated with injury severity on day 1. CONCLUSION The results demonstrate that serum metabolomics has diagnostic potential for sTBI and may reflect molecular mechanisms of primary and secondary brain injuries when comparing metabolite profiles between day 1 and day 4 post-injury. These early changes in serum metabolites may provide insight into molecular pathways or mechanisms of primary injury and ongoing secondary injuries, revealing potential therapeutic targets for sTBI. This work also highlights the need for further research and validation of sTBI metabolite biomarkers in a larger cohort.
Collapse
Affiliation(s)
- Mohammad M Banoei
- Department of Critical Care Medicine, Cumming School of Medicine, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - James Hutchison
- Department of Critical Care and Neuroscience and Mental Health Research Program, The Hospital for Sick Children and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - William Panenka
- Department of Psychiatry, Faculty of Medicine, University of British Colombia, Vancouver, BC, Canada
| | - Andy Wong
- Department of Critical Care Medicine, Cumming School of Medicine, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - David S Wishart
- Departments of Biological Sciences, Computing Sciences and Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Brent W Winston
- Department of Critical Care Medicine, Cumming School of Medicine, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada.
- Dr. Brent W. Winston, Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
8
|
Ahn H, Lee H, Choi W, Lee H, Lee KG, Youn I, Hur W, Han S, Song C. Discovery of the therapeutic potential of naltriben against glutamate-induced neurotoxicity. Neurochem Int 2025; 183:105928. [PMID: 39756586 DOI: 10.1016/j.neuint.2025.105928] [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: 10/11/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Glutamate-induced neuronal death is associated with neurodegeneration including cerebral ischemia. Several μ-opioid receptor antagonists exhibit a neuroprotective activity and have been considered as a potential therapeutic option for neurodegenerative disorders. For the first time, our current study unveiled the neuroprotective activity of selective δ-opioid receptor antagonists. A potent, selective δ-opioid receptor antagonist naltriben, also known as a potent TRPM7 agonist, displayed the prominent protective effect against glutamate-induced toxicity through opioid receptor-independent, TRPM7-independent mechanisms in HT22 cells. Naltriben activated Nrf2 pathway, and alleviated glutamate-induced Ca2+ influx, ROS production, and apoptosis. Moreover, intraperitoneal administration of naltriben at 20 mg/kg greatly reduced the infarct volume in the subcortical photothrombotic ischemia mouse model in vivo. The neuroprotective activity of naltriben was enhanced by a longer pretreatment, indicating that like Nrf2 activators, naltriben also requires the cellular priming for its full protective effects. Together, these results suggested naltriben as a potential therapeutic agent in conditions related with glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- Hyomin Ahn
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Life Sciences, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hyomin Lee
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wonseok Choi
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyebin Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Pharmacology, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Kang-Gon Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Inchan Youn
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wooyoung Hur
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; HY-KIST Bioconvergence, Hanyang University, 222 Wangsimniro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Sungmin Han
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; KHU-KIST, Department of Converging Science and Technology, Kyung Hee University, 26 Kyungheedaero, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Chiman Song
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
9
|
Oruç KY, Ağtürk G, Oruç A, Yanar K, Seymen HO. Protective effect of Apelin-13 on D-glutamic acid-induced excitotoxicity in SH-SY5Y cell line: An in-vitro study. Neuropeptides 2025; 109:102483. [PMID: 39547009 DOI: 10.1016/j.npep.2024.102483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/08/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024]
Abstract
Excitotoxicity, resulting from excessive accumulation of glutamate in the extracellular space, leads to neuronal cell death. This study investigates the protective effects of Apelin-13 on D-Glutamic acid-induced excitotoxicity in SH-SY5Y human neuroblastoma cells, an in-vitro model for neurodegenerative diseases. Unlike the commonly studied L-glutamic acid, this research focuses on D-Glutamic acid to understand its specific impacts. SH-SY5Y cells were treated with varying concentrations of D-Glutamic acid and Apelin-13, followed by analyses at 12 and 24 h to evaluate cell viability, oxidative stress markers, and inflammatory cytokine levels. Cell viability assays revealed significant cytotoxic effects of D-Glutamic acid at doses of 10 mM and 20 mM, reducing viability by over 50 %. However, Apelin-13 treatment mitigated these effects, especially at 2 μg/ml, enhancing cell viability and reducing inflammatory cytokine levels (IL-1β and TNF-α). Apelin-13 also increased anti-inflammatory cytokine levels (IL-10 and TGF-β1) and brain-derived neurotrophic factor (BDNF), indicating its neuroprotective role. Oxidative stress markers, including ROS, AGE, AOPP, DT, T-SH, were significantly elevated by D-Glutamic acid but effectively reduced by Apelin-13. The neuroprotective mechanisms of Apelin-13 involve modulation of cAMP/PKA and MAPK signaling pathways, enhancing BDNF synthesis and suppressing oxidative stress and inflammatory responses. This study is the first to demonstrate the effects of D-Glutamic acid on SH-SY5Y cells. It highlights Apelin-13's potential as a therapeutic agent against excitotoxicity-induced neuronal damage, emphasizing its ability to modulate key molecular pathways involved in inflammation and oxidative stress. Further in-vivo studies are warranted to explore the long-term neuroprotective effects of Apelin-13 in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Kadriye Yağmur Oruç
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Physiology, Istanbul, Turkey; Istinye University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey.
| | - Gökhan Ağtürk
- Haliç University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| | - Aykut Oruç
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| | - Karolin Yanar
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Hakkı Oktay Seymen
- Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| |
Collapse
|
10
|
Xie Y, Zhang L, Chen S, Xie C, Tong J, Shen Y. The potential role of amino acids in myopia: inspiration from metabolomics. Metabolomics 2024; 21:6. [PMID: 39676079 DOI: 10.1007/s11306-024-02207-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Due to the high prevalence of myopia, there is a growing need for the identification of myopia intervention mechanisms and targets. Metabolomics has been gradually used to investigate changes in myopia tissue metabolites over the last few years, but the potential physiological and pathological roles of amino acids and their downstream metabolites discovered by metabolomics in myopia are not fully understood. AIM OF REVIEW Aim to explore the possible relationship between amino acid metabolism and the occurrence and development of myopia, we collected a total of 21 experimental studies related to myopia metabolomics. Perform pathway analysis using MetaboAnalyst online software. We have identified over 20 amino acids that may be associated with the development of myopia. Among them, 19 types of amino acids are common amino acids. We discussed their possible mechanisms affecting myopia and proposed future prospects for treating myopia. KEY SCIENTIFIC CONCEPTS OF REVIEW Our analysis results show that metabolomics research on myopia involves many important amino acids. We have collected literature and found that research on amino acid metabolism in myopia mainly focuses on downstream small molecule substances. Amino acids and their downstream metabolites affect the development of myopia by participating in important biochemical processes such as oxidative stress, glucose metabolism, and lipid metabolism. Enzymes, receptors, and cytokines that regulate amino acid metabolism may become potential targets for myopia treatment.
Collapse
Affiliation(s)
- Ying Xie
- The Department of Ophthalmology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Liyue Zhang
- The Department of Ophthalmology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Siyi Chen
- The Alfred, 55 Commercial Rd, Melbourne, VIC, Australia
| | - Chen Xie
- The Department of Ophthalmology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianping Tong
- The Department of Ophthalmology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| | - Ye Shen
- The Department of Ophthalmology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
11
|
Seo CR, Lee BK, Jee HJ, Yoo JR, Lee CK, Park JW, Jung YS. Ameliorating Effect of Fermented Perilla frutescens on Sleep Deprivation-Induced Cognitive Impairment Through Antioxidant and BDNF Signaling in Mice. Nutrients 2024; 16:4224. [PMID: 39683616 DOI: 10.3390/nu16234224] [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: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Adequate sleep is essential for maintaining cognitive function, as evidenced by literature. Perilla frutescens var. acuta Kudo (PF) is a traditional medicinal herb reported to improve vascular cognitive impairment and induce sedation. However, the effects of PF on cognitive impairment caused by sleep deprivation (SD) have not yet been evaluated. This study aims to evaluate the effects of fermented PF (FPF) and its underlying mechanisms in a model of SD-induced cognitive impairment. Methods: Mice were subjected to SD to establish cognitive impairment, and FPF was administered once daily for 3 days. Cognitive performance was assessed using Y-maze and passive avoidance tests, followed by molecular mechanisms analyses. Results: FPF treatment improved SD-induced cognitive impairment, as evidenced by increased spontaneous alternation and extended latency time. Histological analysis revealed that SD impaired the hippocampus, and this impairment was alleviated by FPF treatment. FPF demonstrated antioxidant activity by increasing glutathione levels and decreasing malondialdehyde levels. Furthermore, the decreased levels of brain-derived neurotrophic factor (BDNF) observed in sleep-deprived mice were restored with FPF treatment. FPF also enhanced the phosphorylation of tropomyosin receptor kinase B, extracellular signal-regulated kinase, and cAMP response element-binding protein. Conclusions: These results indicate that FPF may have beneficial effects on SD-induced cognitive impairment by protecting against oxidative stress and increasing BDNF expression.
Collapse
Affiliation(s)
- Chae-Ryeong Seo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Bo Kyung Lee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hye Jin Jee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Jae Ryeong Yoo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Chul-Kyu Lee
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Jin Wook Park
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Yi-Sook Jung
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
12
|
Bai L, Li F. To explore the protective mechanism of promethazine against hippocampal neuron injury based on network pharmacology and experimental verification. Medicine (Baltimore) 2024; 103:e40550. [PMID: 39654167 PMCID: PMC11631019 DOI: 10.1097/md.0000000000040550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/16/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
This study aims to investigate the effect of promethazine (PMZ) on hippocampal neuronal injury through network pharmacology and in vivo experiments. Network pharmacology: The intersection genes of PMZ and Alzheimer Disease (AD) were obtained, and the core genes of PMZ in AD were screened. The intersection genes were enriched by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. In the in vitro experiment, mouse hippocampal neurons (HT22) were divided into control, glutamate (GLU) model, and GLU + PMZ treatment groups. The control group was given a complete culture medium, the model group was given GLU for 24 hours, the treatment group was given PMZ pretreatment for 3 hours, and then GLU was administered for 24 hours. Cell viability was determined, cell morphology was observed by microscopy, reactive oxygen species levels and glutathione content were detected, and protein expression of P53, PTGS2, SLC7A11, and GPX4 was detected by western blotting. Network pharmacology: A total of 317 PMZ targets, 1934 AD genes, 125 intersection genes, and 18 core genes, including P53 and PTGS2. Gene Ontology enrichment analysis showed that the effect of PMZ on AD was mainly related to cell proliferation, inflammation, hypoxia, synaptic structure, plasma membrane, and oxidoreductase activity. Kyoto Encyclopedia of Genes and Genomes results showed neuroactive ligand-receptor interaction, cell senescence, cancer pathway, PI3K-AKT signal pathway, neurodegeneration, and HIF-1 signal pathway. In vitro experiments: PMZ improved the GLU-induced decrease in cell viability and morphological changes in hippocampal neurons. PMZ inhibited reactive oxygen species levels and increased glutathione content in injured hippocampal neurons. Up-regulated of P53, SLC7A11 and GPX4 expression, and inhibited expression of PTGS2. PMZ regulates the SLC7A11-GPX4 antioxidant system to protect hippocampal neurons from oxidative stress injury.
Collapse
Affiliation(s)
- Li Bai
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
- Bazhong Central Hospital, Bazhong, China
| | - Fang Li
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
13
|
Yuan R, Gao D, Yang G, Zhuoma D, Pu Z, Ciren Y, Li B, Yu J. Oxysophocarpine Prevents the Glutamate-Induced Apoptosis of HT-22 Cells via the Nrf2/HO-1 Signaling Pathway. Curr Issues Mol Biol 2024; 46:13035-13049. [PMID: 39590371 PMCID: PMC11593028 DOI: 10.3390/cimb46110777] [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: 10/25/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Oxysophocarpine (OSC), a quinolizidine alkaloid, shows neuroprotective potential, though its mechanisms are unclear. The aim of the present study was to investigate the neuroprotective effects of OSC through the nuclear factor erythroid 2-related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathway using the HT-22 cell line. Assessments of cell viability were conducted utilizing the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Assessments of oxidative stress (OS) were conducted through the quantification of reactive oxygen species (ROS). The integrity of the mitochondrial membrane potential (MMP) was scrutinized using fluorescent probe technology. Apoptosis levels were quantified using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The trafficking of Nrf2 within the cell nucleus was examined through immunofluorescence analysis. Furthermore, Western blotting (WB) was applied to evaluate the expression levels of proteins implicated in apoptosis and the Nrf2/HO-1 pathway. To further probe the influence of OSC on the overexpression of antioxidant enzymes, cells were subjected to transfection with HO-1 siRNA. The results showed that OSC inhibited glutamate-induced OS, as evidenced by reduced cell viability and ROS levels. Furthermore, the apoptotic condition induced by glutamate in HT-22 cells was significantly reduced following OSC treatment. More interestingly, the Nrf2/HO-1 signaling pathway was upregulated following OSC treatment. These results suggest that OSC can exert neuroprotective effects by regulating the Nrf2/HO-1 pathway to inhibit neuronal cell apoptosis, potentially aiding in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruiying Yuan
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
- College of Pharmacy, Wuhan University, Wuhan 430000, China;
| | - Dan Gao
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Guibing Yang
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Dongzhi Zhuoma
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Zhen Pu
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Yangzhen Ciren
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Bin Li
- Department of Pharmaceutical Sciences, School of Medicine, Tibet University, Lhasa 850000, China; (R.Y.); (D.G.); (G.Y.); (D.Z.); (Z.P.); (Y.C.)
| | - Jianqing Yu
- College of Pharmacy, Wuhan University, Wuhan 430000, China;
| |
Collapse
|
14
|
Iglesias Pastrana C, Sgobba MN, Navas González FJ, Delgado Bermejo JV, Pierri CL, Lentini G, Musio B, Osman TKS, Gallo V, Duarte IF, Guerra L, Ciani E. Factors influencing the bioactivity of natural matrices: The case of osmolarity-dependent modulation of cell viability by different dilutions of camel urines. Res Vet Sci 2024; 180:105419. [PMID: 39341022 DOI: 10.1016/j.rvsc.2024.105419] [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: 03/22/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
The widespread practice of dromedary urinotherapy as a remedy for various illnesses, including cancer, is well-established in traditional dromedary countries. Researchers attempted to demonstrate anticancer properties of camel urine through in vitro experiments with debated outcomes. Notably, two critical aspects remained unexplored in those assays: (i) the osmolarity of tested urines, which can significantly influence in vitro results; (ii) the potential morphological changes of cells, following exposure to camel urines. In this study, we addressed these gaps by evaluating the osmolarity-dependent modulation of cell viability in human renal cell lines. In this regard, we assessed the impact of hyperosmolar mannitol-based solutions and dromedary urine on the viability and morphology of human non-tumor (HK2) and tumor renal cells (Caki-1). The results indicate that cell viability or morphology in both HK2 and Caki-1 cells are not significantly affected only if mannitol-induced hyperosmolarity is lower than 500 mOsm/L. Notably, when exposed to urine solution, diluted to <500 mOsm/L, statistically significant antiproliferative effects were observed primarily in Caki-1 cells (in presence of two out of ten tested urine samples). Conversely, alterations in cell morphology were observed exclusively in HK2 cells when exposed to the same diluted camel urines. In order to investigate, at molecular level, the observed antiproliferative effects, a preliminary metabolomics analysis of the tested urine samples was performed to identify potential bioactive compounds. The Nuclear Magnetic Resonance (NMR) metabolic profiling revealed the presence of three antioxidant compounds, namely trigonelline, pyruvic acid and N-acetylglucosamine. In conclusion, our results highlight the importance of considering the critical role of osmolarity when evaluating the bioactive properties of camel urine in vitro, which should not be used to treat any illness as it is. Conversely, it can be considered the possibility to use camel urines as a source of bioactive compounds.
Collapse
Affiliation(s)
- Carlos Iglesias Pastrana
- Faculty of Veterinary Sciences, Department of Genetics, University of Córdoba, 14071 Córdoba, Spain
| | - Maria Noemi Sgobba
- Department of Biosciences, Biotechnologies and Environment, University of Bari 'Aldo Moro', 70125 Bari, Italy
| | | | | | - Ciro Leonardo Pierri
- Department of Pharmacy- Pharmaceutical Sciences, University of Bari 'Aldo Moro', 70125 Bari, Italy.
| | - Giovanni Lentini
- Department of Pharmacy- Pharmaceutical Sciences, University of Bari 'Aldo Moro', 70125 Bari, Italy
| | - Biagia Musio
- Department of Civil, Environmental, Land, Construction Engineering and Chemistry (DICATECh), Polytechnic University of Bari, 70125 Bari, Italy
| | | | - Vito Gallo
- Department of Civil, Environmental, Land, Construction Engineering and Chemistry (DICATECh), Polytechnic University of Bari, 70125 Bari, Italy; Innovative Solutions S.r.l, Spin Off Company at Polytechnic University of Bari, 70015 Noci (BA), Italy
| | - Iola F Duarte
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Environment, University of Bari 'Aldo Moro', 70125 Bari, Italy.
| | - Elena Ciani
- Department of Biosciences, Biotechnologies and Environment, University of Bari 'Aldo Moro', 70125 Bari, Italy
| |
Collapse
|
15
|
Moharram FA, Hamed FM, El-Sayed EK, Mohamed SK, Ahmed AA, Elgayed SH, Abdelrazek M, Lai KH, Mansour YE, Mady MS, Elsayed HE. Chemical characterization, neuroprotective effect, and in-silico evaluation of the petroleum ether extract of three palm tree species against glutamate-induced excitotoxicity in rats. Heliyon 2024; 10:e39207. [PMID: 39640788 PMCID: PMC11620252 DOI: 10.1016/j.heliyon.2024.e39207] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
The burden of neurological disorders is growing substantially with limited therapeutic options, urging the consideration and assessment of alternative strategies. In this regard, we aimed to elucidate the phytochemical profile of the petroleum ether extract (PEE) of three palm tree species: Aiphanes eggersii Burret, Carpoxylon macrospermum H. Wendl. & Drude, and Jubaeopsis caffra Becc. (Family Arecaceae), and to evaluate their neuroprotective effect in monosodium glutamate (MSG)-induced excitotoxicity model for the first time. We identified a total of 48, 18, and 45 compounds in A. eggersii, C. macrospermum, and J. caffra, constituting 79.41 %, 60.45 %, and 76.35 % of the total detected compounds, respectively. A. eggersii extract was rich in the methyl esters of fatty acids (65.08 %) especially methyl dodecanoate (17.72 %). C. macrospermum was exclusively prolific by the triterpene 3β-methoxy-d:c-friedo-b':a'-neogammacer-9(11)-ene (40.36 %), while J. caffra was noticeable by hydrocarbons (30.14 %) and lupeol derivatives (19.79 %). The biochemical and histopathological analysis showed that the tested extracts significantly reduced the oxidative stress, especially at the highest tested dose (1000 mg/kg). The extracts also reduced the activity of induced nitric oxide synthetase, Ca+2 level, and NR2B subunit expression and attenuated apoptosis and DNA damage. The docking results show that most active natural compounds bind to SOD-1 and NR2B-NMDARs, verifying the credibility of the biological findings. To sum up, the PEE of the three investigated palm tree species possessed a unique blend of lipophilic bioactive constituents that exert promising neuroprotective potential against MSG-induced excitoneurotoxicity. However, further preclinical investigation and pharmaceutical formulation are needed.
Collapse
Affiliation(s)
- Fatma A. Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| | - Fadila M. Hamed
- Department of Pharmacognosy, Faculty of Pharmacy, October 6 University, 6th of October City, Giza, 12585, Egypt
| | - Elsayed K. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| | - Shimaa K. Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| | - Asmaa A. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| | - Sabah H. Elgayed
- Department of Pharmacognosy, Faculty of Pharmacy, October 6 University, 6th of October City, Giza, 12585, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mohammed Abdelrazek
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Yara E. Mansour
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| | - Mohamed S. Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| | - Heba E. Elsayed
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, 11795, Egypt
| |
Collapse
|
16
|
Zhang G, Liang Z, Wang Y, Zhang Z, Hoi PM. Tetramethylpyrazine Analogue T-006 Protects Neuronal and Endothelial Cells Against Oxidative Stress via PI3K/AKT/mTOR and Nrf2 Signaling. Antioxidants (Basel) 2024; 13:1272. [PMID: 39456524 PMCID: PMC11505549 DOI: 10.3390/antiox13101272] [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: 09/23/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND T-006, a novel neuroprotective derivative of tetramethylpyrazine (TMP), exhibits multifunctional neuroprotective properties. T-006 has been shown to improve neurological and behavioral functions in animal models of ischemic stroke and neurodegenerative diseases. The present study aims to further elucidate the mechanisms underlying the protective effects of T-006 against oxidative injuries induced by glutamate or hypoxia. METHODS Mouse hippocampal HT22 cells were used to evaluate the neuroprotective effects of T-006 against glutamate-induced injuries, while mouse brain endothelial bEnd.3 cells were used to evaluate the cerebrovascular protective effects of T-006 against oxygen-glucose deprivation followed by reperfusion (OGD/R)-induced injuries. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry were used to measure cell viability and oxidative stress. Western blot and immunofluorescence analyses of protein expression were used to study cell signaling pathways. RESULTS T-006 exhibited significant protective effects in both oxidative injury models. In HT22 cells, T-006 reduced cell death and enhanced antioxidant capacity by upregulating mTOR and nuclear factor erythroid 2-related factor 2/Heme oxygenase-1 (Nrf2/HO-1) signaling. Similarly, in bEnd.3 cells, T-006 reduced oxidative injuries and preserved tight junction integrity through Nrf2/HO-1 upregulation. These effects were inhibited by LY294002, a Phosphoinositide 3-kinase (PI3K) inhibitor. CONCLUSIONS T-006 may exert its neuroprotective and cerebrovascular protective effects via the regulation of PI3K/AKT-mediated pathways, which facilitate downstream mTOR and Nrf2 signaling, leading to improved cell survival and antioxidant defenses.
Collapse
Affiliation(s)
- Guiliang Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; (G.Z.); (Z.L.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Zirong Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; (G.Z.); (Z.L.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Yuqiang Wang
- Institute of New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University College of Pharmacy, Guangzhou 510632, China; (Y.W.); (Z.Z.)
- Guangdong-Hong Kong-Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University College of Pharmacy, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Zaijun Zhang
- Institute of New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University College of Pharmacy, Guangzhou 510632, China; (Y.W.); (Z.Z.)
- Guangdong-Hong Kong-Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University College of Pharmacy, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Pui-Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; (G.Z.); (Z.L.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, China
| |
Collapse
|
17
|
Poejo J, Gumerova NI, Rompel A, Mata AM, Aureliano M, Gutierrez-Merino C. Unveiling the agonistic properties of Preyssler-type Polyoxotungstates on purinergic P2 receptors. J Inorg Biochem 2024; 259:112640. [PMID: 38968927 DOI: 10.1016/j.jinorgbio.2024.112640] [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: 03/18/2024] [Revised: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 07/07/2024]
Abstract
The Preyssler-type polyoxotungstate ({P5W30}) belongs to the family of polyanionic metal-oxides formed by group V and VI metal ions, such as V, Mo and W, commonly known as polyoxometalates (POMs). POMs have demonstrated inhibitory effect on a significant number of ATP-binding proteins in vitro. Purinergic P2 receptors, widely expressed in eukaryotic cells, contain extracellularly oriented ATP-binding sites and play many biological roles with health implications. In this work, we use the immortalized mouse hippocampal neuronal HT-22 cells in culture to study the effects of {P5W30} on the cytosolic Ca2+ concentration. Changes in cytosolic Ca2+ concentration were monitored using fluorescence microscopy of HT-22 cells loaded with the fluorescent Ca2+ indicator Fluo3. 31P-Nuclear magnetic resonance measurements of {P5W30} indicate its stability in the medium used for cytosolic Ca2+ measurements for over 30 min. The findings reveal that addition of {P5W30} to the extracellular medium induces a sustained increase of the cytosolic Ca2+ concentration within minutes. This Ca2+ increase is triggered by extracellular Ca2+ entry into the cells and is dose-dependent, with a half-of-effect concentration of 0.25 ± 0.05 μM {P5W30}. In addition, after the {P5W30}-induced cytosolic Ca2+ increase, the transient Ca2+ peak induced by extracellular ATP is reduced up to 100% with an apparent half-of-effect concentration of 0.15 ± 0.05 μM {P5W30}. Activation of metabotropic purinergic P2 receptors affords about 80% contribution to the increase of Fluo3 fluorescence elicited by {P5W30} in HT-22 cells, whereas ionotropic receptors contribute, at most, with 20%. These results suggest that {P5W30} could serve as a novel agonist of purinergic P2 receptors.
Collapse
Affiliation(s)
- Joana Poejo
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Nadiia I Gumerova
- Universität Wien, Fakultät für Chemie, Institut für Biophysikalische Chemie, 1090 Vienna, Austria
| | - Annette Rompel
- Universität Wien, Fakultät für Chemie, Institut für Biophysikalische Chemie, 1090 Vienna, Austria.
| | - Ana M Mata
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain; Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Manuel Aureliano
- DCBB, Faculdade de Ciências e Tecnologia, Universidade do Algarve, 8000-139 Faro, Portugal; Centro de Ciências do Mar, Universidade do Algarve, 8000-139 Faro, Portugal.
| | - Carlos Gutierrez-Merino
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain.
| |
Collapse
|
18
|
Belenichev I, Bukhtiyarova N, Ryzhenko V, Makyeyeva L, Morozova O, Oksenych V, Kamyshnyi O. Methodological Approaches to Experimental Evaluation of Neuroprotective Action of Potential Drugs. Int J Mol Sci 2024; 25:10475. [PMID: 39408802 PMCID: PMC11477376 DOI: 10.3390/ijms251910475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The authors propose a novel approach to a comprehensive evaluation of neuroprotective effects using both in vitro and in vivo methods. This approach allows for the initial screening of numerous newly synthesized chemical compounds and substances from plant and animal sources while saving animal life by reducing the number of animals used in research. In vitro techniques, including mitochondrial suspensions and neuronal cell cultures, enable the assessment of neuroprotective activity, which can be challenging in intact organisms. The preliminary methods help outline the neuroprotection mechanism depending on the neurodestruction agent. The authors have validated a model of acute cerebrovascular accident, which simulates key cerebrovascular phenomena such as reduced cerebral blood flow, energy deficit, glutamate-calcium excitotoxicity, oxidative stress, and early gene expression. A significant advantage of this model is its ability to reproduce the clinical picture of cerebral ischemia: impaired motor activity; signs of neurological deficits (paresis, paralysis, etc.); as well as disturbances in attention, learning, and memory. Crucial to this approach is the selection of biochemical, molecular, and cellular markers to evaluate nerve tissue damage and characterize potential neuroprotective agents. Additionally, a comprehensive set of molecular, biochemical, histological, and immunohistochemical methods is proposed for evaluating neuroprotective effects and underlying mechanisms of potential pharmaceutical compounds.
Collapse
Affiliation(s)
- Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69035 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69035 Zaporizhzhia, Ukraine
| | - Victor Ryzhenko
- Department of Medical and Pharmaceutical Informatics and Advanced Technologies, Zaporizhzhia State Medical and Pharmaceutical University, 69035 Zaporizhzhia, Ukraine
| | - Lyudmyla Makyeyeva
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69035 Zaporizhzhia, Ukraine
| | - Oksana Morozova
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69035 Zaporizhzhia, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| |
Collapse
|
19
|
Li Z, Cao Z, Chen F, Li B, Jin H. Lutein inhibits glutamate-induced apoptosis in HT22 cells via the Nrf2/HO-1 signaling pathway. Front Neurosci 2024; 18:1432969. [PMID: 39193525 PMCID: PMC11347311 DOI: 10.3389/fnins.2024.1432969] [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: 05/15/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Excessive glutamate levels induce oxidative stress, resulting in neuronal damage, and cell death. While natural antioxidants show promise for neuroprotection, their effectiveness in the central nervous system (CNS) is limited by the blood -brain barrier. Lutein, a neuroprotective carotenoid, has gained attention for its ability to traverse this barrier and accumulate in various brain regions. This study aimed to elucidate the mechanisms underlying the protective effects of lutein against glutamateinduced cell death in HT22 cells. Methods HT22 cells were treated with lutein (1.25-20 μM) for 24 hours. Cell viability, ROS levels, apoptosis, and mitochondrial membrane potential were assessed following lutein pretreatment and glutamate exposure. Protein expression of apoptotic markers was analyzed using Western blotting. Results Lutein effectively attenuated glutamate-induced apoptosis due to its antioxidant properties. Additionally, lutein inhibited glutamate-induced mitochondrial-mediated apoptosis. We observed that lutein modulated the nuclear translocation of nuclear factor erythroid 2 -related factor 2 (Nrf2) and upregulated the expression of heme oxygenase-1 (HO-1). Inhibition of HO-1 by tin protoporphyrin (SnPP), a synthetic inhibitor, weakened the protective effect of lutein. Furthermore, we demonstrated that lutein prevented the aberrant activation of MAPKs induced by glutamate, including ERK1/2, p38, and JNK, thereby conferring oxidative protection. Discussion Our study highlights the potent antioxidant properties of lutein, which effectively safeguards against glutamate-induced mitochondrial apoptotic cell death through the Nrf2/HO-1 signaling pathway and inhibition of MAPK activation. These findings demonstrate that lutein exerts a neuroprotective effect against glutamate-induced neuronal cell damage.
Collapse
Affiliation(s)
- Zhenhua Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Zhuohua Cao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Fangmei Chen
- Institute of Science and Technology Information Research of Tibet Autonomous Region, Lhasa, China
| | - Bin Li
- Key Laboratory of Pharmaceutical Research for Metabolic Diseases, Department of Pharmacy, Qingdao University of Science and Technology, Qingdao, China
- Department of Medicament, College of Medicine, Tibet University, Lhasa, China
| | - Hanyong Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| |
Collapse
|
20
|
Bartman AE, Raeisi M, Peiris CD, Jacobsen IE, Martin DB, Doorn JA. A Novel Analog of the Natural Product Fraxinellone Protects against Endogenous and Exogenous Neurotoxicants. ACS Chem Neurosci 2024; 15:2612-2622. [PMID: 38925635 PMCID: PMC11258694 DOI: 10.1021/acschemneuro.4c00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Numerous insults, both endogenous (e.g., glutamate) and exogenous (e.g., pesticides), compromise the function of the nervous system and pose risk factors for damage or later disease. In previous reports, limonoids such as fraxinellone showed significant neuroprotective activity against glutamate (Glu) excitotoxicity and reactive oxygen species (ROS) production in vitro, albeit with minimal mechanistic information provided. Given these findings, a library of novel fraxinellone analogs (including analogs 1 and 2 described here) was synthesized with the goal of identifying compounds exhibiting neuroprotection against insults. Analog 2 was found to be protective against Glu-mediated excitotoxicity with a measured EC50 of 44 and 39 nM for in vitro assays using PC12 and SH-SY5Y cells, respectively. Pretreatment with analog 2 yielded rapid induction of antioxidant genes, namely, Gpx4, Sod1, and Nqo1, as measured via qPCR. Analog 2 mitigated Glu-mediated ROS. Cytoprotection could be replicated using sulforaphane (SFN), a Nrf2 activator, and inhibited via ML-385, which inhibits Nrf2 binding to regulatory DNA sequences, thereby blocking downstream gene expression. Nrf2 DNA-binding activity was demonstrated using a Nrf2 ELISA-based transcription factor assay. In addition, we found that pretreatment with the thiol N-acetyl Cys completely mitigated SFN-mediated induction of antioxidant genes but had no effect on the activity of analog 2, suggesting thiol modification is not critical for its mechanism of action. In summary, our data demonstrate a fraxinellone analog to be a novel, potent, and rapid activator of the Nrf2-mediated antioxidant defense system, providing robust protection against insults.
Collapse
Affiliation(s)
- Anna E. Bartman
- Department
of Pharmaceutical Sciences & Experimental Therapeutics, College
of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States
| | - Mersad Raeisi
- Department
of Chemistry, College of Liberal Arts & Sciences, University of Iowa, Iowa City, Iowa 52242, United States
| | - Clarence D. Peiris
- Department
of Chemistry, College of Liberal Arts & Sciences, University of Iowa, Iowa City, Iowa 52242, United States
| | - Isabella E. Jacobsen
- Department
of Chemistry, College of Liberal Arts & Sciences, University of Iowa, Iowa City, Iowa 52242, United States
| | - David B.C. Martin
- Department
of Chemistry, College of Liberal Arts & Sciences, University of Iowa, Iowa City, Iowa 52242, United States
| | - Jonathan A. Doorn
- Department
of Pharmaceutical Sciences & Experimental Therapeutics, College
of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
21
|
Alberto-Silva C, da Silva BR, da Silva JCA, da Cunha e Silva FA, Kodama RT, da Silva WD, Costa MS, Portaro FCV. Small Structural Differences in Proline-Rich Decapeptides Have Specific Effects on Oxidative Stress-Induced Neurotoxicity and L-Arginine Generation by Arginosuccinate Synthase. Pharmaceuticals (Basel) 2024; 17:931. [PMID: 39065782 PMCID: PMC11279908 DOI: 10.3390/ph17070931] [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: 06/22/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION The proline-rich decapeptide 10c (Bj-PRO-10c; ENWPHPQIPP) from the Bothrops jararaca snake modulates argininosuccinate synthetase (AsS) activity to stimulate L-arginine metabolite production and neuroprotection in the SH-SY5Y cell line. The relationships between structure, interactions with AsS, and neuroprotection are little known. We evaluated the neuroprotective effects of Bj-PRO-10c and three other PROs (Bn-PRO-10a, METHODS Cell integrity, metabolic activity, reactive oxygen species (ROS) production, and arginase activity were examined after 4 h of PRO pre-treatment and 20 h of H2O2-induced damage. RESULTS Only Bn-PRO-10a-MK and Bn-PRO-10c restored cell integrity and arginase function under oxidative stress settings, but they did not reduce ROS or cell metabolism. The MK dipeptide in Bn-PRO-10a-MK and valine (V8) in Bn-PRO-10c are important to these effects when compared to Bn-PRO-10a. Bj-PRO-10c is not neuroprotective in PC12 cells, perhaps because of their limited NMDA-type glutamate receptor activity. The PROs interaction analysis on AsS activation can be rated as follows: Bj-PRO-10c > Bn-PRO-10c > Bn-PRO-10a-MK > Bn-PRO-10a. The structure of PROs and their correlations with enzyme activity revealed that histidine (H5) and glutamine (Q7) in Bj-PRO-10c potentiated their affinity for AsS. CONCLUSIONS Our investigation provides the first insights into the structure and molecular interactions of PROs with AsS, which could possibly further their neuropharmacological applications.
Collapse
Affiliation(s)
- Carlos Alberto-Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Brenda Rufino da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Julio Cezar Araujo da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Felipe Assumpção da Cunha e Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Roberto Tadashi Kodama
- Structure and Functions of Biomolecules Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (R.T.K.); (F.C.V.P.)
| | - Wilmar Dias da Silva
- Laboratory of Immunochemistry, Butantan Institute, São Paulo 05503-900, SP, Brazil;
| | - Maricilia Silva Costa
- Instituto de Pesquisa & Desenvolvimento—IP&D, Universidade do Vale do Paraíba—UNIVAP, Av. Shishima Hifumi, 2911, São José dos Campos 12244-390, SP, Brazil;
| | - Fernanda Calheta Vieira Portaro
- Structure and Functions of Biomolecules Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (R.T.K.); (F.C.V.P.)
| |
Collapse
|
22
|
Song C, Chu Z, Dai J, Xie D, Qin T, Xie L, Zhai Z, Huang S, Xu Y, Sun T. Water extract of moschus alleviates erastin-induced ferroptosis by regulating the Keap1/Nrf2 pathway in HT22 cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117937. [PMID: 38423409 DOI: 10.1016/j.jep.2024.117937] [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: 12/20/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 03/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Moschus, first described in the Shennong's Classic of the Materia medicine, is a scarce and precious animal medicine. Modern pharmacological researches have suggested that Moschus has neuroprotective actions, and its mechanism is related to anti-inflammatory, antioxidant, and anti-apoptosis effects. Ferroptosis is one of the major pathologies of Alzheimer's disease (AD) and is widely implicated in the pathogenesis and progression of AD. Although previous studies have suggested that Moschus possesses neuroprotective effect, whether Moschus could mitigate neuronal damages by inhibiting the onset of ferroptosis is unknown in model cells of AD. AIM OF THE STUDY The aim of study was to explore the water extract of Moschus (WEM) on ferroptosis caused by erastin and the potential mechanism. MATERIALS AND METHODS Erastin was used to stimulate HT22 cells to form ferroptosis model to evaluate the anti-ferroptosis effect of WEM by cell counting kit-8 and lactic dehydrogenase (LDH) tests. The malondialdehyde (MDA) and glutathione (GSH) kits are used for detection of MDA and GSH levels, and 2',7'-dichlorofluorescein diacetate and C11 BODIPY 581/591 fluorescence probe are used for evaluation of reactive oxygen species (ROS) and lipid peroxide (LOOH) levels. And Western blot was used to test nuclear factor erythroid 2-related factor 2 (Nrf2), Kelch-like ECH-associated protein 1 (Keap1), heme oxygenase-1 (HO-1), and ferroptosis associated proteins including glutathione peroxidase 4 (GPX4), cystine/glutamate antiporter subunit (SLC7A11), ferritin heavy chain 1 (FTH1), ferroportin1 (FPN1), transferrin receptor (TFRC). In addition, the Nrf2 inhibitor ML385 was applied to verify whether WEM prevents erastin-induced ferroptosis by activating the Keap1/Nrf2 pathway. RESULTS After WEM treatment, erastin-induced HT22 cell survival was significantly elevated, the accumulation of intracellular MDA, ROS, and LOOH were significantly reduced, the level of GSH and expressions of ferroptosis inhibitors GPX4 and SLC7A11 were significantly increased, and iron metabolism-related proteins TFRC, FPN1, and FTH1 were regulated. These effects of WEM are implemented by activating the Keap1/Nrf2 pathway. CONCLUSIONS This study demonstrated that WEM could perform neuroprotective effects by alleviating ferroptosis, verified that WEM treatment of AD can be mediated by the Keap1/Nrf2 pathway, and provided theoretical support for the application of WEM in the treatment of AD.
Collapse
Affiliation(s)
- Caiyou Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhili Chu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jingyi Dai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Linjiang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhenwei Zhai
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Sha Huang
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
23
|
Yang Y, He B, Mu X, Qi S. Exposure to flutolanil at environmentally relevant concentrations can induce image and non-image-forming failure of zebrafish larvae through neuro and visual disruptions. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:134108. [PMID: 38521039 DOI: 10.1016/j.jhazmat.2024.134108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Numerous pesticides pose a threat to aquatic ecosystems, jeopardizing aquatic animal species and impacting human health. While the contamination of aquatic environment by flutolanil and its adverse effects on animal in the treatment of rich sheath blight have been reported, the neuro-visual effects of flutolanil at environmentally relevant concentrations remain unknown. In this study, we administered flutolanil to zebrafish embryos (0, 0.125, 0.50 and 2.0 mg/L) for 4 days to investigate its impact on the neuro and visual system. The results revealed that flutolanil induced abnormal behavior in larvae, affecting locomotor activity, stimuli response and phototactic response. Additionally, it led to defective brain and ocular development and differentiation. The disruption extended to the neurological system and visual phototransduction of larvae, evidenced by significant disturbances in genes and proteins related to neurodevelopment, neurotransmission, eye development, and visual function. Untargeted metabolomics analysis revealed that the GABAergic signaling pathway and increased levels of glutamine, glutamate, andγ-aminobutyric acid were implicated in the response to neuro and visual system injury induced by flutolanil, contributing to aberrant development, behavioral issues, and endocrine disruption. This study highlights the neuro-visual injury caused by flutolanil in aquatic environment, offering fresh insights into the mechanisms underlying image and non-image effects.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Bin He
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Sciences, Wuhan 430070, People's Republic of China
| | - Xiyan Mu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, People's Republic of China.
| | - Suzhen Qi
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, People's Republic of China.
| |
Collapse
|
24
|
Matus V, Castro-Guarda M, Cárcamo-Fierro J, Morera FJ, Zambrano A. Interleukin 3 Inhibits Glutamate-Cytotoxicity in Neuroblastoma Cell Line. Neurochem Res 2024; 49:1373-1386. [PMID: 38512424 DOI: 10.1007/s11064-024-04123-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: 09/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 03/23/2024]
Abstract
Interleukin 3 (IL-3) is a well-known pleiotropic cytokine that regulates the proliferation and differentiation of hematopoietic progenitor cells, triggering classical signaling pathways such as JAK/STAT, Ras/MAPK, and PI3K/Akt to carry out its functions. Interestingly, the IL-3 receptor is also expressed in non-hematopoietic cells, playing a crucial role in cell survival. Our previous research demonstrated the expression of the IL-3 receptor in neuron cells and its protective role in neurodegeneration. Glutamate, a principal neurotransmitter in the central nervous system, can induce cellular stress and lead to neurotoxicity when its extracellular concentrations surpass normal levels. This excessive glutamate presence is frequently observed in various neurological diseases. In this study, we uncover the protective role of IL-3 as an inhibitor of glutamate-induced cell death, analyzing the cytokine's signaling pathways during its protective effect. Specifically, we examined the relevance of JAK/STAT, Ras/MAPK, and PI3 K signaling pathways in the molecular mechanism triggered by IL-3. Our results show that the inhibition of JAK, ERK, and PI3 K signaling pathways, using pharmacological inhibitors, effectively blocked IL-3's protective role against glutamate-induced cell death. Additionally, our findings suggest that Bcl-2 and Bax proteins may be involved in the molecular mechanism triggered by IL-3. Our investigation into IL-3's ability to protect neuronal cells from glutamate-induced damage offers a promising therapeutic avenue with potential clinical implications for several neurological diseases characterized by glutamate neurotoxicity.
Collapse
Affiliation(s)
- Verónica Matus
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Marcos Castro-Guarda
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Joaquín Cárcamo-Fierro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Francisco J Morera
- Applied Biochemistry Laboratory, Escuela de Medicina Veterinaria, Facultad de Agronomía y Sistemas Naturales, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, 7820436, Santiago, Chile
| | - Angara Zambrano
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
25
|
Lai Y, Han J, Qiu D, Liu X, Sun K, Fan Y, Wang C, Zhang S. The protective effects of methylene blue on astrocytic swelling after cerebral ischemia-reperfusion injuries are mediated by Aquaporin-4 and metabotropic glutamate receptor 5 activation. Heliyon 2024; 10:e29483. [PMID: 38644842 PMCID: PMC11031768 DOI: 10.1016/j.heliyon.2024.e29483] [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: 10/11/2023] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024] Open
Abstract
Methylene blue (MB) was found to exert neuroprotective effect on different brain diseases, such as ischemic stroke. This study assessed the MB effects on ischemia induced brain edema and its role in the inhibition of aquaporin 4 (AQP4) and metabotropic glutamate receptor 5 (mGluR5) expression. Rats were exposed 1 h transient middle cerebral artery occlusion (tMCAO), and MB was injected intravenously following reperfusion (3 mg/kg). Magnetic resonance imaging (MRI) and 2,3,5-triphenyltetrazolium chloride (TTC) staining was performed 48 h after the onset of tMCAO to evaluate the brain infarction and edema. Brain tissues injuries as well as the glial fibrillary acidic protein (GFAP), AQP4 and mGluR5 expressions were detected. Oxygen and glucose deprivation/reoxygenation (OGD/R) was performed on primary astrocytes (ASTs) to induce cell swelling. MB was administered at the beginning of reoxygenation, and the perimeter of ASTs was measured by GFAP immunofluorescent staining. 3,5-dihydroxyphenylglycine (DHPG) and fenobam were given at 24 h before OGD to examine their effects on MB functions on AST swelling and AQP4 expression. MB remarkably decreased the volumes of T2WI and ADC lesions, as well as the cerebral swelling. Consistently, MB treatment significantly decreased GFAP, mGluR5 and AQP4 expression at 48 h after stroke. In the cultivated primary ASTs, OGD/R and DHPG significantly increased ASTs volume as well as AQP4 expression, which was reversed by MB and fenobam treatment. The obtained results highlight that MB decreases the post-ischemic brain swelling by regulating the activation of AQP4 and mGluR5, suggesting potential applications of MB on clinical ischemic stroke treatment.
Collapse
Affiliation(s)
- Yu Lai
- Department of Cardiovascular, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Jie Han
- Department of Cardiovascular, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Dongxian Qiu
- Department of Dermatology, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Xinyan Liu
- Medical Insurance Division, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Kan Sun
- Department of Cardiovascular, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Yuzhu Fan
- Department of Endocrinology, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Chunliang Wang
- Department of Cardiovascular, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| | - Song Zhang
- Department of Cardiovascular, The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
26
|
Chen LC, Lai MC, Hong TY, Liu IM. γ-Oryzanol from Rice Bran Antagonizes Glutamate-Induced Excitotoxicity in an In Vitro Model of Differentiated HT-22 Cells. Nutrients 2024; 16:1237. [PMID: 38674927 PMCID: PMC11053564 DOI: 10.3390/nu16081237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/03/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
The excessive activation of glutamate in the brain is a factor in the development of vascular dementia. γ-Oryzanol is a natural compound that has been shown to enhance brain function, but more research is needed to determine its potential as a treatment for vascular dementia. This study investigated if γ-oryzanol can delay or improve glutamate neurotoxicity in an in vitro model of differentiated HT-22 cells and explored its neuroprotective mechanisms. The differentiated HT-22 cells were treated with 0.1 mmol/L glutamate for 24 h then given γ-oryzanol at appropriate concentrations or memantine (10 µmol/L) for another 24 h. Glutamate produced reactive oxygen species and depleted glutathione in the cells, which reduced their viability. Mitochondrial dysfunction was also observed, including the inhibition of mitochondrial respiratory chain complex I activity, the collapse of mitochondrial transmembrane potential, and the reduction of intracellular ATP levels in the HT-22 cells. Calcium influx triggered by glutamate subsequently activated type II calcium/calmodulin-dependent protein kinase (CaMKII) in the HT-22 cells. The activation of CaMKII-ASK1-JNK MAP kinase cascade, decreased Bcl-2/Bax ratio, and increased Apaf-1-dependent caspase-9 activation were also observed due to glutamate induction, which were associated with increased DNA fragmentation. These events were attenuated when the cells were treated with γ-oryzanol (0.4 mmol/L) or the N-methyl-D-aspartate receptor antagonist memantine. The results suggest that γ-oryzanol has potent neuroprotective properties against glutamate excitotoxicity in differentiated HT-22 cells. Therefore, γ-oryzanol could be a promising candidate for the development of therapies for glutamate excitotoxicity-associated neurodegenerative diseases, including vascular dementia.
Collapse
Affiliation(s)
- Li-Chai Chen
- Department of Pharmacy and Master Program, Collage of Pharmacy and Health Care, Tajen University, Pingtung County 90741, Taiwan; (L.-C.C.); (M.-C.L.)
| | - Mei-Chou Lai
- Department of Pharmacy and Master Program, Collage of Pharmacy and Health Care, Tajen University, Pingtung County 90741, Taiwan; (L.-C.C.); (M.-C.L.)
| | - Tang-Yao Hong
- Department of Environmental Science and Occupational Safety and Hygiene, Graduate School of Environmental Management, Collage of Pharmacy and Health Care, Tajen University, Pingtung County 90741, Taiwan;
| | - I-Min Liu
- Department of Pharmacy and Master Program, Collage of Pharmacy and Health Care, Tajen University, Pingtung County 90741, Taiwan; (L.-C.C.); (M.-C.L.)
| |
Collapse
|
27
|
Vongthip W, Nilkhet S, Boonruang K, Sukprasansap M, Tencomnao T, Baek SJ. Neuroprotective mechanisms of luteolin in glutamate-induced oxidative stress and autophagy-mediated neuronal cell death. Sci Rep 2024; 14:7707. [PMID: 38565590 PMCID: PMC10987666 DOI: 10.1038/s41598-024-57824-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Neurodegenerative diseases, characterized by progressive neuronal dysfunction and loss, pose significant health challenges. Glutamate accumulation contributes to neuronal cell death in diseases such as Alzheimer's disease. This study investigates the neuroprotective potential of Albizia lebbeck leaf extract and its major constituent, luteolin, against glutamate-induced hippocampal neuronal cell death. Glutamate-treated HT-22 cells exhibited reduced viability, altered morphology, increased ROS, and apoptosis, which were attenuated by pre-treatment with A. lebbeck extract and luteolin. Luteolin also restored mitochondrial function, decreased mitochondrial superoxide, and preserved mitochondrial morphology. Notably, we first found that luteolin inhibited the excessive process of mitophagy via the inactivation of BNIP3L/NIX and inhibited lysosomal activity. Our study suggests that glutamate-induced autophagy-mediated cell death is attenuated by luteolin via activation of mTORC1. These findings highlight the potential of A. lebbeck as a neuroprotective agent, with luteolin inhibiting glutamate-induced neurotoxicity by regulating autophagy and mitochondrial dynamics.
Collapse
Affiliation(s)
- Wudtipong Vongthip
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Program in Clinical Biochemistry and Molecular Medicine, Chulalongkorn University, 10330, Bangkok, Thailand
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Sunita Nilkhet
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Program in Clinical Biochemistry and Molecular Medicine, Chulalongkorn University, 10330, Bangkok, Thailand
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Kanokkan Boonruang
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Monruedee Sukprasansap
- Food Toxicology Unit, Institute of Nutrition, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Seung Joon Baek
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
28
|
Rong J, Sun S, Xu SX, Xie XH, Wang C, Chen G, Kang L, Xiang D, Liu Z. The Neuroprotective Effects of BMSC-Derived Exosomes against Glutamate-Induced HT22 Cell Cytotoxicity. Neuroscience 2024; 542:1-10. [PMID: 38342336 DOI: 10.1016/j.neuroscience.2024.01.023] [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: 06/30/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/13/2024]
Abstract
Many central nervous system diseases are closely related to nerve damage caused by dysregulation of the endogenous neurotransmitter glutamate. Exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos) play an important role in improving injury and regeneration functions. However, its mechanism remains unknown. Therefore, the aim of this study is to investigate whether and how BMSC-Exos improve neurotoxicity caused by glutamate and to fill the gap in the literature. In this study, glutamate-treated HT22 cells were first exposed to mouse-derived BMSC-Exos at different concentrations to observe their effects on HT22 apoptosis. Next, we treated glutamate-treated HT22 cells with mouse-derived BMSC-Exos. We then inhibited the PI3K/Akt/mTOR signaling pathways using the PI3K/Akt inhibitor and the mTOR inhibitor, respectively, and observed the protective effect of mouse-derived BMSC-Exos on HT22 cells treated with glutamate. Our results show that BMSC-Exos reduced apoptosis triggered by glutamate stimulation, increased cell vitality, and decreased the levels of proapoptotic proteins while increasing the levels of anti-apoptotic proteins. The protective effect of BMSC-Exos was weakened when PI3K/Akt inhibitor and mTOR inhibitor were added. To sum up, we draw the following conclusions: BMSC-Exos can reduce neuronal apoptosis and apoptosis-related protein expression after glutamate stimulation by regulating the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jingtong Rong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Siqi Sun
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Shu-Xian Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Xin-Hui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Chao Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Guopeng Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Lijun Kang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Dan Xiang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060 China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072 China.
| |
Collapse
|
29
|
Zhao Y, Li B, Cao H, Wang F, Mu M, Jin H, Liu J, Fan Z, Tao X. Maternal nicotine exposure promotes hippocampal CeRNA-mediated excitotoxicity and social barriers in adolescent offspring mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116079. [PMID: 38377778 DOI: 10.1016/j.ecoenv.2024.116079] [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: 10/25/2023] [Revised: 01/18/2024] [Accepted: 02/04/2024] [Indexed: 02/22/2024]
Abstract
Nicotine, an addictive component of cigarettes, causes cognitive defects, particularly when exposure occurs early in life. However, the exact mechanism through which nicotine causes toxicity and alters synaptic plasticity is still not fully understood. The aim of the current study is to examine how non-coding developmental regulatory RNA impacts the hippocampus of mice offspring whose mothers were exposed to nicotine. Female C57BL/6J mice were given nicotine water from one week before pregnancy until end of lactation. Hippocampal tissue from offspring at 20 days post-birth was used for LncRNA and mRNA microarray analysis. Differential expression of LncRNAs and mRNAs associated with neuronal development were screened and validated, and the CeRNA pathway mediating neuronal synaptic plasticity GM13530/miR-7119-3p/mef2c was predicted using LncBase Predicted v.2. Using protein immunoblotting, Golgi staining and behavioral tests, our findings revealed that nicotine exposure in offspring mice increased hippocampal NMDAR receptor, activated receptor-dependent calcium channels, enhanced the formation of NMDAR/nNOS/PSD95 ternary complexes, increased NO synthesis, mediated p38 activation, induced neuronal excitability toxicity. Furthermore, an epigenetic CeRNA regulatory mechanism was identified, which suppresses Mef2c-mediated synaptic plasticity and leads to modifications in the learning and social behavior of the offspring during adolescence. This study uncovers the way in which maternal nicotine exposure results in neurotoxicity in offspring.
Collapse
Affiliation(s)
- Yehong Zhao
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Bing Li
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Hangbing Cao
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Fei Wang
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Min Mu
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Haibo Jin
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Jing Liu
- The First Hospital of Anhui University of Science and Technology, Huainan, China
| | - Zhenzhen Fan
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Xinrong Tao
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232000, China; Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China.
| |
Collapse
|
30
|
Feng L, Dai S, Zhang C, Zhang W, Zhu W, Wang C, He Y, Song W. Ripa-56 protects retinal ganglion cells in glutamate-induced retinal excitotoxic model of glaucoma. Sci Rep 2024; 14:3834. [PMID: 38360971 PMCID: PMC10869350 DOI: 10.1038/s41598-024-54075-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/08/2024] [Indexed: 02/17/2024] Open
Abstract
Glaucoma is a prevalent cause of blindness globally, characterized by the progressive degeneration of retinal ganglion cells (RGCs). Among various factors, glutamate excitotoxicity stands out as a significant contributor of RGCs loss in glaucoma. Our study focused on Ripa-56 and its protective effect against NMDA-induced retinal damage in mice, aiming to delve into the potential underlying mechanism. The R28 cells were categorized into four groups: glutamate (Glu), Glu + Ripa-56, Ripa-56 and Control group. After 24 h of treatment, cell death was assessed by PI / Hoechst staining. Mitochondrial membrane potential changes, apoptosis and reactive oxygen species (ROS) production were analyzed using flow cytometry. The alterations in the expression of RIP-1, p-MLKL, Bcl-2, BAX, Caspase-3, Gpx4 and SLC7A11 were examined using western blot analysis. C57BL/6j mice were randomly divided into NMDA, NMDA + Ripa-56, Ripa-56 and control groups. Histological changes in the retina were evaluated using hematoxylin and eosin (H&E) staining. RGCs survival and the protein expression changes of RIP-1, Caspase-3, Bcl-2, Gpx4 and SLC7A11 were observed using immunofluorescence. Ripa-56 exhibited a significant reduction in the levels of RIP-1, p-MLKL, Caspase-3, and BAX induced by glutamate, while promoting the expression of Bcl-2, Gpx-4, and SLC7A1 in the Ripa-56-treated group. In our study, using an NMDA-induced normal tension glaucoma mice model, we employed immunofluorescence and H&E staining to observe that Ripa-56 treatment effectively ameliorated retinal ganglion cell loss, mitigating the decrease in retinal ganglion cell layer and bipolar cell layer thickness caused by NMDA. In this study, we have observed that Ripa-56 possesses remarkable anti- necroptotic, anti-apoptotic and anti-ferroptosis properties. It demonstrates the ability to combat not only glutamate-induced excitotoxicity in R28 cells, but also NMDA-induced retinal excitotoxicity in mice. Therefore, Ripa-56 could be used as a potential retinal protective agent.
Collapse
Affiliation(s)
- Lemeng Feng
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Shirui Dai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, People's Republic of China
| | - Cheng Zhang
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Wulong Zhang
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Weiming Zhu
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Chao Wang
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Ye He
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Weitao Song
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
31
|
Zhang Y, Shaabani S, Vowinkel K, Trombetta-Lima M, Sabogal-Guáqueta AM, Chen T, Hoekstra J, Lembeck J, Schmidt M, Decher N, Dömling A, Dolga AM. Novel SK channel positive modulators prevent ferroptosis and excitotoxicity in neuronal cells. Biomed Pharmacother 2024; 171:116163. [PMID: 38242037 DOI: 10.1016/j.biopha.2024.116163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Small conductance calcium-activated potassium (SK) channel activity has been proposed to play a role in the pathology of several neurological diseases. Besides regulating plasma membrane excitability, SK channel activation provides neuroprotection against ferroptotic cell death by reducing mitochondrial Ca2+ uptake and reactive oxygen species (ROS). In this study, we employed a multifaceted approach, integrating structure-based and computational techniques, to strategically design and synthesize an innovative class of potent small-molecule SK2 channel modifiers through highly efficient multicomponent reactions (MCRs). The compounds' neuroprotective activity was compared with the well-studied SK positive modulator, CyPPA. Pharmacological SK channel activation by selected compounds confers neuroprotection against ferroptosis at low nanomolar ranges compared to CyPPA, that mediates protection at micromolar concentrations, as shown by an MTT assay, real-time cell impedance measurements and propidium iodide staining (PI). These novel compounds suppress increased mitochondrial ROS and Ca2+ level induced by ferroptosis inducer RSL3. Moreover, axonal degeneration was rescued by these novel SK channel activators in primary mouse neurons and they attenuated glutamate-induced neuronal excitability, as shown via microelectrode array. Meanwhile, functional afterhyperpolarization of the novel SK2 channel modulators was validated by electrophysiological measurements showing more current change induced by the novel modulators than the reference compound, CyPPA. These data support the notion that SK2 channel activation can represent a therapeutic target for brain diseases in which ferroptosis and excitotoxicity contribute to the pathology.
Collapse
Affiliation(s)
- Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Shabnam Shaabani
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Kirsty Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technologies and Biopharmacy, Research Institute of Pharmacy, University of Groningen, the Netherlands
| | | | - Tingting Chen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Hoekstra
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Lembeck
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| |
Collapse
|
32
|
Liang L, Heinrichs RW, Liddle PF, Jeon P, Théberge J, Palaniyappan L. Cortical impoverishment in a stable subgroup of schizophrenia: Validation across various stages of psychosis. Schizophr Res 2024; 264:567-577. [PMID: 35644706 DOI: 10.1016/j.schres.2022.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cortical thinning is a well-known feature in schizophrenia. The considerable variation in the spatial distribution of thickness changes has been used to parse heterogeneity. A 'cortical impoverishment' subgroup with a generalized reduction in thickness has been reported. However, it is unclear if this subgroup is recoverable irrespective of illness stage, and if it relates to the glutamate hypothesis of schizophrenia. METHODS We applied hierarchical cluster analysis to cortical thickness data from magnetic resonance imaging scans of three datasets in different stages of psychosis (n = 288; 160 patients; 128 healthy controls) and studied the cognitive and symptom profiles of the observed subgroups. In one of the samples, we also studied the subgroup differences in 7-Tesla magnetic resonance spectroscopy glutamate concentration in the dorsal anterior cingulate cortex. RESULTS Our consensus-based clustering procedure consistently produced 2 subgroups of participants. Patients accounted for 75%-100% of participants in one subgroup that was characterized by significantly lower cortical thickness. Both subgroups were equally symptomatic in clinically unstable stages, but cortical impoverishment indicated a higher symptom burden in a clinically stable sample and higher glutamate levels in the first-episode sample. There were no subgroup differences in cognitive and functional outcome profiles or antipsychotic exposure across all stages. CONCLUSIONS Cortical thinning does not vary with functioning or cognitive impairment, but it is more prevalent among patients, especially those with glutamate excess in early stages and higher residual symptom burden at later stages, providing an important mechanistic clue to one of the several possible pathways to the illness.
Collapse
Affiliation(s)
- Liangbing Liang
- Graduate Program in Neuroscience, Western University, London, Ontario, Canada; Robarts Research Institute, Western University, London, Ontario, Canada
| | | | - Peter F Liddle
- Institute of Mental Health, Division of Mental Health and Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Peter Jeon
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Jean Théberge
- Department of Medical Biophysics, Western University, London, Ontario, Canada; Department of Psychiatry, Western University, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada
| | - Lena Palaniyappan
- Robarts Research Institute, Western University, London, Ontario, Canada; Department of Medical Biophysics, Western University, London, Ontario, Canada; Department of Psychiatry, Western University, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada; Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
33
|
Hu L, Wang S, Zhang L, Shang L, Zong R, Li J, Wu Z, Meng Y, Dai Y, Huang Y, Wei G. Wild imitating vs greenhouse cultivated Dendrobium huoshanense: Chemical quality differences. PLoS One 2024; 19:e0291376. [PMID: 38271357 PMCID: PMC10810538 DOI: 10.1371/journal.pone.0291376] [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: 06/12/2023] [Accepted: 08/28/2023] [Indexed: 01/27/2024] Open
Abstract
Dendrobium huoshanense (D. huoshanense) has been used as functional food supplements and herbal medicines for preventing and managing diseases with a long history in China. Due to its endangered natural resources and huge demand, people tend to cultivate D. huoshanense to protect this species. However, the quality of wild and cultivated herbs of the same species may change. This work quantified and compared the main quality traits and chemical components of wild imitating and greenhouse cultivated D. huoshanense with different growth years. As a result, wild and cultivated D. huoshanense had similar chemical composition, but there are significant differences in the content of many ingredients (polysaccharides, flavonoids, nucleosides, bibenzyls, lignans and volatile compounds). And the contents of many of these components increased with growing years. In addition, multivariate statistical analyses have been applied to classify and evaluate samples from different cultivation modes according to these components. In conclusion, our results demonstrated that the overall quality of greenhouse cultivated D. huoshanense was not as good as wild-grown, but this mode can be a promising and sustainable way of producing D. huoshanense.
Collapse
Affiliation(s)
- Li Hu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shiwen Wang
- Jiuxianzun Dendrobium Huoshanense Co. Ltd., Lu’an, China
| | - Lin Zhang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Ruiye Zong
- Jiuxianzun Dendrobium Huoshanense Co. Ltd., Lu’an, China
| | - Jinyan Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhanghua Wu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanjun Meng
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafeng Dai
- Jiuxianzun Dendrobium Huoshanense Co. Ltd., Lu’an, China
| | - Yuechun Huang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gang Wei
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
34
|
Indurkar A, Kudale P, Rjabovs V, Heinmaa I, Demir Ö, Kirejevs M, Rubenis K, Chaturbhuj G, Turks M, Locs J. Small organic molecules containing amorphous calcium phosphate: synthesis, characterization and transformation. Front Bioeng Biotechnol 2024; 11:1329752. [PMID: 38283170 PMCID: PMC10811600 DOI: 10.3389/fbioe.2023.1329752] [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: 10/29/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
As the primary solid phase, amorphous calcium phosphate (ACP) is a pivotal precursor in cellular biomineralization. The intrinsic interplay between ACP and Howard factor underscores the significance of understanding their association for advancing biomimetic ACP development. While organic compounds play established roles in biomineralization, this study presents the synthesis of ACP with naturally occurring organic compounds (ascorbate, glutamate, and itaconate) ubiquitously found in mitochondria and vital for bone remodeling and healing. The developed ACP with organic compounds was meticulously characterized using XRD, FTIR, and solid-state 13C and 31P NMR. The morphological analysis revealed the characteristic spherical morphology with particle size close to 20 nm of all synthesized ACP variants. Notably, the type of organic compound strongly influences true density, specific surface area, particle size, and transformation. The in vitro analysis was performed with MC3T3-E1 cells, indicating the highest cell viability with ACP_ASC (ascorbate), followed by ACP_ITA (itaconate). The lowest cell viability was observed with 10 %w/v of ACP_GLU (glutamate); however, 1 %w/v of ACP_GLU was cytocompatible. Further, the effect of small organic molecules on the transformation of ACP to low crystalline apatite (Ap) was examined in Milli-Q® water, PBS, and α-MEM.
Collapse
Affiliation(s)
- Abhishek Indurkar
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Pawan Kudale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Vitālijs Rjabovs
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
| | - Ivo Heinmaa
- National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Öznur Demir
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Matvejs Kirejevs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
| | - Kristaps Rubenis
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Ganesh Chaturbhuj
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Māris Turks
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
| | - Janis Locs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
35
|
Shehata SA, Kolieb E, Ali DA, Maher SA, Korayem HE, Ibrahim MA, Nafie MS, Ameen SH. Selenium alleviates modafinil-induced neurobehavioral toxicity in rat via PI3K/Akt/mTOR/GSK3B signaling pathway and suppression of oxidative stress and apoptosis: in vivo and in silico study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:458-480. [PMID: 38015391 DOI: 10.1007/s11356-023-31093-4] [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: 08/23/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Nonmedical use of modafinil (MOD) led to increased rates of overdose toxicity, road accidents, addiction, withdrawal, suicide, and mental illnesses. The current study aims to determine the probable MOD brain toxicity and elucidate the possible role of selenium (Se) in ameliorating the neurotoxicity in rat models. Fifty-four male Albino rats were randomly assigned into nine groups. The groups were G1 (control negative), G2 (Se0.1), G3 (Se0.2), G4 (MOD300), G5 (MOD600), G6 (Se0.1 + MOD300), G7 (Se0.2 + MOD300), G8 (Se0.1 + MOD600), and G9 (Se0.2 + MOD600). After finishing the experiment, blood and brain tissue were harvested for biochemical and histological investigation. Neurobehavior parameters were assessed. Tissue neurotransmitter levels and oxidative stress markers were assessed. Gene expression of PI3K/Akt/mTOR-GSK3B, orexin, and orexin receptor2 was measured by qRT-PCR. Histological and immunohistochemistry assessments, as well as molecular docking, were carried out. MOD-induced neurobehavioral toxicity exhibited by behavioral and cognitive function impairments, which are associated with decreased antioxidant activities, increased MDA levels, and decreases in neurotransmitter levels. Brain levels of mRNA expression of PI3K, Akt, and mTOR were decreased, while GS3K, orexin, and orexin receptors were significantly elevated. These disturbances were confirmed by histopathological brain changes with increased silver and Bax immunostaining and decreased crystal violet levels. MOD induced neurotoxic effects in a dose-dependent manner. Compared with the MOD groups, SE coadministration significantly attenuates MOD-induced toxic changes. Docking study shows the protective role of Se as an apoptosis inhibitor and inflammation inhibitor. In conclusion, Se could be used as a biologically effective antioxidant compound to protect from MOD neurobehavioral toxicity in Wistar rats by reversing behavioral alterations, inflammation, apoptosis, and oxidative injury.
Collapse
Affiliation(s)
- Shaimaa A Shehata
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Eman Kolieb
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Dina A Ali
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Shymaa Ahmed Maher
- Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Horeya Erfan Korayem
- Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Mahrous A Ibrahim
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
- Forensic Medicine and Clinical Toxicology, College of Medicine, Jouf University, 72341, Aljouf, Saudi Arabia.
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, P. O. Box 27272, Sharjah, United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| | - Shimaa H Ameen
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Alsharqia, Egypt
| |
Collapse
|
36
|
Oh YC, Jeong YH, Yang HJ, Li W, Ma JY. Lumbricus Extract Prevents LPS-Induced Inflammatory Activation of BV2 Microglia and Glutamate-Induced Hippocampal HT22 Cell Death by Suppressing MAPK/NF-κB/NLRP3 Signaling and Oxidative Stress. Curr Issues Mol Biol 2023; 45:9926-9942. [PMID: 38132466 PMCID: PMC10742620 DOI: 10.3390/cimb45120620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
Microglia-induced inflammatory signaling and neuronal oxidative stress are mutually reinforcing processes central to the pathogenesis of neurodegenerative diseases. Recent studies have shown that extracts of dried Pheretima aspergillum (Lumbricus) can inhibit tissue fibrosis, mitochondrial damage, and asthma. However, the effects of Lumbricus extracts on neuroinflammation and neuronal damage have not been previously studied. Therefore, to evaluate the therapeutic potential of Lumbricus extract for neurodegenerative diseases, the current study assessed the extract's anti-inflammatory and antioxidant activities in BV2 microglial cultures stimulated with lipopolysaccharide (LPS) along with its neuroprotective efficacy in mouse hippocampal HT22 cell cultures treated with excess glutamate. Lumbricus extract dose-dependently inhibited the LPS-induced production of multiple proinflammatory cytokines (tumor necrosis factor-α, interleukin (IL)-6, and IL-1β) and reversed the upregulation of proinflammatory enzymes (inducible nitric oxide synthase and cyclooxygenase-2). Lumbricus also activated the antioxidative nuclear factor erythroid 2-relayed factor 2/heme oxygenase-1 pathway and inhibited LPS-induced activation of the nuclear factor-κB/mitogen-activated protein kinases/NOD-like receptor family pyrin domain containing 3 inflammatory pathway. In addition, Lumbricus extract suppressed the glutamate-induced necrotic and apoptotic death of HT22 cells, effects associated with upregulated expression of antiapoptotic proteins, downregulation of pro-apoptotic proteins, and reduced accumulation of reactive oxygen species. Chromatography revealed that the Lumbricus extract contained uracil, hypoxanthine, uridine, xanthine, adenosine, inosine, and guanosine. Its effects against microglial activation and excitotoxic neuronal death reported herein support the therapeutic potential of Lumbricus for neurodegenerative diseases.
Collapse
Affiliation(s)
- You-Chang Oh
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine, 70, Cheomdanro, Dong-gu, Daegu 41062, Republic of Korea; (Y.H.J.); (H.J.Y.); (W.L.)
| | | | | | | | - Jin Yeul Ma
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine, 70, Cheomdanro, Dong-gu, Daegu 41062, Republic of Korea; (Y.H.J.); (H.J.Y.); (W.L.)
| |
Collapse
|
37
|
Šachlevičiūtė U, Gonzalez G, Kvasnicová M, Štěpánková Š, Kleizienė N, Bieliauskas A, Zatloukal M, Strnad M, Sløk FA, Kvasnica M, Šačkus A, Žukauskaitė A. Synthesis and neuroprotective activity of 3-aryl-3-azetidinyl acetic acid methyl ester derivatives. Arch Pharm (Weinheim) 2023; 356:e2300378. [PMID: 37797174 DOI: 10.1002/ardp.202300378] [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: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
A library of 3-aryl-3-azetidinyl acetic acid methyl ester derivatives was prepared from N-Boc-3-azetidinone employing the Horner-Wadsworth-Emmons reaction, rhodium(I)-catalyzed conjugate addition of arylboronic acids, and subsequent elaborations to obtain N-unprotected hydrochlorides, N-alkylated and N-acylated azetidine derivatives. The compounds were evaluated for acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitory activity, revealing several derivatives to possess AChE inhibition comparable to that of the AChE inhibitor rivastigmine. The binding mode of the AChE inhibitor donepezil and selected active compounds 26 and 27 within the active site of AChE was studied using molecular docking. Furthermore, the neuroprotective activity of the prepared compounds was evaluated in models associated with Parkinson's disease (salsolinol-induced) and aspects of Alzheimer's disease (glutamate-induced oxidative damage). Compound 28 showed the highest neuroprotective effect in both salsolinol- and glutamate-induced neurodegeneration models, and its protective effect in the glutamate model was revealed to be driven by a reduction in oxidative stress and caspase-3/7 activity.
Collapse
Affiliation(s)
- Urtė Šachlevičiūtė
- Institute of Synthetic Chemistry, Kaunas University of Technology, Kaunas, Lithuania
| | - Gabriel Gonzalez
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
- Department of Neurology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | - Marie Kvasnicová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Šárka Štěpánková
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Pardubice, Czech Republic
| | - Neringa Kleizienė
- Institute of Synthetic Chemistry, Kaunas University of Technology, Kaunas, Lithuania
| | - Aurimas Bieliauskas
- Institute of Synthetic Chemistry, Kaunas University of Technology, Kaunas, Lithuania
| | - Marek Zatloukal
- Department of Chemical Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences & Palacký University, Olomouc, Czech Republic
| | | | - Miroslav Kvasnica
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences & Palacký University, Olomouc, Czech Republic
| | - Algirdas Šačkus
- Institute of Synthetic Chemistry, Kaunas University of Technology, Kaunas, Lithuania
| | - Asta Žukauskaitė
- Department of Chemical Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| |
Collapse
|
38
|
Yao X, Xu X, Hu K, Yang Z, Deng S. BANF1 promotes glutamate-induced apoptosis of HT-22 hippocampal neurons. Mol Biol Rep 2023; 50:9441-9452. [PMID: 37838622 DOI: 10.1007/s11033-023-08889-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Glutamate exposure was fatal to HT-22 neuronal cells that derived from mouse hippocampus. This is often used as a model for hippocampus neurodegeneration in vitro. The targets relevant to glutamate-induced neuronal toxicity is not fully understood. In this study, we aimed to identify crucial factors associated with glutamate-induced cytotoxicity in HT-22 cells. METHODS HT-22 cells were treated with 7.5 mM glutamate for 24 h and isobaric tags for relative and absolute quantitation (iTRAQ) proteomic analysis conducted to identify the differentially expressed proteins. Differential proteins were subjected to Gene Ontology analyses. Upregulation of barrier to autointegration factor (BANF1/BANF1) protein was confirmed by RT-qPCR and western blotting. Cell viability was measured by CKK-8 and MTT assays. Cell apoptosis rates and intracellular reactive oxygen species (ROS) levels were detected using flow cytometry. RESULTS A total of 5811 proteins were quantified by iTRAQ, 50 of which were recognized as significantly differential proteins (fold change ≥ 1.5 and P ≤ 0.05); 26 proteins were up-regulated and 24 were down-regulated after exposure to glutamate. GO enrichment analysis showed that the apoptotic signaling pathway was involved in cell death induced by glutamate. BANF1 expression level was markedly increased in HT-22 cells after glutamate treatment. Further, knockdown of BANF1 alleviated glutamate-mediated cell death with lower ROS levels. CONCLUSIONS In conclusion, we successfully filtered out differential proteins relevant to glutamate-mediated cytotoxicity. BANF1 upregulation promoted glutamate-induced apoptosis of HT-22 cells by enhancing ROS generation.
Collapse
Affiliation(s)
- Xinyu Yao
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiaoyi Xu
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, Guangdong, China
| | - Kunhua Hu
- Proteomics Research Center, Sun Yat-Sen Medical College of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhaoshou Yang
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Shaodong Deng
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, 523710, Guangdong, China.
- Scientific Research Platform, The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
39
|
El-Hashash SA, El-Sakhawy MA, Eldamaty HS, Alqasem AA. Experimental evidence of the neurotoxic effect of monosodium glutamate in adult female Sprague Dawley rats: The potential protective role of Zingiber officinale Rosc. rhizomes. Saudi J Biol Sci 2023; 30:103824. [PMID: 37869363 PMCID: PMC10587751 DOI: 10.1016/j.sjbs.2023.103824] [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: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023] Open
Abstract
Strategies to prevent the health abnormalities associated with the extensive use of MSG (monosodium glutamate) as a flavoring booster are badly needed. The current study was conducted to investigate oxidative stress, inflammation, and abnormal lipid profile as the main risk factors of neurotoxicity in MSG-exposed female albino rats. Besides, the effect of concurrent consumption of Zingiber officinale rhizomes powder was studied at low doses. Twenty rats (total) were split into 4 separate groups. The 1st group was a negative control group (without any treatment), while the others received 6 mg MSG/kg. The 2nd group was left untreated, whereas the 3rd and 4th groups were given a regular laboratory diet that included ginger rhizome powder supplements (GRP, 0.5 & 1%, respectively) for six weeks. In brain tissue homogenates, exposure to MSG caused a significant depletion of gamma-aminobutyric acid (GABA) and total protein levels, while triglycerides and cholesterol contents were significantly elevated. Moreover, a noteworthy upsurge in oxidative load and inflammation markers was also noticed associated with a marked reduction of antioxidant levels, which histopathological staining verified further. The rat diet formulated with GRP, with a dose-dependent effect, resulted in increased GABA and total protein contents and attenuated inflammation, oxidative stress, abnormal lipid profile, and marked histological changes in cerebral cortical neurons of MSG-administered animals. Therefore, this study reveals that GRP shields rats against the neurotoxicity that MSG causes. The anti-inflammatory as well as antioxidant, and lipid-normalizing properties of rhizomes of ginger may be accountable for their observed neuroprotective action.
Collapse
Affiliation(s)
- Samah A. El-Hashash
- Department of Nutrition and Food Science, Faculty of Home Economics, Al-Azhar University, Nawag, Tanta City, P.O. Box 31732, Egypt
| | - Mohamed A. El-Sakhawy
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Medicinal and Aromatic Plants, Desert Research Center, Cairo, Egypt
| | - Hanan S.E. Eldamaty
- Department of Nutrition and Food Science, Faculty of Home Economics, Al-Azhar University, Nawag, Tanta City, P.O. Box 31732, Egypt
| | - Abdullah A. Alqasem
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
40
|
Borgonetti V, Caroli C, Governa P, Virginia B, Pollastro F, Franchini S, Manetti F, Les F, López V, Pellati F, Galeotti N. Helichrysum stoechas (L.) Moench reduces body weight gain and modulates mood disorders via inhibition of silent information regulator 1 (SIRT1) by arzanol. Phytother Res 2023; 37:4304-4320. [PMID: 37433745 DOI: 10.1002/ptr.7941] [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: 04/20/2023] [Revised: 06/06/2023] [Accepted: 06/25/2023] [Indexed: 07/13/2023]
Abstract
The prevalence of obesity is steadily rising, making safe and more efficient anti-obesity treatments an urgent medical need. Growing evidence correlates obesity and comorbidities, including anxiety and depression, with the development of a low-grade inflammation in peripheral and central tissues. We hypothesized that attenuating neuroinflammation might reduce weight gain and improve mood. We investigated the efficacy of a methanolic extract from Helichrysum stoechas (L.) Moench (HSE), well-known for its anti-inflammatory properties, and its main constituent arzanol (AZL). HPLC-ESI-MS2 and HPLC-UV were used to characterize the extract. HSE effects on mood and feeding behavior was assessed in mice. The mechanism of action of HSE and AZL was investigated in hippocampus samples and SH-SY5Y cells by western blotting and immunofluorescence. Oral administration of HSE for 3 weeks limited weight gain with no significant decrease in food intake. HSE produced an anxiolytic-like and antidepressant-like phenotype comparable to diazepam and amitriptyline, respectively, in the absence of locomotor and cognitive impairments and induced neuroprotective effects in glutamate-exposed SH-SY5Y cells. A dose-dependent reduction of SIRT1 expression was detected in SH-SY5Y cells and in hippocampal samples from HSE-treated mice. The inhibition of the SIRT1-FoxO1 pathway was induced in the hypothalamus. Molecular docking studies proposed a mechanism of SIRT1 inhibition by AZL, confirmed by the evaluation of inhibitory effects on SIRT1 enzymatic activity. HSE limited weight gain and comorbidities through an AZL-mediated SIRT1 inhibition. These activities indicate HSE an innovative therapeutic perspective for obesity and associated mood disorders.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
- Department of Molecular Medicine and Neuroscience, Scripps Research Institute, La Jolla, California, USA
| | - Clarissa Caroli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, California, USA
| | - Brighenti Virginia
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Novara, Italy
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Zaragoza, Spain
| | - Victor López
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Zaragoza, Spain
| | - Federica Pellati
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
41
|
Abstract
Iron accumulation in the CNS occurs in many neurological disorders. It can contribute to neuropathology as iron is a redox-active metal that can generate free radicals. The reasons for the iron buildup in these conditions are varied and depend on which aspects of iron influx, efflux, or sequestration that help maintain iron homeostasis are dysregulated. Iron was shown recently to induce cell death and damage via lipid peroxidation under conditions in which there is deficient glutathione-dependent antioxidant defense. This form of cell death is called ferroptosis. Iron chelation has had limited success in the treatment of neurological disease. There is therefore much interest in ferroptosis as it potentially offers new drugs that could be more effective in reducing iron-mediated lipid peroxidation within the lipid-rich environment of the CNS. In this review, we focus on the molecular mechanisms that induce ferroptosis. We also address how iron enters and leaves the CNS, as well as the evidence for ferroptosis in several neurological disorders. Finally, we highlight biomarkers of ferroptosis and potential therapeutic strategies.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Fari Ryan
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Priya Jhelum
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
42
|
Li H, Liu X, Zhong H, Fang J, Li X, Shi R, Yu Q. Research progress on the pathogenesis of diabetic retinopathy. BMC Ophthalmol 2023; 23:372. [PMID: 37697295 PMCID: PMC10494348 DOI: 10.1186/s12886-023-03118-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
Diabetic retinopathy is one of the most common and serious microvascular complications of diabetes mellitus. There are many factors leading to diabetic retinopathy, and its pathogenesis is still unclear. At present, there are still no effective measures for the early treatment of diabetic retinopathy, and the treatment options available when diabetes progresses to advanced stages are very limited, and the treatment results are often unsatisfactory. Detailed studies on the molecular mechanisms of diabetic retinopathy pathogenesis and the development of new therapeutic agents are of great importance. This review describes the potential pathogenesis of diabetic retinopathy for experimental studies and clinical practice.
Collapse
Affiliation(s)
- Hongbo Li
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.
| | - Xinyu Liu
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Hua Zhong
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Jiani Fang
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Xiaonan Li
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Rui Shi
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Qi Yu
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| |
Collapse
|
43
|
Pak ME, Li W. Neuroprotective Effects of Sparassis crispa Ethanol Extract through the AKT/NRF2 and ERK/CREB Pathway in Mouse Hippocampal Cells. J Fungi (Basel) 2023; 9:910. [PMID: 37755018 PMCID: PMC10532724 DOI: 10.3390/jof9090910] [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: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Sparassis crispa, known as the "Cauliflower mushroom", is an edible medicinal fungus found in Asia, Europe, and North America. Its fruiting bodies contain active biological and pharmacological ingredients with antitumor and anti-inflammatory properties. In this study, we investigated the neuroprotective effect of various Sparassis crispa extract against glutamate-induced toxicity and oxidative stress in hippocampal HT22 cells. Cell viability and reactive oxygen species (ROS) analyses served to evaluate the neuroprotective effects of Sparassis crispa ethanol extract (SCE) and their fractions partitioned with ethyl acetate (EtOAc; SCE-E) and water (SCE-W) in HT22 cells. SCE and SCE-E treatment reduced glutamate-induced cell death and ROS generation. SCE-E reduced apoptosis and ROS levels by regulating anti-apoptotic proteins. Under glutamate treatment, SCE-E activated nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and regulated extracellular signal-regulated kinase (ERK) and AKT signals at late stages. SCE-E increased the protein expression of cAMP response element binding (CREB), brain-derived neurotrophic factor (BDNF), and Kelch-like ECH-associated protein 1 (Keap1), and decreased the Nrf2 protein expression. Moreover, co-treatment of SCE-E and wortmannin did not activate Nrf2 expression. Thus, the neuroprotective effect of SCE-E is likely due to Nrf2 and CREB activation through AKT and ERK phosphorylation, which effectively suppress glutamate-induced oxidative stress in HT22 cells. Accordingly, a daily supplement of SCE-E could become a potential treatment for oxidative-stress-related neurological diseases.
Collapse
Affiliation(s)
| | - Wei Li
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| |
Collapse
|
44
|
Putralis R, Korotkaja K, Kaukulis M, Rudevica Z, Jansons J, Nilova O, Rucins M, Krasnova L, Domracheva I, Plotniece M, Pajuste K, Sobolev A, Rumnieks F, Bekere L, Zajakina A, Plotniece A, Duburs G. Styrylpyridinium Derivatives for Fluorescent Cell Imaging. Pharmaceuticals (Basel) 2023; 16:1245. [PMID: 37765053 PMCID: PMC10535741 DOI: 10.3390/ph16091245] [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: 06/15/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
A set of styrylpyridinium (SP) compounds was synthesised in order to study their spectroscopic and cell labelling properties. The compounds comprised different electron donating parts (julolidine, p-dimethylaminophenyl, p-methoxyphenyl, 3,4,5-trimethoxyphenyl), conjugated linkers (vinyl, divinyl), and an electron-withdrawing N-alkylpyridinium part. Geminal or bis-compounds incorporating two styrylpyridinium (bis-SP) moieties at the 1,3-trimethylene unit were synthesised. Compounds comprising a divinyl linker and powerful electron-donating julolidine donor parts possessed intensive fluorescence in the near-infrared region (maximum at ~760 nm). The compounds had rather high cytotoxicity towards the cancerous cell lines HT-1080 and MH-22A; at the same time, basal cytotoxicity towards the NIH3T3 fibroblast cell line ranged from toxic to harmful. SP compound 6e had IC50 values of 1.0 ± 0.03 µg/mL to the cell line HT-1080 and 0.4 µg/mL to MH-22A; however, the basal toxicity LD50 was 477 mg/kg (harmful). The compounds showed large Stokes' shifts, including 195 nm for 6a,b, 240 nm for 6e, and 325 and 352 nm for 6d and 6c, respectively. The highest photoluminescence quantum yield (PLQY) values were observed for 6a,b, which were 15.1 and 12.2%, respectively. The PLQY values for the SP derivatives 6d,e (those with a julolidinyl moiety) were 0.5 and 0.7%, respectively. Cell staining with compound 6e revealed a strong fluorescent signal localised in the cell cytoplasm, whereas the cell nuclei were not stained. SP compound 6e possessed self-assembling properties and formed liposomes with an average diameter of 118 nm. The obtained novel data on near-infrared fluorescent probes could be useful for the development of biocompatible dyes for biomedical applications.
Collapse
Affiliation(s)
- Reinis Putralis
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, LV-1007 Riga, Latvia;
| | - Ksenija Korotkaja
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (K.K.); (Z.R.); (J.J.); (O.N.); (F.R.); (A.Z.)
| | - Martins Kaukulis
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1048 Riga, Latvia
| | - Zhanna Rudevica
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (K.K.); (Z.R.); (J.J.); (O.N.); (F.R.); (A.Z.)
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (K.K.); (Z.R.); (J.J.); (O.N.); (F.R.); (A.Z.)
| | - Olga Nilova
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (K.K.); (Z.R.); (J.J.); (O.N.); (F.R.); (A.Z.)
| | - Martins Rucins
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| | - Laura Krasnova
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| | - Ilona Domracheva
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| | - Mara Plotniece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, LV-1007 Riga, Latvia;
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1048 Riga, Latvia
| | - Karlis Pajuste
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| | - Arkadij Sobolev
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| | - Felikss Rumnieks
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (K.K.); (Z.R.); (J.J.); (O.N.); (F.R.); (A.Z.)
| | - Laura Bekere
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| | - Anna Zajakina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (K.K.); (Z.R.); (J.J.); (O.N.); (F.R.); (A.Z.)
| | - Aiva Plotniece
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, LV-1007 Riga, Latvia;
| | - Gunars Duburs
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (R.P.); (M.K.); (M.R.); (L.K.); (I.D.); (K.P.); (A.S.); (L.B.)
| |
Collapse
|
45
|
Hou Y, Michiels J, Kerschaver CV, Vandaele M, Majdeddin M, Vossen E, Degroote J. The kinetics of glutathione in the gastrointestinal tract of weaned piglets supplemented with different doses of dietary reduced glutathione. Front Vet Sci 2023; 10:1220213. [PMID: 37635757 PMCID: PMC10448897 DOI: 10.3389/fvets.2023.1220213] [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: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
This study aimed to investigate the kinetics of dietary GSH in the gastrointestinal tract and the effect of GSH on the intestinal redox status of weaned piglets. Forty-eight piglets with an average age of 26 days and an average body weight of 7.7 kg were used in this study. The piglets were divided into three treatment groups including the control group with a basal diet (CON) and two GSH groups with a basal diet supplemented with 0.1% GSH (LGSH) and 1.0% GSH (HGSH), respectively. The basal diet did not contain any GSH. The experiment lasted for 14 days, with eight animals sampled from each group on d5 and 14. The parts of 0-5%, 5-75%, and 75-100% of the length of the small intestine were assigned to SI1, SI2, and SI3. The results showed that GSH almost completely disappeared from the digesta at SI2. However, no difference in the GSH level in mucosa, liver, and blood erythrocytes was found. The level of cysteine (CYS) in SI1 digesta was significantly higher in HGSH than CON and LGSH on d14, and similar findings were observed for cystine (CYSS) in SI3 digesta on d5. The CYSS level in HGSH was also significantly higher than LGSH in the stomach on d14, while no CYS or CYSS was detected in the stomach for control animals, indicating the breakdown of GSH to CYS already occurred in the stomach. Irrespective of the dietary treatment, the CYS level on d14 and the CYSS level on d5 and 14 were increased when moving more distally into the gastrointestinal tract. Furthermore, the mucosal CYS level was significantly increased at SI1 in the LGSH and HGSH group compared with CON on d5. Glutathione disulfide (GSSG) was recovered in the diets and digesta from the LGSH and HGSH group, which could demonstrate the auto-oxidation of GSH. It is, therefore, concluded that GSH supplementation could not increase the small intestinal mucosal GSH level of weaned piglets, and this could potentially relate to the kinetics of GSH in the digestive tract, where GSH seemed to be prone to the breakdown to CYS and CYSS and the auto-oxidation to GSSG.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jeroen Degroote
- Laboratory of Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, Ghent, Belgium
| |
Collapse
|
46
|
Takahashi K, Ishibashi Y, Chujo K, Suzuki I, Sato K. Neuroprotective Potential of L-Glutamate Transporters in Human Induced Pluripotent Stem Cell-Derived Neural Cells against Excitotoxicity. Int J Mol Sci 2023; 24:12605. [PMID: 37628787 PMCID: PMC10454411 DOI: 10.3390/ijms241612605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived neural cells have started to be used in safety/toxicity tests at the preclinical stage of drug development. As previously reported, hiPSC-derived neurons exhibit greater tolerance to excitotoxicity than those of primary cultures of rodent neurons; however, the underlying mechanisms remain unknown. We here investigated the functions of L-glutamate (L-Glu) transporters, the most important machinery to maintain low extracellular L-Glu concentrations, in hiPSC-derived neural cells. We also clarified the contribution of respective L-Glu transporter subtypes. At 63 days in vitro (DIV), we detected neuronal circuit functions in hiPSC-derived neural cells by a microelectrode array system (MEA). At 63 DIV, exposure to 100 μM L-Glu for 24 h did not affect the viability of neural cells. 100 µM L-Glu in the medium decreased to almost 0 μM in 60 min. Pharmacological inhibition of excitatory amino acid transporter 1 (EAAT1) and EAAT2 suppressed almost 100% of L-Glu decrease. In the presence of this inhibitor, 100 μM L-Glu dramatically decreased cell viability. These results suggest that in hiPSC-derived neural cells, EAAT1 and EAAT2 are the predominant L-Glu transporters, and their uptake potentials are the reasons for the tolerance of hiPSC-derived neurons to excitotoxicity.
Collapse
Affiliation(s)
- Kanako Takahashi
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-city, Kanagawa 210-9501, Japan; (K.T.); (K.C.)
| | - Yuto Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Miyagi 982-8577, Japan; (Y.I.); (I.S.)
| | - Kaori Chujo
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-city, Kanagawa 210-9501, Japan; (K.T.); (K.C.)
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Miyagi 982-8577, Japan; (Y.I.); (I.S.)
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-city, Kanagawa 210-9501, Japan; (K.T.); (K.C.)
| |
Collapse
|
47
|
Pap R, Pandur E, Jánosa G, Sipos K, Fritz FR, Nagy T, Agócs A, Deli J. Protective Effects of 3'-Epilutein and 3'-Oxolutein against Glutamate-Induced Neuronal Damage. Int J Mol Sci 2023; 24:12008. [PMID: 37569384 PMCID: PMC10418699 DOI: 10.3390/ijms241512008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Dietary lutein can be naturally metabolized to 3'-epilutein and 3'-oxolutein in the human body. The epimerization of lutein can happen in acidic pH, and through cooking, 3'-epilutein can be the product of the direct oxidation of lutein in the retina, which is also present in human serum. The 3'-oxolutein is the main oxidation product of lutein. Thus, the allylic oxidation of dietary lutein can result in the formation of 3'-oxolutein, which may undergo reduction either to revert to dietary lutein or epimerize to form 3'-epilutein. We focused on the effects of 3'-epilutein and 3'-oxolutein itself and on glutamate-induced neurotoxicity on SH-SY5Y human neuroblastoma cells to identify the possible alterations in oxidative stress, inflammation, antioxidant capacity, and iron metabolism that affect neurological function. ROS measurements were performed in the differently treated cells. The inflammatory state of cells was followed by TNFα, IL-6, and IL-8 cytokine ELISA measurements. The antioxidant status of the cells was determined by the total antioxidant capacity kit assay. The alterations of genes related to ferroptosis and lipid peroxidation were followed by gene expression measurements; then, thiol measurements were performed. Lutein metabolites 3'-epilutein and 3'-oxolutein differently modulated the effect of glutamate on ROS, inflammation, ferroptosis-related iron metabolism, and lipid peroxidation in SH-SY5Y cells. Our results revealed the antioxidant and anti-inflammatory features of 3'-epilutein and 3'-oxolutein as possible protective agents against glutamate-induced oxidative stress in SH-SY5Y cells, with greater efficacy in the case of 3'-epilutein.
Collapse
Affiliation(s)
- Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Gergely Jánosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Ferenc Rómeó Fritz
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Tamás Nagy
- Department of Laboratory Medicine, Faculty of Medical Sciences, University of Pécs, Ifjúság út 13, H-7624 Pécs, Hungary;
| | - Attila Agócs
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary;
| | - József Deli
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary;
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| |
Collapse
|
48
|
Phoraksa O, Chimkerd C, Thiyajai P, Judprasong K, Tuntipopipat S, Tencomnao T, Charoenkiatkul S, Muangnoi C, Sukprasansap M. Neuroprotective Effects of Albizia lebbeck (L.) Benth. Leaf Extract against Glutamate-Induced Endoplasmic Reticulum Stress and Apoptosis in Human Microglial Cells. Pharmaceuticals (Basel) 2023; 16:989. [PMID: 37513900 PMCID: PMC10384906 DOI: 10.3390/ph16070989] [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: 06/17/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Endoplasmic reticulum (ER) stress caused by excessive glutamate in the central nervous system leads to neurodegeneration. Albizia lebbeck (L.) Benth. has been reported to possess neuroprotective properties. We aimed to investigate the effect and mechanism of A. lebbeck leaf extracts on glutamate-induced neurotoxicity and apoptosis linked to ER stress using human microglial HMC3 cells. A. lebbeck leaves were extracted using hexane (AHE), mixed solvents, and ethanol. Each different extract was evaluated for cytotoxic effects on HMC3 cells, and then non-cytotoxic concentrations of the extracts were pretreated with the cells, followed by glutamate. Our results showed that AHE treatment exhibited the highest protective effect and was thus selected for finding the mechanistic approach. AHE inhibited the specific ER stress proteins (calpain1 and caspase-12). AHE also suppressed the apoptotic proteins (Bax, cytochrome c, cleaved caspase-9, and cleaved caspase-3); however, it also increased the antiapoptotic Bcl-2 protein. Remarkably, AHE increased cellular antioxidant activities (SOD, CAT, and GPx). To support the activation of antioxidant defense and inhibition of apoptosis in our HMC3 cell model, the bioactive phytochemicals within AHE were identified by HPLC analysis. We found that AHE had high levels of carotenoids (α-carotene, β-carotene, and lutein) and flavonoids (quercetin, luteolin, and kaempferol). Our novel findings indicate that AHE can inhibit glutamate-induced neurotoxicity via ER stress and apoptosis signaling pathways by activating cellular antioxidant enzymes in HMC3 cells, suggesting a potential mechanism for neuroprotection. As such, A. lebbeck leaf might potentially represent a promising source and novel alternative approach for preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Onuma Phoraksa
- Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok 10400, Thailand
| | - Chanika Chimkerd
- Center of Analysis for Product Quality (Natural Products Division), Faculty of Pharmacy, Mahidol University, Rajathevi, Bangkok 10400, Thailand
| | - Parunya Thiyajai
- Food Chemistry Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| | - Kunchit Judprasong
- Food Chemistry Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| | - Siriporn Tuntipopipat
- Cell and Animal Model Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| | - Tewin Tencomnao
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand
| | - Somsri Charoenkiatkul
- Food Chemistry Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| | - Chawanphat Muangnoi
- Cell and Animal Model Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| | - Monruedee Sukprasansap
- Food Toxicology Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| |
Collapse
|
49
|
Sharma A, Sari E, Lee Y, Patel S, Brenner M, Marambaud P, Wang P. Extracellular CIRP Induces Calpain Activation in Neurons via PLC-IP 3-Dependent Calcium Pathway. Mol Neurobiol 2023; 60:3311-3328. [PMID: 36853429 PMCID: PMC10506840 DOI: 10.1007/s12035-023-03273-3] [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: 11/07/2022] [Accepted: 02/16/2023] [Indexed: 03/01/2023]
Abstract
Abnormal calcium homeostasis, activation of protease calpain, generation of p25 and hyperactivation of cyclin-dependent kinase 5 (Cdk5) have all been implicated in the pathogenesis of neurogenerative diseases including Alzheimer's disease. We have recently shown that extracellular cold-inducible RNA-binding protein (eCIRP) induces Cdk5 activation via p25. However, the precise molecular mechanism by which eCIRP regulates calcium signaling and calpain remains to be addressed. We hypothesized that eCIRP regulates p25 via Ca2+-dependent calpain activation. eCIRP increased calpain activity and decreased the endogenous calpain inhibitor calpastatin in Neuro 2a (N2a) cells. Calpain inhibition with calpeptin attenuated eCIRP-induced calpain activity and p25. eCIRP specifically upregulated cytosolic calpain 1, and calpain 1 silencing attenuated the eCIRP-induced increase in p25. eCIRP stimulation increased cytosolic free Ca2+, especially in hippocampal neuronal HT22 cells, which was attenuated by the eCIRP inhibitor Compound 23 (C23). Endoplasmic reticulum (ER) inositol 1,4,5-trisphosphate receptor (IP3R) inhibition using 2-aminoethoxy-diphenyl-borate or xestospongin-C (X-C), interleukin-6 receptor alpha (IL-6Rα)-neutralization, and phospholipase C (PLC) inhibition with U73122 attenuated eCIRP-induced Ca2+ increase, while Ca2+ influx across the plasma membrane remained unaffected by eCIRP. Finally, C23, IL-6Rα antibody, U73122 and X-C attenuated eCIRP-induced p25 in HT-22 cells. In conclusion, the current study uncovers eCIRP-triggered Ca2+ release from ER stores in an IL-6Rα/PLC/IP3-dependent manner as a novel molecular mechanism underlying eCIRP's induction of Cdk5 activity and potential involvement in neurodegeneration.
Collapse
Affiliation(s)
- Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Ezgi Sari
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Shivani Patel
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Philippe Marambaud
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
- The Litwin-Zucker Center for Alzheimer's Disease Research, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| |
Collapse
|
50
|
Prasansuklab A, Sukjamnong S, Theerasri A, Hu VW, Sarachana T, Tencomnao T. Transcriptomic analysis of glutamate-induced HT22 neurotoxicity as a model for screening anti-Alzheimer's drugs. Sci Rep 2023; 13:7225. [PMID: 37142620 PMCID: PMC10160028 DOI: 10.1038/s41598-023-34183-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
Glutamate-induced neurotoxicity in the HT22 mouse hippocampal neuronal cell line has been recognized as a valuable cell model for the study of neurotoxicity associated with neurodegenerative diseases including Alzheimer's disease (AD). However, the relevance of this cell model for AD pathogenesis and preclinical drug screening remains to be more elucidated. While there is increasing use of this cell model in a number of studies, relatively little is known about its underlying molecular signatures in relation to AD. Here, our RNA sequencing study provides the first transcriptomic and network analyses of HT22 cells following glutamate exposure. Several differentially expressed genes (DEGs) and their relationships specific to AD were identified. Additionally, the usefulness of this cell model as a drug screening system was assessed by determining the expression of those AD-associated DEGs in response to two medicinal plant extracts, Acanthus ebracteatus and Streblus asper, that have been previously shown to be protective in this cell model. In summary, the present study reports newly identified AD-specific molecular signatures in glutamate-injured HT22 cells, suggesting that this cell can be a valuable model system for the screening and evaluation of new anti-AD agents, particularly from natural products.
Collapse
Affiliation(s)
- Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Suporn Sukjamnong
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Atsadang Theerasri
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Valerie W Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Tewarit Sarachana
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|