1
|
Uy NP, He MT, Lee CD, Lee Y, Kang KS, Lee S. Artemisia extracts and phytochemicals inhibit glutamate-induced oxidative cell death and upregulate the Nrf2/HO-1 signaling pathway in HT22 neuronal cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119933. [PMID: 40339835 DOI: 10.1016/j.jep.2025.119933] [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: 03/03/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia (Asteraceae) is a diverse genus of plants with ecological, economic, and therapeutic significance. Some species have been traditionally used for neuroprotection; however, their bioactive potential remains underexplored. AIM OF THE STUDY To identify Artemesia extracts and phytochemicals with neuroprotective effects that could provide alternative sources of functional foods, nutraceuticals, and therapeutics. METHODS Twelve Artemisia extracts were tested for their neuroprotective effects, followed by phytochemical profiling and quantification using HPLC. Compounds with superior activity were subjected to western blot analysis. RESULTS Pretreatment with Artemesia stolonifera, A. rubripes, A. sieversiana, A. keiskeana, A. scoparia, and A. annua (125 μg/mL) attenuated glutamate-induced cytotoxicity. The findings revealed a significant (p < 0.05) heterogeneity in the phytochemical profiles of the species. Among the evaluated compounds, rutin (5) significantly protected HT22 cells from glutamate-induced damage. Pretreatment with rutin (5) upregulated Nrf2 and HO-1. CONCLUSIONS The study findings highlight the potential of Artemisia species as a source of bioactive chemicals in functional foods, nutraceuticals, and medicines. Further research on the bioactivity of these chemicals and the mechanisms responsible for their diversity could help develop cultivation strategies and potential medicinal applications.
Collapse
Affiliation(s)
- Neil Patrick Uy
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| | - Mei Tong He
- College of Korean Medicine, Gachon University, Seongnam, 13120, Republic of Korea.
| | - Chang-Dae Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| | - Yunji Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Eumseong, 27709, Republic of Korea.
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, 13120, Republic of Korea.
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea; Natural Product Institute of Science and Technology, Anseong, 17546, Republic of Korea.
| |
Collapse
|
2
|
Chu Z, Chen Y, Xie D, Song C, Yang L, Qin T, Zhai Z, Cao Z, Xu Y, Sun T. Ethanol extract of Moschus attenuates glutamate-induced cytotoxicity in HT22 cells by regulating the Nrf2 and MAPK pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119879. [PMID: 40288659 DOI: 10.1016/j.jep.2025.119879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Moschus is a traditional Chinese materia medica for treating central nervous system disorders. Oxidative stress is a key pathogenic mechanism of Alzheimer's disease (AD) and serves as a critical bridge linking various pathological processes of AD. Previous studies have shown that Moschus can exert neuroprotective effects by inhibiting glutamate-induced neuronal cell damage. However, its underlying mechanisms remain unclear. AIM OF THE STUDY This study aimed to evaluate the effects and potential mechanisms of the ethanol extract of Moschus (EEM) on glutamate-induced oxidative damage in HT22 cells. MATERIALS AND METHODS The components of EEM were identified using GC-MS. An oxidative toxicity cell model was established by exposing HT22 cells to glutamate. Cell viability was assessed through CCK8 and LDH assays, and the modes of cell death were evaluated using FITC-Annexin V staining and TUNEL assays. Intracellular and mitochondrial ROS levels were measured with DCFH-DA and MitoSOX Red probes. Intracellular Ca2+ levels were measured with the Fluo-4 AM fluorescent probe. Mitochondrial function was analyzed using the JC-1 fluorescent probe. Protein expression levels of Bid, Calpain-1, Bax, Bcl-2, AIF, P-ERK, ERK, P-JNK, JNK, P-P38, P38, Nrf2, HO-1, Keap1, and NQO-1 were analyzed through western blotting. The distribution of AIF and Nrf2 in the cytoplasm and nucleus was examined through immunofluorescence staining. RESULTS Using GC-MS, 18 major components were identified in EEM. EEM significantly inhibited apoptosis, reduced ROS generation, and alleviated Ca2+ overload. EEM restored mitochondrial dysfunction by regulating the expression of mitochondria-related apoptotic proteins, including the downregulation of Calpain-1 and Bax, upregulation of Bid and Bcl-2, and inhibition of AIF nuclear translocation. EEM inhibited MAPK phosphorylation while activating the Nrf2/Keap1 signaling pathway. CONCLUSIONS Our study shows that EEM protects HT22 cells from glutamate-induced damage by regulating the MAPK and Nrf2 pathways, effectively reducing oxidative stress and apoptosis. In summary, this study first demonstrates at the cellular level that EEM exerts neuroprotective effects by modulating the MAPK and Nrf2 pathways. These findings provide new insights into the mechanism of Moschus against AD and establish a foundation for its potential application in AD.
Collapse
Affiliation(s)
- Zhili Chu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yubing Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, 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
| | - Caiyou Song
- Traditional Chinese Medicine Factory Co. Ltd, Taiji Group Chongqing, Chongqing, 402284, China
| | - Lin Yang
- 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
| | - Zhenwei Zhai
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, 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
|
3
|
Chen J, Gou Z, Huang Y, Yu Q, Kim AN, Shi W, Zhou Y. Research Progress on Phytochemicals from Mulberry with Neuroprotective Effects: A Review. Pharmaceuticals (Basel) 2025; 18:695. [PMID: 40430516 PMCID: PMC12114994 DOI: 10.3390/ph18050695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
With the intensification of the population aging worldwide, neurological disorders (NDs) are seriously threatening human society. Mulberry, a traditional economic crop, is a significant medicinal plant. Increasing evidence suggests that phytochemicals from mulberry play critical roles in the prevention and treatment of NDs. This paper reviews the recently reported phytochemicals from mulberry with neuroprotective effects and systematically summarizes neuroprotective mechanisms and their classifications. Based on their origins from different parts of mulberry, the extracts with neuroprotective effects are classified into mulberry fruit extract and mulberry leaf extract. According to the compound structures, the compounds are divided into flavonoids, Diels-Alder-type adducts (DAAs), benzofurans, quinones, stilbenes, and alkaloids. This aims to provide a future reference for their pharmaceutical development and utilization.
Collapse
Affiliation(s)
- Junwei Chen
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (J.C.); (Z.G.); (W.S.)
| | - Zhonglang Gou
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (J.C.); (Z.G.); (W.S.)
| | - Yufei Huang
- Westa College, Southwest University, Chongqing 400715, China; (Y.H.); (Q.Y.)
| | - Qianhui Yu
- Westa College, Southwest University, Chongqing 400715, China; (Y.H.); (Q.Y.)
| | - An Na Kim
- CDD Engine, Lino Lakes, MN 55014, USA;
| | - Wenchao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (J.C.); (Z.G.); (W.S.)
| | - You Zhou
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; (J.C.); (Z.G.); (W.S.)
| |
Collapse
|
4
|
Liu Y, Chen X, Liu XJ, Liu R, Hu HL, Min SX, Huang CH, Liu L, Tan GS. Five racemic phthalides from the aerial parts of Lycopodiastrum casuarinoides and their neuroprotective activities. PHYTOCHEMISTRY 2025; 233:114384. [PMID: 39756559 DOI: 10.1016/j.phytochem.2025.114384] [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: 08/14/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Five racemic phthalides (1-5), including four undescribed phthalides monomers [(+)-1, (+)-2, (-)-2 and (-)-3], four undescribed phthalide dimers [(+)-4, (-)-4, (+)-5 and (-)-5], together with two known compounds [(-)-1 and (+)-3], were isolated from the aerial parts of Lycopodistrum casuarinoides. Their chemical structures were delineated by extensive spectroscopic data (UV, 1D/2D NMR, HRESIMS), in combination with the comparison of the experimental and calculated electronic circular dichroism spectra, calculated spin-spin coupling constants, and calculated NMR. All compounds were reported from Lycopodiaceae family for the first time. In addition, all isolates were tested for their neuroprotective effects on HT-22 cell injury induced by glutamate. Interestingly, among the five racemic phthalides, only the homologous dimers [(±)-5] displayed significant differences in neuroprotective effects, and (-)-5 exhibited the best neuroprotective activity against glutamate-induced HT-22 cells damage, with 29.3% increase rate in cell survival at 5 μM concentration. The neuroprotective effect of (-)-5 at different concentrations is equivalent to that of the positive control drug D/L-3-n-butylphthalide (racemic NBP). Furthermore, the biological evaluation revealed that (-)-5 could ameliorate glutamate-induced neuronal cell death via the Bax/Bcl-2 anti-apoptotic pathway.
Collapse
Affiliation(s)
- Yang Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; School of Pharmacy, Jiangxi Provincial Education Department Key Laboratory for the Application of Key Technologies in Drug Screening for Inflammatory Diseases and Phlegm Syndrome, Nanchang Medical College, Nanchang, Jiangxi, 330052, China; Jiangxi Provincial Key Laboratory of Tissue Engineering, 2024SSY06291, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Xi Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Xue-Jin Liu
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Ren Liu
- School of Pharmacy, Jiangxi Provincial Education Department Key Laboratory for the Application of Key Technologies in Drug Screening for Inflammatory Diseases and Phlegm Syndrome, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Hui-Ling Hu
- School of Pharmacy, Jiangxi Provincial Education Department Key Laboratory for the Application of Key Technologies in Drug Screening for Inflammatory Diseases and Phlegm Syndrome, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Shan-Xue Min
- School of Pharmacy, Jiangxi Provincial Education Department Key Laboratory for the Application of Key Technologies in Drug Screening for Inflammatory Diseases and Phlegm Syndrome, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Chao-Hui Huang
- School of Pharmacy, Jiangxi Provincial Education Department Key Laboratory for the Application of Key Technologies in Drug Screening for Inflammatory Diseases and Phlegm Syndrome, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Lin Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; Jiangxi Provincial Key Laboratory of Tissue Engineering, 2024SSY06291, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Gui-Shan Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
5
|
Jiang C, Liu R, Chang Y, Zhang S, Li X, Zhao Z, Quan M, Wang Q, Zhou H, Hou X, Fang H. Design and synthesis of novel benzoic acid derivatives as striatal-enriched protein tyrosine phosphatase (STEP) inhibitors with neuroprotective properties. Eur J Med Chem 2025; 283:117135. [PMID: 39657460 DOI: 10.1016/j.ejmech.2024.117135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
As a central nervous system-specific member of the protein tyrosine phosphatase (PTP) family, the striatal-enriched protein tyrosine phosphatase (STEP) is an attractive drug target for neurodegenerative diseases. Here, we reported the discovery of a series of benzoic acid derivatives as new STEP inhibitors. Among them, compound 14b exhibited good STEP inhibitory activity and displayed selectivity against other PTPs. The neuroprotective activity of compound 14b was evaluated against glutamate-induced oxidative cell death in HT22 cells. Results indicated that compound 14b co-treatment prevented cell death and reduced cellular ROS accumulation. Compound 14b inhibited cell apoptosis by upregulating BCL-2 expression and downregulating BAX and C-caspase3 expression. Moreover, compound 14b was also found to provide neuroprotection to primary cortical neurons after oxygen-glucose deprivation/reoxygenation (OGD/R). Further structural elaboration of compound 14b may provide new drug candidates for neurodegenerative diseases.
Collapse
Affiliation(s)
- Chunxue Jiang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Renshuai Liu
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yong Chang
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Shiji Zhang
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xue Li
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhongcheng Zhao
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Mengyao Quan
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Quande Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Normal University, Guilin, 541004, China
| | - Hengxing Zhou
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Xuben Hou
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Hao Fang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
6
|
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] [MESH Headings] [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
|
7
|
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
|
8
|
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
|
9
|
Sun S, Hou X, Li K, Huang C, Rong Y, Bi J, Li X, Wu D. Curcumin and Metformin Infinite Coordination Polymer Nanoparticles for Combined Therapy of Diabetic Mice via Intraperitoneal Injections. J Funct Biomater 2024; 15:388. [PMID: 39728188 DOI: 10.3390/jfb15120388] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/01/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
Metformin (Met) is one of the most commonly prescribed first-line drugs for diabetes treatment. However, it has several issues, including low bioavailability, therapeutic platform, and side effects at high doses. In order to improve the therapeutic efficiency of Met, this study proposes a strategy of using Met and curcumin (Cur) to prepare Cur-Zn(II)-Met infinite coordination polymer nanoparticles (CM ICP NPs), and combining this with intraperitoneal injections, for the treatment of diabetic mice. Fourier transform infrared (FTIR) spectroscopy, X-ray photoelectron spectroscopy (XPS), transmission electron microscopy (TEM), nanoparticle analysis, cytotoxicity experiments, and mice experiments were used to investigate structure, properties, and application effects. The results showed that CM ICP NPs exhibit a high drug encapsulation rate (100%), good stability, and an absence of in vivo and in vitro toxicity. The blood glucose level of diabetic mice after treatment was reduced to 6.7 ± 0.65 mmol/L at the seventh week. In terms of therapeutic mechanism, it appears that Met and Cur can synergistically regulate blood glucose in mice from multiple paths. This study provides a promising method for the treatment of diabetes using Met and other drugs.
Collapse
Affiliation(s)
- Siwei Sun
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Xinyi Hou
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
- School of Pharmacy, Xi'an Medical University, Xi'an 710021, China
| | - Ke Li
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Chenqi Huang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yu Rong
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Jiao Bi
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
| | - Xueping Li
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Xi'an Medical University, Xi'an 710021, China
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
10
|
Nguyen CD, Yoo J, Jeong SJ, Ha HA, Yang JH, Lee G, Shin JC, Kim JH. Melittin - the main component of bee venom: a promising therapeutic agent for neuroprotection through keap1/Nrf2/HO-1 pathway activation. Chin Med 2024; 19:166. [PMID: 39605070 PMCID: PMC11603938 DOI: 10.1186/s13020-024-01020-x] [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: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
The Nuclear factor erythroid 2-related factor (Nrf2)/ Heme oxygenase-1 (HO-1) pathway, known for its significant role in regulating innate antioxidant defense mechanisms, is increasingly being recognized for its potential in neuroprotection studies. Derived from bee venom, melittin's neuroprotective effects have raised interest. This study confirmed that melittin specificity upregulated the weakened Nrf2/HO-1 signaling in mice brain. Interestingly, we also revealed melittin's efficient tactic, as it was suggested to first restore redox balance and then gradually stabilized other regulations of the mouse hippocampus. Using a neuro-stress-induced scopolamine model, chromatography and mass spectrometry analysis revealed that melittin crossed the compromised blood-brain barrier and accumulated in the hippocampus, which provided the chance to interact directly to weakened neurons. A wide range of improvements of melittin action were observed from various tests from behavior Morris water maze, Y maze test to immune florescent staining, western blots. As we need to find out what is the focus of melittin action, we conducted a careful observation in mice which showed that: the first signs of changes, in the hippocampus, within 5 h after melittin administration were the restoration of the Nrf2/HO-1 system and suppression of oxidative stress. After this event, from 7 to 12.5 h after administration, a diversity of conditions was all ameliorated: inflammation, apoptosis, neurotrophic factors, cholinergic function, and tissue ATP level. This chain reaction underscores that melittin focus was on redox balance's role, which revived multiple neuronal functions. Evidence of enhancement in the mouse hippocampus led to further exploration with hippocampal cell line HT22 model. Immunofluorescence analysis showed melittin-induced Nrf2 translocation to the nucleus, which would initiating the translation of antioxidant genes like HO-1. Pathway inhibitors pinpointed melittin's direct influence on the Nrf2/HO-1 pathway. 3D docking models and pull-down assays suggested melittin's direct interaction with Keap1, the regulator of the Nrf2/HO-1 pathway. Overall, this study not only highlighted melittin specifically effect on Nrf2/HO-1, thus rebalancing cellular redox, and also showed that this is an effective multi-faceted therapeutic strategy against neurodegeneration.
Collapse
Affiliation(s)
- Cong Duc Nguyen
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Jaehee Yoo
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Sang Jun Jeong
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Hai-Anh Ha
- Faculty of Pharmacy, College of Medicine and Pharmacy, Duy Tan University, Da Nang, 550000, Vietnam
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Gihyun Lee
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Jeong Cheol Shin
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea.
- Department of Acupuncture and Moxibustion Medicine, Dongshin University Gwangju Korean Medicine Hospital, 141, Wolsan-ro, Nam-gu, Gwangju City 61619, Republic of Korea , 141 Wolsan-Ro Nam-Gu, Gwangju, 61619, Republic of Korea.
| | - Jae-Hong Kim
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea.
- Department of Acupuncture and Moxibustion Medicine, Dongshin University Gwangju Korean Medicine Hospital, 141, Wolsan-ro, Nam-gu, Gwangju City 61619, Republic of Korea , 141 Wolsan-Ro Nam-Gu, Gwangju, 61619, Republic of Korea.
| |
Collapse
|
11
|
Shen H, Nie J, Li G, Tian H, Zhang J, Luo X, Xu D, Sun J, Zhang D, Zhang H, Zhao G, Wang W, Zheng Z, Yang S, Jin Y. Stem cell factor restrains endoplasmic reticulum stress-associated apoptosis through c-Kit receptor activation of JAK2/STAT3 axis in hippocampal neuronal cells. PLoS One 2024; 19:e0310872. [PMID: 39546459 PMCID: PMC11567559 DOI: 10.1371/journal.pone.0310872] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/07/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a common elderly disorder characterized by cognitive decline. Endoplasmic reticulum (ER) stress has been implicated in various neurodegenerative diseases, including AD. Stem cell factor (SCF) performs its biological functions by binding to and activating receptor tyrosine kinase c-Kit. We aimed to investigate the effects of SCF/c-Kit and JAK2/STAT3 on ER stress and apoptosis in AD. METHODS The study employed L-glutamic acid (L-Glu)-treated HT22 cells as sporadic AD cell model and APP/PS1 mice as an animal model of familiar AD. SCF, c-Kit inhibitor ISCK03 or JAK2/STAT3 inhibitor WP1066 was treated to verify the effects of SCF/c-Kit and JAK2/STAT3 on ER stress and apoptosis of L-Glu-exposed HT22 cells. Cell viability was assessed by MTT. BrdU detected cell proliferation. Flow cytometry measured cell apoptosis. The expression levels of ER stress markers GRP78, PERK, CHOP, and apoptosis protein caspase3 were determined by western blot. The effect on the mRNA of ER stress markers GRP78, PERK, CHOP and apoptotic caspase3 were quantified by RT-qPCR in primary cultured hippocampal neurons from APP/PS1 transgenic mice. RESULTS Administration of SCF significantly augmented the activity and proliferation of hippocampal neuronal cells, protecting cells against L-Glu induced ER stress-associated apoptosis. Moreover, the addition of ISCK03 (c-Kit inhibitor) or WP1066 (JAK2/STAT3 inhibitor) reversed SCF effects on ER stress and apoptosis in vitro. CONCLUSION We found that SCF inhibits L-Glu-induced ER stress-associated apoptosis via JAK2/STAT3 axis in HT22 hippocampal neuronal cells, as well as in primary hippocampal neurons from APP/PS1 mice, which provides valuable insights into the molecular mechanisms underlying the pathogenesis of AD and explores novel therapeutic targets for both sporadic and familial AD.
Collapse
Affiliation(s)
- Haiying Shen
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Junjie Nie
- Department of Nuclear Medicine, Jilin People’s Hospital, Jilin, Jilin Province, P.R. China
| | - Guangqing Li
- Department of Computer Application, School of Biomedical Engineering, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Hongyan Tian
- Department of Histoembryology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Jun Zhang
- Department of Histoembryology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Xiaofeng Luo
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Da Xu
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Jie Sun
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Dongfang Zhang
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Hong Zhang
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Guifang Zhao
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Weiyao Wang
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Zhonghua Zheng
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Shuyan Yang
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| | - Yuji Jin
- Department of Medical Genetics, School of Basic Medicine, Jilin Medical University, Jilin, Jilin Province, P.R. China
| |
Collapse
|
12
|
Nguyen QNS, Yoo KY, Pham TTT, Selvaraj B, Vu HT, Le TT, Lee H, Tran QL, Thuong PT, Pae AN, Jung SH, Lee JW. Neuroprotective Effects of Ethanol Extract Polyscias guilfoylei (EEPG) Against Glutamate Induced Neurotoxicity in HT22 Cells. Int J Mol Sci 2024; 25:12153. [PMID: 39596219 PMCID: PMC11595212 DOI: 10.3390/ijms252212153] [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: 09/30/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress induced by glutamate is a significant contributor to neuronal cell damage and can lead to neurodegenerative diseases such as Alzheimer's, Huntington's, and ischemic brain injury. At the cellular level, oxidative stress increases Ca2+ ion influx and reactive oxygen species (ROS), which activate the MAPK signaling pathway. Additionally, the generation of ROS causes mitochondrial dysfunction, triggering apoptosis by promoting the translocation of AIF to the nucleus from the mitochondria. The neuroprotective potential of Polyscias guilfoylei has not yet been reported. Therefore, in this study, the ethanol extract of Polyscias guilfoylei (EEPG) was examined for its protective effect against oxidative cell damage caused by glutamate in neuronal cells. EEPG treatment increased the viability of HT22 cells exposed to high concentrations of glutamate. Cellular Ca2+ ion influx and ROS generation decreased with EEPG treatment in glutamate-treated HT22 cells. EEPG treatment inhibited MAPK activation and AIF nuclear translocation. In an in vivo study, EEPG attenuated brain cell death in an ischemic brain injury rat model. This study demonstrates the potential therapeutic effects of Polyscias guilfoylei in the treatment of ischemic brain injury.
Collapse
Affiliation(s)
- Qui Ngoc Sang Nguyen
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; (Q.N.S.N.); (T.T.T.P.); (B.S.); (T.T.L.)
- Department of Anatomy, College of Dentistry and Research Institute for Dental Engineering, Gangneung Wonju National University, 7 Jukheon-gil, Gangneung 25457, Republic of Korea; (K.-Y.Y.); (H.L.)
- Institute of Natural Product Chemistry, Vietnamese Academy Science and Technology, 1H Building, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 100000, Vietnam
| | - Ki-Yeon Yoo
- Department of Anatomy, College of Dentistry and Research Institute for Dental Engineering, Gangneung Wonju National University, 7 Jukheon-gil, Gangneung 25457, Republic of Korea; (K.-Y.Y.); (H.L.)
| | - Thi Thu Trang Pham
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; (Q.N.S.N.); (T.T.T.P.); (B.S.); (T.T.L.)
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung 25451, Republic of Korea
| | - Baskar Selvaraj
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; (Q.N.S.N.); (T.T.T.P.); (B.S.); (T.T.L.)
| | - Huong Thuy Vu
- Traphaco Join-Stock Company, 75 P. Yên Ninh, Quán Thánh, Ba Đình, Hanoi 1000000, Vietnam; (H.T.V.); (Q.L.T.)
- Faculty of Herbal Medicine, Traditional Pharmacy, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Ba Dinh, Hanoi 100000, Vietnam
| | - Tam Thi Le
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; (Q.N.S.N.); (T.T.T.P.); (B.S.); (T.T.L.)
| | - Heesu Lee
- Department of Anatomy, College of Dentistry and Research Institute for Dental Engineering, Gangneung Wonju National University, 7 Jukheon-gil, Gangneung 25457, Republic of Korea; (K.-Y.Y.); (H.L.)
| | - Quang Luc Tran
- Traphaco Join-Stock Company, 75 P. Yên Ninh, Quán Thánh, Ba Đình, Hanoi 1000000, Vietnam; (H.T.V.); (Q.L.T.)
| | - Phuong Thien Thuong
- Division of Biotechnology, Vietnam Korea Institute of Science and Technology, Hoa lac High-tech Park, km29 Thang Long Boulevard, Hanoi 100000, Vietnam;
| | - Ae Nim Pae
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea;
- Center for Brain Disorders, Brain Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sang Hoon Jung
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; (Q.N.S.N.); (T.T.T.P.); (B.S.); (T.T.L.)
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung 25451, Republic of Korea
| | - Jae Wook Lee
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; (Q.N.S.N.); (T.T.T.P.); (B.S.); (T.T.L.)
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung 25451, Republic of Korea
| |
Collapse
|
13
|
Zhang W, Sun H, Zhao W, Li J, Meng H. Suppression of JNK pathway protects neurons from oxidative injury via attenuating parthanatos in glutamate-treated HT22 neurons. Sci Rep 2024; 14:25793. [PMID: 39468165 PMCID: PMC11519538 DOI: 10.1038/s41598-024-76640-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
Oxidative stress causes diverse neurological disorders. Parthanatos is a type of programmed cell death, characterised by strong activation of poly (ADP-ribose) (PAR) polymerase-1 (PARP-1), PAR polymer accumulation, and nuclear translocation of apoptosis-inducing factor (AIF), and is involved in cellular oxidative injury. Signalling by c-Jun-N-terminal protein kinase (JNK) is activated by reactive oxygen species (ROS), and this also contributes to ROS production. However, the exact relationship between JNK signalling and parthanatos in neurological disorders triggered by oxidative stress is unclear. In this study, glutamate-treated HT22 neurons were used to investigate whether the signalling by JNK contributes a regulatory role to parthanatos in oxidative stress-related neurological disease. JNK signalling was activated in glutamate-treated HT22 neurons, demonstrated via upregulation of p-JNK levels. Pre-treatment with SP600125 markedly inhibited JNK signalling, increased cell viability, and significantly reversed PARP-1 overproduction, PAR polymer accumulation, and nuclear AIF translocation. In addition, inhibition of JNK signalling severely reduced the production of both intracellular ROS and mitochondria superoxide. This study indicated that parthanatos in glutamate-treated HT22 neurons could be suppressed by JNK signalling inhibition. JNK activation participated in parthanatos via an increase in intracellular ROS levels.
Collapse
Affiliation(s)
- Wuqiong Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Huaiyu Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Weixuan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Jiaai Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Hongmei Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
14
|
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
|
15
|
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: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
16
|
Yang Y, Zhou ZD, Yi L, Tan BJW, Tan EK. Interaction between caffeine consumption & genetic susceptibility in Parkinson's disease: A systematic review. Ageing Res Rev 2024; 99:102381. [PMID: 38914264 DOI: 10.1016/j.arr.2024.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Caffeine is one of the most consumed psychoactive substances globally. Caffeine-gene interactions in Parkinson's disease (PD) has not been systematically examined. OBJECTIVES To conduct a systematic review on the interaction between caffeine consumption and genetic susceptibility to PD. METHODOLOGY We conducted PubMed and Embase search using terms "Genetic association studies", "Caffeine", "polymorphism" and "Parkinson's disease", from inception till 2023. Of the initial 2391 studies, 21 case-control studies were included. The demographic, genetic and clinical data were extracted and analyzed. RESULTS We identified 21 studies which involved a total of 607,074 study subjects and 17 gene loci (SNCA, MAPT, HLA-DRA, NOS1, NOS3, GBA, ApoE, BST1, ESR2, NAT2, SLC2A13, LRRK2, NOS2A, GRIN2A, CYP1A2, ESR1, ADORA2A) have been investigated for the effect of gene-caffeine interaction and PD risk. The genes were identified through PD GWAS or involved in caffeine or related metabolism pathways. Based on the genetic association and interaction studies, only MAPT, SLC2A13, LRRK2, ApoE, NOS2A, GRIN2A, CYP1A2, and ADORA2A have been shown by at least one study to have a positive caffeine-gene interaction influencing the risk of PD. CONCLUSION Studies have shown an interaction between caffeine with genetic variants of MAPT, SLC2A13, LRRK2, ApoE, NOS2A, GRIN2A, CYP1A2, and ADORA2A in modulating the risk of PD. Due to the potential limitations of these discovery/pilot studies, further independent replication studies are needed. Better designed genetic association studies in multi-ancestry and admixed cohorts to identify potential shared or unique multivariate gene-environmental interactions, as well as functional studies of gene-caffeine interactions will be useful.
Collapse
Affiliation(s)
- Yujuan Yang
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore.
| | - Zhi Dong Zhou
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore.
| | - Lingxiao Yi
- Department of Neurology, National Neuroscience Institute, Singapore.
| | | | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore.
| |
Collapse
|
17
|
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
|
18
|
Yan F, Zhang L, Duan L, Li L, Liu X, Liu Y, Qiao T, Zeng Y, Fang H, Wu D, Wang X. Roles of glutamic pyruvate transaminase 2 in reprogramming of airway epithelial lipidomic and metabolomic profiles after smoking. Clin Transl Med 2024; 14:e1679. [PMID: 38706045 PMCID: PMC11070440 DOI: 10.1002/ctm2.1679] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Metabolic abnormalities represent one of the pathological features of chronic obstructive pulmonary disease (COPD). Glutamic pyruvate transaminase 2 (GPT2) is involved in glutamate metabolism and lipid synthesis pathways, whilst the exact roles of GPT2 in the occurrence and development of COPD remains uncertain. This study aims at investigating how GPT2 and the associated genes modulate smoking-induced airway epithelial metabolism and damage by reprogramming lipid synthesis. The circulating or human airway epithelial metabolomic and lipidomic profiles of COPD patients or cell-lines explored with smoking were assessed to elucidate the pivotal roles of GPT2 in reprogramming processes. We found that GPT2 regulate the reprogramming of lipid metabolisms caused by smoking, especially phosphatidylcholine (PC) and triacylglycerol (TAG), along with changes in the expression of lipid metabolism-associated genes. GPT2 modulated cell sensitivities and survival in response to smoking by enhancing mitochondrial functions and maintaining lipid and energy homeostasis. Our findings provide evidence for the involvement of GPT2 in the reprogramming of airway epithelial lipids following smoking, as well as the molecular mechanisms underlying GPT2-mediated regulation, which may offer an alternative of therapeutic strategies for chronic lung diseases.
Collapse
Affiliation(s)
- Furong Yan
- Center for Tumor Diagnosis & TherapyJinshan HospitalFudan UniversityShanghaiChina
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
- Center of Molecular Diagnosis and TherapyThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Linlin Zhang
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Lian Duan
- Department of Pediatric SurgeryFaculty of Pediatricsthe Seventh Medical Center of PLA General HospitalBeijingChina
| | - Liyang Li
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Xuanqi Liu
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Yifei Liu
- Center of Molecular Diagnosis and TherapyThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Tiankui Qiao
- Center for Tumor Diagnosis & TherapyJinshan HospitalFudan UniversityShanghaiChina
| | - Yiming Zeng
- Center of Molecular Diagnosis and TherapyThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Hao Fang
- Department of AnesthesiologyShanghai Geriatic Medical CenterShanghaiChina
- Department of AnesthesiologyZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Duojiao Wu
- Center for Tumor Diagnosis & TherapyJinshan HospitalFudan UniversityShanghaiChina
| | - Xiangdong Wang
- Center for Tumor Diagnosis & TherapyJinshan HospitalFudan UniversityShanghaiChina
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
19
|
Wang H, Hou MJ, Liao L, Li P, Chen T, Wang P, Zhu BT. Strong Protection by 4-Hydroxyestrone against Erastin-Induced Ferroptotic Cell Death in Estrogen Receptor-Negative Human Breast Cancer Cells: Evidence for Protein Disulfide Isomerase as a Mechanistic Target for Protection. Biochemistry 2024; 63:984-999. [PMID: 38569593 PMCID: PMC11025120 DOI: 10.1021/acs.biochem.3c00261] [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: 05/17/2023] [Revised: 12/31/2023] [Accepted: 01/03/2024] [Indexed: 04/05/2024]
Abstract
Ferroptosis is a recently identified form of regulated cell death, characterized by excessive iron-dependent lipid peroxidation. Recent studies have demonstrated that protein disulfide isomerase (PDI) is an important mediator of chemically induced ferroptosis and also a new target for protection against ferroptosis-associated cell death. In the present study, we identified that 4-hydroxyestrone (4-OH-E1), a metabolic derivative of endogenous estrogen, is a potent small-molecule inhibitor of PDI, and can strongly protect against chemically induced ferroptotic cell death in the estrogen receptor-negative MDA-MB-231 human breast cancer cells. Pull-down and CETSA assays demonstrated that 4-OH-E1 can directly bind to PDI both in vitro and in intact cells. Computational modeling analysis revealed that 4-OH-E1 forms two hydrogen bonds with PDI His256, which is essential for its binding interaction and thus inhibition of PDI's catalytic activity. Additionally, PDI knockdown attenuates the protective effect of 4-OH-E1 as well as cystamine (a known PDI inhibitor) against chemically induced ferroptosis in human breast cancer cells. Importantly, inhibition of PDI by 4-OH-E1 and cystamine or PDI knockdown by siRNAs each markedly reduces iNOS activity and NO accumulation, which has recently been demonstrated to play an important role in erastin-induced ferroptosis. In conclusion, this study demonstrates that 4-OH-E1 is a novel inhibitor of PDI and can strongly inhibit ferroptosis in human breast cancer cells in an estrogen receptor-independent manner. The mechanistic understanding gained from the present study may also aid in understanding the estrogen receptor-independent cytoprotective actions of endogenous estrogen metabolites in many noncancer cell types.
Collapse
Affiliation(s)
- Hongge Wang
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
- School
of Life Sciences, University of Science
and Technology of China, Hefei, Anhui 230026, China
| | - Ming-Jie Hou
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
| | - Lixi Liao
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
| | - Peng Li
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
| | - Tongxiang Chen
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
| | - Pan Wang
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
| | - Bao Ting Zhu
- Shenzhen
Key Laboratory of Steroid Drug Discovery and Development, School of
Medicine, The Chinese University of Hong
Kong, Shenzhen 518172, China
- Shenzhen
Bay Laboratory, Shenzhen 518055, China
| |
Collapse
|
20
|
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
|
21
|
Campos J, Gleitze S, Hidalgo C, Núñez MT. IP 3R-Mediated Calcium Release Promotes Ferroptotic Death in SH-SY5Y Neuroblastoma Cells. Antioxidants (Basel) 2024; 13:196. [PMID: 38397794 PMCID: PMC10886377 DOI: 10.3390/antiox13020196] [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: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death pathway that involves the depletion of intracellular glutathione (GSH) levels and iron-mediated lipid peroxidation. Ferroptosis is experimentally caused by the inhibition of the cystine/glutamate antiporter xCT, which depletes cells of GSH, or by inhibition of glutathione peroxidase 4 (GPx4), a key regulator of lipid peroxidation. The events that occur between GPx4 inhibition and the execution of ferroptotic cell death are currently a matter of active research. Previous work has shown that calcium release from the endoplasmic reticulum (ER) mediated by ryanodine receptor (RyR) channels contributes to ferroptosis-induced cell death in primary hippocampal neurons. Here, we used SH-SY5Y neuroblastoma cells, which do not express RyR channels, to test if calcium release mediated by the inositol 1,4,5-trisphosphate receptor (IP3R) channel plays a role in this process. We show that treatment with RAS Selective Lethal Compound 3 (RSL3), a GPx4 inhibitor, enhanced reactive oxygen species (ROS) generation, increased cytoplasmic and mitochondrial calcium levels, increased lipid peroxidation, and caused cell death. The RSL3-induced calcium signals were inhibited by Xestospongin B, a specific inhibitor of the ER-resident IP3R calcium channel, by decreasing IP3R levels with carbachol and by IP3R1 knockdown, which also prevented the changes in cell morphology toward roundness induced by RSL3. Intracellular calcium chelation by incubation with BAPTA-AM inhibited RSL3-induced calcium signals, which were not affected by extracellular calcium depletion. We propose that GPx4 inhibition activates IP3R-mediated calcium release in SH-SY5Y cells, leading to increased cytoplasmic and mitochondrial calcium levels, which, in turn, stimulate ROS production and induce lipid peroxidation and cell death in a noxious positive feedback cycle.
Collapse
Affiliation(s)
- Joaquín Campos
- Chica and Heinz Schaller Foundation, Institute for Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Silvia Gleitze
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (S.G.); (C.H.)
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (S.G.); (C.H.)
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
- Physiology and Biophysics Program, Institute of Biomedical Sciences and Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
| | - Marco T. Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago 7800024, Chile
| |
Collapse
|
22
|
R R, Devtalla H, Rana K, Panda SP, Agrawal A, Kadyan S, Jindal D, Pancham P, Yadav D, Jha NK, Jha SK, Gupta V, Singh M. A comprehensive update on genetic inheritance, epigenetic factors, associated pathology, and recent therapeutic intervention by gene therapy in schizophrenia. Chem Biol Drug Des 2024; 103:e14374. [PMID: 37994213 DOI: 10.1111/cbdd.14374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/15/2023] [Accepted: 09/29/2023] [Indexed: 11/24/2023]
Abstract
Schizophrenia is a severe psychological disorder in which reality is interpreted abnormally by the patient. The symptoms of the disease include delusions and hallucinations, associated with extremely disordered behavior and thinking, which may affect the daily lives of the patients. Advancements in technology have led to understanding the dynamics of the disease and the identification of the underlying causes. Multiple investigations prove that it is regulated genetically, and epigenetically, and is affected by environmental factors. The molecular and neural pathways linked to the regulation of schizophrenia have been extensively studied. Over 180 Schizophrenic risk loci have now been recognized due to several genome-wide association studies (GWAS). It has been observed that multiple transcription factors (TF) binding-disrupting single nucleotide polymorphisms (SNPs) have been related to gene expression responsible for the disease in cerebral complexes. Copy number variation, SNP defects, and epigenetic changes in chromosomes may cause overexpression or underexpression of certain genes responsible for the disease. Nowadays, gene therapy is being implemented for its treatment as several of these genetic defects have been identified. Scientists are trying to use viral vectors, miRNA, siRNA, and CRISPR technology. In addition, nanotechnology is also being applied to target such genes. The primary aim of such targeting was to either delete or silence such hyperactive genes or induce certain genes that inhibit the expression of these genes. There are challenges in delivering the gene/DNA to the site of action in the brain, and scientists are working to resolve the same. The present article describes the basics regarding the disease, its causes and factors responsible, and the gene therapy solutions available to treat this disease.
Collapse
Affiliation(s)
- Rachana R
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Harshit Devtalla
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Karishma Rana
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Arushi Agrawal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Shreya Kadyan
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Divya Jindal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
- IIT Bombay Monash Research Academy, IIT - Bombay, Bombay, India
| | - Pranav Pancham
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Deepshikha Yadav
- Bhartiya Nirdeshak Dravya Division, CSIR-National Physical Laboratory, New Delhi, India
- Physico-Mechanical Metrology Division, CSIR-National Physical Laboratory, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Niraj Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology (SSET), Sharda University, Greater Noida, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
- Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology (SSET), Sharda University, Greater Noida, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
- Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun, India
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Vivek Gupta
- Macquarie Medical School, Macquarie University (MQU), Sydney, New South Wales, Australia
| | - Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
- Faculty of Health, Graduate School of Public Health, University of Technology Sydney, Sydney, New South Wales, Australia
- Australian Research Consortium in Complementary and Integrative Medicine (ARCCIM), University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
23
|
Xie D, Song C, Qin T, Zhai Z, Cai J, Dai J, Sun T, Xu Y. Moschus ameliorates glutamate-induced cellular damage by regulating autophagy and apoptosis pathway. Sci Rep 2023; 13:18586. [PMID: 37903904 PMCID: PMC10616123 DOI: 10.1038/s41598-023-45878-7] [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: 11/16/2022] [Accepted: 10/25/2023] [Indexed: 11/01/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, causes short-term memory and cognition declines. It is estimated that one in three elderly people die from AD or other dementias. Chinese herbal medicine as a potential drug for treating AD has gained growing interest from many researchers. Moschus, a rare and valuable traditional Chinese animal medicine, was originally documented in Shennong Ben Cao Jing and recognized for its properties of reviving consciousness/resuscitation. Additionally, Moschus has the efficacy of "regulation of menstruation with blood activation, relief of swelling and pain" and is used for treating unconsciousness, stroke, coma, and cerebrovascular diseases. However, it is uncertain whether Moschus has any protective effect on AD patients. We explored whether Moschus could protect glutamate (Glu)-induced PC12 cells from cellular injury and preliminarily explored their related action mechanisms. The chemical compounds of Moschus were analyzed and identified by GC-MS. The Glu-induced differentiated PC12 cell model was thought to be the common AD cellular model. The study aims to preliminarily investigate the intervention effect of Moschus on Glu-induced PC12 cell damage as well as their related action mechanisms. Cell viability, lactate dehydrogenase (LDH), mitochondrial reactive oxygen species, mitochondrial membrane potential (MMP), cell apoptosis, autophagic vacuoles, autolysosomes or autophagosomes, proteins related to apoptosis, and the proteins related to autophagy were examined and analyzed. Seventeen active compounds of the Moschus sample were identified based on GC-MS analysis. In comparison to the control group, Glu stimulation increased cell viability loss, LDH release, mitochondrial damage, loss of MMP, apoptosis rate, and the number of cells containing autophagic vacuoles, and autolysosomes or autophagosomes, while these results were decreased after the pretreatment with Moschus and 3-methyladenine (3-MA). Furthermore, Glu stimulation significantly increased cleaved caspase-3, Beclin1, and LC3II protein expression, and reduced B-cell lymphoma 2/BAX ratio and p62 protein expression, but these results were reversed after pretreatment of Moschus and 3-MA. Moschus has protective activity in Glu-induced PC12 cell injury, and the potential mechanism might involve the regulation of autophagy and apoptosis. Our study may promote research on Moschus in the field of neurodegenerative diseases, and Moschus may be considered as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Caiyou Song
- 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
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jie Cai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jingyi Dai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, 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 Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
24
|
Bagán A, Abás S, Palà-Pujadas J, Irisarri A, Griñán-Ferré C, Pallàs M, Muneta-Arrate I, Muguruza C, Callado LF, Pérez B, Molins E, Morales-García JÁ, Escolano C. Exploring the reactivity of bicyclic α-iminophosphonates to access new imidazoline I 2 receptor ligands. Bioorg Chem 2023; 142:106935. [PMID: 39492364 DOI: 10.1016/j.bioorg.2023.106935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/05/2024]
Abstract
Recent studies pointed out the modulation of imidazoline I2 receptors (I2-IR) by selective ligands as a putative strategy to face neurodegenerative diseases. Foregoing the classical 2-imidazoline/imidazole-containing I2-IR ligands, we report a family of bicyclic α-iminophosphonates endowed with high affinity and selectivity upon I2-IR and we advanced a representative compound B06 in preclinical phases. In this paper, we describe the synthetic possibilities of bicyclic α-iminophosphonates by exploring its ambivalent reactivity, leading to unprecedented molecules that showed promising activities as I2-IR ligands in human brain tissues and good BBB permeation capabilities. After in silico ADME prediction studies, we assessed the neuroprotective properties of selected compounds and beneficial effect in an in vitro model of Alzheimeŕs and Parkinson's disease. Along with their neuroprotective effect, compounds showed a potent anti-inflammatory response when evaluated in a neuroinflammation cellular model. Moreover, this is the first time that the neuroprotective effects of imidazoline I2-IR ligands in a transgenic Alzheimer's disease Caenorhabditis elegans strain are investigated. Using a thrashing assay, we found a significant cognition improvement in this in vivo model after treatment with the new bicyclic α-phosphoprolines. Therefore, our results confirmed the need of exploring structurally new I2-IR ligands and their potential for therapeutic strategies in neurodegeneration.
Collapse
Affiliation(s)
- Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Judith Palà-Pujadas
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Alba Irisarri
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31. E-08028, Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31. E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31. E-08028, Barcelona, Spain
| | - Itziar Muneta-Arrate
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Carolina Muguruza
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Biocruces Bizkaia Health Research Institute Bizkaia, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology. Autonomous University of Barcelona, E-08193 Cerdanyola, Spain
| | - Elies Molins
- Institut de Ciència de Materials de Barcelona (CSIC), Campus UAB, E-08193 Cerdanyola, Spain
| | - José Á Morales-García
- Department of Cell Biology. School of Medicine, Complutense University (UCM), 28040 Madrid, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
25
|
Zhu Z, Yang P, Jia Y, Wang Y, Shi M, Zhong C, Peng H, Sun L, Guo D, Xu Q, Chen J, Wang A, Xu T, He J, Zhang Y. Plasma Amino Acid Neurotransmitters and Ischemic Stroke Prognosis: A Multicenter Prospective Study. Am J Clin Nutr 2023; 118:754-762. [PMID: 37793742 DOI: 10.1016/j.ajcnut.2023.06.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/05/2023] [Accepted: 06/12/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Plasma amino acid neurotransmitter dysregulation is suggested to be implicated in the development of ischemic stroke, but its prognostic value for ischemic stroke remains controversial. OBJECTIVE We aimed to prospectively investigate the associations between plasma amino acid neurotransmitters levels and adverse outcomes after ischemic stroke in a large-scale multicenter cohort study. METHODS We measured 4 plasma amino acid neurotransmitters (glutamic acid, aspartic acid, gamma-aminobutyric acid, and glycine) among 3486 patients with ischemic stroke from 26 hospitals across China. The primary outcome is the composite outcome of death or major disability (modified Rankin Scale score ≥3) at 3 mo after ischemic stroke. RESULTS After multivariate adjustment, the odds ratios of death or major disability for the highest versus the lowest quartile were 2.04 (95% confidence interval [CI]: 1.60,2.59; P-trend < 0.001) for glutamic acid, 2.03 (95% CI: 1.59, 2.59; P-trend < 0.001) for aspartic acid, 1.35 (95% CI: 1.06, 1.71; P-trend = 0.016) for gamma-aminobutyric acid, and 0.54 (95% CI: 0.42, 0.69; P-trend < 0.001) for glycine. Each standard deviation increment of log-transformed glutamic acid, aspartic acid, gamma-aminobutyric acid, and glycine was associated with a 34%, 34%, and 9% increased risk, and a 23% decreased risk of death or major disability, respectively (all P < 0.05), in a linear fashion as indicated by spline regression analyses (all P for linearity < 0.05). Addition of the 4 plasma amino acid neurotransmitters to conventional risk factors significantly improved the risk reclassification, as evidenced by integrated discrimination improvement and net reclassification improvement (all P < 0.05). CONCLUSIONS Increased glutamic acid, aspartic acid, and gamma-aminobutyric acid and decreased glycine in plasma are associated with adverse outcomes after ischemic stroke, suggesting that plasma amino acid neurotransmitters may be potential intervention targets for improving prognosis of ischemic stroke. The CATIS trial was registered at clinicaltrials.gov (registration number: NCT01840072; URL: ===https://clinicaltrials.gov/ct2/show/NCT01840072?cond=NCT01840072&draw=2&rank=1).
Collapse
Affiliation(s)
- Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China; Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yinan Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China; Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China; Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Lulu Sun
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Daoxia Guo
- School of Nursing, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qingyun Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
26
|
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
|
27
|
Yang X, Wang Z, Zandkarimi F, Liu Y, Duan S, Li Z, Kon N, Zhang Z, Jiang X, Stockwell BR, Gu W. Regulation of VKORC1L1 is critical for p53-mediated tumor suppression through vitamin K metabolism. Cell Metab 2023; 35:1474-1490.e8. [PMID: 37467745 PMCID: PMC10529626 DOI: 10.1016/j.cmet.2023.06.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/23/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023]
Abstract
Here, we identified vitamin K epoxide reductase complex subunit 1 like 1 (VKORC1L1) as a potent ferroptosis repressor. VKORC1L1 protects cells from ferroptosis by generating the reduced form of vitamin K, a potent radical-trapping antioxidant, to counteract phospholipid peroxides independent of the canonical GSH/GPX4 mechanism. Notably, we found that VKORC1L1 is also a direct transcriptional target of p53. Activation of p53 induces downregulation of VKORC1L1 expression, thus sensitizing cells to ferroptosis for tumor suppression. Interestingly, a small molecular inhibitor of VKORC1L1, warfarin, is widely prescribed as an FDA-approved anticoagulant drug. Moreover, warfarin represses tumor growth by promoting ferroptosis in both immunodeficient and immunocompetent mouse models. Thus, by downregulating VKORC1L1, p53 executes the tumor suppression function by activating an important ferroptosis pathway involved in vitamin K metabolism. Our study also reveals that warfarin is a potential repurposing drug in cancer therapy, particularly for tumors with high levels of VKORC1L1 expression.
Collapse
Affiliation(s)
- Xin Yang
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhe Wang
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fereshteh Zandkarimi
- Department of Chemistry, Columbia University, New York, NY, USA; Mass Spectrometry Core Facility, Columbia University, New York, NY, USA
| | - Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Shoufu Duan
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhiming Li
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ning Kon
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhiguo Zhang
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pediatrics, and Department of Genetics and Development, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
28
|
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: 15] [Impact Index Per Article: 7.5] [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
|
29
|
Josifovska S, Panov S, Hadzi-Petrushev N, Mitrokhin V, Kamkin A, Stojchevski R, Avtanski D, Mladenov M. Positive Tetrahydrocurcumin-Associated Brain-Related Metabolomic Implications. Molecules 2023; 28:3734. [PMID: 37175144 PMCID: PMC10179939 DOI: 10.3390/molecules28093734] [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: 02/10/2023] [Revised: 03/22/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Tetrahydrocurcumin (THC) is a metabolite of curcumin (CUR). It shares many of CUR's beneficial biological activities in addition to being more water-soluble, chemically stable, and bioavailable compared to CUR. However, its mechanisms of action have not been fully elucidated. This paper addresses the preventive role of THC on various brain dysfunctions as well as its effects on brain redox processes, traumatic brain injury, ischemia-reperfusion injury, Alzheimer's disease, and Parkinson's disease in various animal or cell culture models. In addition to its strong antioxidant properties, the effects of THC on the reduction of amyloid β aggregates are also well documented. The therapeutic potential of THC to treat patterns of mitochondrial brain dysmorphic dysfunction is also addressed and thoroughly reviewed, as is evidence from experimental studies about the mechanism of mitochondrial failure during cerebral ischemia/reperfusion injury. THC treatment also results in a dose-dependent decrease in ERK-mediated phosphorylation of GRASP65, which prevents further compartmentalization of the Golgi apparatus. The PI3K/AKT signaling pathway is possibly the most involved mechanism in the anti-apoptotic effect of THC. Overall, studies in various animal models of different brain disorders suggest that THC can be used as a dietary supplement to protect against traumatic brain injury and even improve brain function in Alzheimer's and Parkinson's diseases. We suggest further preclinical studies be conducted to demonstrate the brain-protective, anti-amyloid, and anti-Parkinson effects of THC. Application of the methods used in the currently reviewed studies would be useful and should help define doses and methods of THC administration in different disease conditions.
Collapse
Affiliation(s)
- Slavica Josifovska
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Sasho Panov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Vadim Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| | - Andre Kamkin
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| |
Collapse
|
30
|
Neuroprotective Effects of Ethanol Extract of Polyscias fruticosa (EEPF) against Glutamate-Mediated Neuronal Toxicity in HT22 Cells. Int J Mol Sci 2023; 24:ijms24043969. [PMID: 36835378 PMCID: PMC9959701 DOI: 10.3390/ijms24043969] [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: 01/11/2023] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
In traditional herbal medicine, the Polyscias fruticosa has been frequently used for the treatment of ischemia and inflammation. Oxidative stress mediated by elevated glutamate levels cause neuronal cell death in ischemia and various neurodegenerative diseases. However, so far, the neuroprotective effects of this plant extract against glutamate-mediated cell death have not been investigated in cell models. The current study investigates the neuroprotective effects of ethanol extracts of Polyscias fruticosa (EEPF) and elucidates the underlying molecular mechanisms of EEPFs relevant to neuroprotection against glutamate-mediated cell death. The oxidative stress-mediated cell death was induced by 5 mM glutamate treatment in HT22 cells. The cell viability was measured by a tetrazolium-based EZ-Cytox reagent and Calcein-AM fluorescent dye. Intracellular Ca2+ and ROS levels were measured by fluorescent dyes, fluo-3 AM and 2',7'-dichlorodihydrofluorescein diacetate (DCF-DA), respectively. Protein expressions of p-AKT, BDNF, p-CREB, Bax, Bcl-2, and apoptosis-inducing factor (AIF) were determined by western blot analysis. The apoptotic cell death was measured by flow cytometry. The in vivo efficacy of EEPF was evaluated using the Mongolian gerbil mouse by surgery-induced brain ischemia. EEPF treatment showed a neuroprotective effect against glutamate-induced cell death. The EEPF co-treatment reduced the intracellular Ca2+ and ROS and apoptotic cell death. Furthermore, it recovered the p-AKT, p-CREB, BDNF, and Bcl-2 levels decreased by glutamate. The EEPF co-treatment suppressed the activation of apoptotic Bax, the nuclear translocation of AIF, and mitogen-activated protein kinase (MAPK) pathway proteins (ERK1/2, p38, JNK). Further, EEPF treatment significantly rescued the degenerative neurons in the ischemia-induced Mongolian gerbil in vivo model. EEPF exhibited neuroprotective properties that suppress glutamate-mediated neurotoxicity. The underlying mechanism of EEPF is increasing the level of p-AKT, p-CREB, BDNF, and Bcl-2 associated with cell survival. It has therapeutic potential for the treatment of glutamate-mediated neuropathology.
Collapse
|
31
|
Jeong EK, Selvaraj B, Clovis S, Son YJ, Park TH, Veeramanoharan A, Kim HI, Yoo KY, Lee JW, Park CM. Synthesis and neuroprotective effects of H 2S-donor-peptide hybrids on hippocampal neuronal cells. Free Radic Biol Med 2023; 194:316-325. [PMID: 36528123 DOI: 10.1016/j.freeradbiomed.2022.12.012] [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: 11/05/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S) has emerged as an endogenous signaling molecule that functions in many physiological and pathological processes of human cells in health and disease, including neuromodulation and neuroprotection, inflammation, angiogenesis, and vasorelaxation. The limited clinical applications of current H2S donors have led to the development of H2S donor hybrid compounds that combine current H2S donors with bioactive molecules. Finely tuned multi-targeting hybrid molecules have been shown to have complementary neuroprotective effects against reactive oxygen species (ROS)-induced oxidative stress. In this study, we developed hybrid molecules combining a dithiolethione-based slow-releasing H2S donor that exerts neuroprotective effects, with the tripeptides glycyl-L-histidyl-l-lysine (GHK) and L-alanyl-L-cystinyl-l-glutamine (ACQ), two natural products that exhibit powerful antioxidant effects. In particular, a hybrid combination of a dithiolethione-based slow-releasing H2S donor and ACQ exhibited significant neuroprotective effects against glutamate-induced oxidative damage in HT22 hippocampal neuronal cells. This hybrid remarkably suppressed Ca2+ accumulation and ROS production. Furthermore, it efficiently inhibited apoptotic neuronal cell death by blocking apoptosis-inducing factor release and its translocation to the nucleus. These results indicate that the hybrid efficiently inhibited apoptotic neuronal cell damage by complementary neuroprotective actions.
Collapse
Affiliation(s)
- Eui Kyun Jeong
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea
| | - Baskar Selvaraj
- Natural Product Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangneung, Gangwon, 25451, South Korea
| | - Shyaka Clovis
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea
| | - Yun Jeong Son
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea
| | - Tae Hoo Park
- Natural Product Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangneung, Gangwon, 25451, South Korea
| | - Ashokkumar Veeramanoharan
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea
| | - Hoe-In Kim
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea
| | - Ki-Yeon Yoo
- Department of Anatomy, College of Dentistry, Reseach Institute of Oral Sciences, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea
| | - Jae Wook Lee
- Natural Product Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangneung, Gangwon, 25451, South Korea.
| | - Chung-Min Park
- Department of Chemistry, Gangneung-Wonju National University, Gangneung, Gangwon, 25457, South Korea.
| |
Collapse
|
32
|
Germinated brown rice protects against glutamate toxicity in HT22 hippocampal neurons through the jnk-mediated apoptotic pathway via the GABA A receptor. IBRO Neurosci Rep 2022; 14:38-49. [PMID: 36590249 PMCID: PMC9800259 DOI: 10.1016/j.ibneur.2022.12.004] [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: 07/23/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The anti-apoptosis effect of germinated brown rice (GBR) focusing on differentiated HT22 cells results in improved nutritional values after the germination process of GBR which contains total phenolic compounds and γ-aminobutyric acid (GABA). Cell death induced by 5 mM glutamate was investigated for 24 h to determine whether GBR mediates cell death through GABA receptors by using antagonists. The results showed that GBR (100 µg/ml) suppressed glutamate-induced cytotoxicity and caused arrest at the G1/S phase of the cell cycle in differentiated HT22 cells. Furthermore, GBR significantly decreased the expression level of c-Jun, while its active form, p-c-Jun, is the downstream product of the JNK-mediated apoptotic pathway and causes subsequent cell death. In addition, bicuculline (12.5 nM), a GABAA antagonist, could eliminate GBR effects, but phaclofen (1 mM), a GABAB antagonist, could not. Surprisingly, GBR exhibited a better neuroprotective effect than a pure commercial GABA compound (0.115 µM). These results indicated that GBR possessed high anti-apoptotic activity and inhibited cell death in differentiated HT22 cells by perturbing re-entry of the cell cycle and apoptosis via the GABAA receptor. Hence, GBR could be further used as a valuable nutritional compound to prevent apoptosis-induced neurodegenerative diseases.
Collapse
Key Words
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid
- Apoptosis
- Bic, bicuculline
- Cell cycle
- Differentiated HT22 cells
- GABA, gamma-aminobutyric acid
- GABAA receptor
- GABRG2, GABAA receptor (γ2 subunit)
- GBR
- GBR, germinated brown rice
- Glu, glutamate
- HT22, mouse hippocampal neuronal cell line
- JNKs
- JNKs, c-Jun N-terminal kinases
- MAPKs, mitogen-activated protein kinase
- NMDA, N-methyl-D-aspartate receptors
- Pha, phaclofen
- ROS, reactive oxygen species
Collapse
|
33
|
Han EJ, Zhang C, Kim HS, Kim JY, Park SM, Jung WK, Ahn G, Cha SH. Sargachromenol Isolated from Sargassum horneri Attenuates Glutamate-Induced Neuronal Cell Death and Oxidative Stress through Inhibition of MAPK/NF-κB and Activation of Nrf2/HO-1 Signaling Pathway. Mar Drugs 2022; 20:710. [PMID: 36421988 PMCID: PMC9695719 DOI: 10.3390/md20110710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 10/29/2023] Open
Abstract
Oxidative stress-induced neuronal cell loss is considered to be the major mechanism underlying the pathogenesis of neurodegenerative diseases, which could be induced by a high concentration of glutamate. In this study, sargachromenol (SC) was isolated from a marine brown seaweed Sargassum horneri (S. horneri) and its neuroprotective effects against glutamate-induced oxidative stress in HT22 cells were investigated. An MTT assay was applied to assess the cytotoxicity of the SC, and the efficacies of SC were determined by flow cytometry, an analysis of ROS production, quantitative Real-Time PCR, and the Western blot assay. Our results showed that the pretreatment of SC reduced glutamate-induced apoptosis in HT22 cells via inhibiting the sub-G1 population, DNA fragmentation, and nuclear condensation, as well as up-regulating anti-apoptotic protein (Bcl-2) and down-regulating apoptotic proteins (Bax, p53, cleaved-PARP, caspase-3, caspase-9, and cytochrome c). Additionally, SC attenuated glutamate-induced oxidative stress by suppressing mitogen-activated protein kinases (MAPKs;ERK, JNK, and p38) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling (IκBα and NF-κB p65), while activating nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signaling (Nrf2; HO-1, and NQO-1). Our results suggest that SC could be used as a pharmacological candidate for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Eui-Jeong Han
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea
| | - Chunying Zhang
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 32158, Korea
| | - Hyun-Soo Kim
- National Marine Biodiversity Institute of Korea, Seocheon-kun 33662, Korea
| | - Ji-Yul Kim
- National Marine Biodiversity Institute of Korea, Seocheon-kun 33662, Korea
| | - Sang-Muyn Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Won-Kyo Jung
- Research Center for Marine Integrated Bionics Technology and Marine Integrated Biomedical Technology Center, Pukyong National University, Busan 48513, Korea
- Department of Biomedical Engineering, New Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Korea
| | - Ginnae Ahn
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 32158, Korea
| |
Collapse
|
34
|
Choi HJ, Chen TX, Hou MJ, Song JH, Li P, Liu CF, Wang P, Zhu BT. Protection against glutathione depletion-associated oxidative neuronal death by neurotransmitters norepinephrine and dopamine: Protein disulfide isomerase as a mechanistic target for neuroprotection. Acta Pharmacol Sin 2022; 43:2527-2541. [PMID: 35347247 PMCID: PMC9525605 DOI: 10.1038/s41401-022-00891-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/17/2022] [Indexed: 12/11/2022]
Abstract
Oxidative stress is extensively involved in neurodegeneration. Clinical evidence shows that keeping the mind active through mentally-stimulating physical activities can effectively slow down the progression of neurodegeneration. With increased physical activities, more neurotransmitters would be released in the brain. In the present study, we investigated whether some of the released neurotransmitters might have a beneficial effect against oxidative neurodegeneration in vitro. Glutamate-induced, glutathione depletion-associated oxidative cytotoxicity in HT22 mouse hippocampal neuronal cells was used as an experimental model. We showed that norepinephrine (NE, 50 µM) or dopamine (DA, 50 µM) exerted potent protective effect against glutamate-induced cytotoxicity, but this effect was not observed when other neurotransmitters such as histamine, γ-aminobutyric acid, serotonin, glycine and acetylcholine were tested. In glutamate-treated HT22 cells, both NE and DA significantly suppressed glutathione depletion-associated mitochondrial dysfunction including mitochondrial superoxide accumulation, ATP depletion and mitochondrial AIF release. Moreover, both NE and DA inhibited glutathione depletion-associated MAPKs activation, p53 phosphorylation and GADD45α activation. Molecular docking analysis revealed that NE and DA could bind to protein disulfide isomerase (PDI). In biochemical enzymatic assay in vitro, NE and DA dose-dependently inhibited the reductive activity of PDI. We further revealed that the protective effect of NE and DA against glutamate-induced oxidative cytotoxicity was mediated through inhibition of PDI-catalyzed dimerization of the neuronal nitric oxide synthase. Collectively, the results of this study suggest that NE and DA may have a protective effect against oxidative neurodegeneration through inhibition of protein disulfide isomerase and the subsequent activation of the MAPKs‒p53‒GADD45α oxidative cascade.
Collapse
Affiliation(s)
- Hye Joung Choi
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tong-Xiang Chen
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Ji Hoon Song
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Peng Li
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Chun-Feng Liu
- Institute of Neuroscience, Soochow University, and Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China.
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
35
|
Tran TTT, Nguyen TKA, Nguyen BN, Hoang TMN, Doan LP, Phan MG, Lee H, Kim DW, Lee JW. Six new polyoxygenated xanthones from Garcinia cowa and their neuroprotective effects on glutamate-mediated hippocampal neuronal HT22 cell death. Chem Biodivers 2022; 19:e202200376. [PMID: 35927784 DOI: 10.1002/cbdv.202200376] [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: 04/19/2022] [Accepted: 08/04/2022] [Indexed: 11/06/2022]
Abstract
Six new polyoxygenated xanthones, garcicowanones F-H (1-3), norcowanol A-B (4-5), and garcinone F (6) along with twelve known compounds 7-18 were obtained from the latex of Garcinia cowa Roxb. ex Choisy. All new compounds have a 1,3,7-trioxygenated or 1,3,6,7-tetraoxygenated xanthone nucleus and differ from majority of xanthones from G. cowa by hydrated side chains. Compounds 1, 7, 8 and 18 exhibited significant neuroprotective effects on glutamate-mediated hippocampal neuronal HT22 cell death. In particular, compound 1 exhibited the most potent neuroprotective effect with >80% cell viability in the concentration range of 2.9-115 µM. Further studies on compound 1 showed that it decreased cellular Ca2+ influx and inhibits cellular reactive oxygen species generation in HT22 cells. A Western blot analysis showed that MAPK phosphorylation, Bax, and AIF translocation dramatically increased upon treatment with 5 mM glutamate and decreased upon a co-treatment with compound 1.
Collapse
Affiliation(s)
- Thi Thu Thuy Tran
- Vietnam Academy of Science and Technology, Institute of Natural Products Chemistry, 1H, 18 Hoang Quoc Viet, Cau Giay, 122045, Hanoi, VIET NAM
| | - Thi Kim An Nguyen
- Vietnam Academy of Science and Technology, Chemistry, 1H, 18 Hoang Quoc Viet, Cau Giay, 122045, Hanoi, VIET NAM
| | - Bao Ngoc Nguyen
- Korea Institute of Science and Technology, Natural Product Research Center, 679 Saimdang-ro, Gyeongpo-dong, Gangneung-si, Gangwon-do, Gangneung, KOREA, REPUBLIC OF
| | - Thi Minh Nguyet Hoang
- Vietnam Academy of Science and Technology, Institute of Natural Products Chemistry, 1H, 18 Hoang Quoc Viet, Cau Giay, 122045, Hanoi, VIET NAM
| | - Lan Phuong Doan
- Vietnam Academy of Science and Technology, Institute of Natural Products Chemistry, 1H, 18 Hoang Quoc Viet, Cau Giay, 122045, Hanoi, VIET NAM
| | - Minh Giang Phan
- Vietnam National University Hanoi, Chemistry, 334 Nguyen Trai, 122045, Hanoi, VIET NAM
| | - Heesu Lee
- Gangneung-Wonju National University, College of Dentistry, 7, Jukheon-gil, Gangneung, KOREA, REPUBLIC OF
| | - Dae Won Kim
- Gangneung-Wonju National University, College of Dentistry, 7, Jukheon-gil, Gangneung, KOREA, REPUBLIC OF
| | - Jae Wook Lee
- Korea Institute of Science and Technology, Natural Product Research Center, 679 Saimdang-ro, Gyeongpo-dong, Gangneung-si, Gangwon-do, Gangneung, KOREA, REPUBLIC OF
| |
Collapse
|
36
|
Rakkhittawattana V, Panichayupakaranant P, Prasanth MI, Brimson JM, Tencomnao T. Rhinacanthin-C but Not -D Extracted from Rhinacanthus nasutus (L.) Kurz Offers Neuroprotection via ERK, CHOP, and LC3B Pathways. Pharmaceuticals (Basel) 2022; 15:627. [PMID: 35631453 PMCID: PMC9145051 DOI: 10.3390/ph15050627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Neurodegenerative diseases present an increasing problem as the world's population ages; thus, the discovery of new drugs that prevent diseases such as Alzheimer's, and Parkinson's diseases are vital. In this study, Rhinacanthin-C and -D were isolated from Rhinacanthus nasustus, using ethyl acetate, followed by chromatography to isolate Rhinacanthin-C and -D. Both compounds were confirmed using NMR and ultra-performance-LCMS. Using glutamate toxicity in HT-22 cells, we measured cell viability and apoptosis, ROS build-up, and investigated signaling pathways. We show that Rhinacanthin-C and 2-hydroxy-1,4-naphthoquinone have neuroprotective effects against glutamate-induced apoptosis in HT-22 cells. Furthermore, we see that Rhinacanthin-C resulted in autophagy inhibition and increased ER stress. In contrast, low concentrations of Rhinacanthin-C and 2-hydroxy-1,4-naphthoquinone prevented ER stress and CHOP expression. All concentrations of Rhinacanthin-C prevented ROS production and ERK1/2 phosphorylation. We conclude that, while autophagy is present in HT-22 cells subjected to glutamate toxicity, its inhibition is not necessary for cryoprotection.
Collapse
Affiliation(s)
- Varaporn Rakkhittawattana
- Ph.D. Program in Clinical Biochemistry and Molecular Medicine, 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;
| | - Pharkphoom Panichayupakaranant
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 90112, Thailand;
| | - Mani I. Prasanth
- 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
| | - James M. Brimson
- 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
| | - 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
|
37
|
Mechanism of Erastin-Induced Ferroptosis in MDA-MB-231 Human Breast Cancer Cells: Evidence for a Critical Role of Protein Disulfide Isomerase. Mol Cell Biol 2022; 42:e0052221. [PMID: 35499331 PMCID: PMC9202373 DOI: 10.1128/mcb.00522-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Ferroptosis is a form of regulated cell death resulting predominantly from catastrophic accumulation of lipid reactive oxygen species (ROS). While the antioxidant systems that counter ferroptosis have been well characterized, the mechanism underlying ferroptosis-associated accumulation of lipid ROS remains unclear. In this study, we demonstrated that protein disulfide isomerase (PDI) is a novel mediator of ferroptosis, which is responsible for the accumulation of lipid ROS and ultimately ferroptosis in MDA-MB-231 human breast cancer cells. Treatment with erastin led to a significant increase in inducible nitric oxide synthase (iNOS)-mediated nitric oxide production, which contributes to the accumulation of the death-inducing cellular lipid ROS. Small interfering RNA (siRNA)-mediated PDI knockdown or pharmacological inhibition of PDI’s isomerase activity with cystamine strongly suppressed iNOS dimerization and its catalytic activation, subsequently prevented lipid ROS accumulation, and conferred strong protection against erastin-induced ferroptosis. Remarkably, PDI knockdown in MDA-MB-231 cells also largely abrogated the protective effect of cystamine against erastin-induced ferroptotic cell death. Together, these experimental observations demonstrate a noncanonical role of PDI in ferroptosis, which may serve as a potential therapeutic target for ferroptosis-related diseases.
Collapse
|
38
|
AL-Nasser MN, Mellor IR, Carter WG. Is L-Glutamate Toxic to Neurons and Thereby Contributes to Neuronal Loss and Neurodegeneration? A Systematic Review. Brain Sci 2022; 12:577. [PMID: 35624964 PMCID: PMC9139234 DOI: 10.3390/brainsci12050577] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
L-glutamate (L-Glu) is a nonessential amino acid, but an extensively utilised excitatory neurotransmitter with critical roles in normal brain function. Aberrant accumulation of L-Glu has been linked to neurotoxicity and neurodegeneration. To investigate this further, we systematically reviewed the literature to evaluate the effects of L-Glu on neuronal viability linked to the pathogenesis and/or progression of neurodegenerative diseases (NDDs). A search in PubMed, Medline, Embase, and Web of Science Core Collection was conducted to retrieve studies that investigated an association between L-Glu and pathology for five NDDs: Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Together, 4060 studies were identified, of which 71 met eligibility criteria. Despite several inadequacies, including small sample size, employment of supraphysiological concentrations, and a range of administration routes, it was concluded that exposure to L-Glu in vitro or in vivo has multiple pathogenic mechanisms that influence neuronal viability. These mechanisms include oxidative stress, reduced antioxidant defence, neuroinflammation, altered neurotransmitter levels, protein accumulations, excitotoxicity, mitochondrial dysfunction, intracellular calcium level changes, and effects on neuronal histology, cognitive function, and animal behaviour. This implies that clinical and epidemiological studies are required to assess the potential neuronal harm arising from excessive intake of exogenous L-Glu.
Collapse
Affiliation(s)
- Maryam N. AL-Nasser
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| | - Ian R. Mellor
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Wayne G. Carter
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| |
Collapse
|
39
|
Oikawa T, Fukuda T, Yamashita T, Tomita H, Ozaki T. Lentiviral expression of calpain-1 C2-like domain peptide prevents glutamate-induced cell death in mouse hippocampal neuronal HT22 cells. In Vitro Cell Dev Biol Anim 2022; 58:289-294. [PMID: 35469046 DOI: 10.1007/s11626-022-00683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/30/2022] [Indexed: 11/05/2022]
Abstract
Glutamate neurotoxicity is involved in neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. Excess glutamate causes caspase-independent programmed cell death via oxidative stress and calcium influx. Our previous study showed that calpain-1 localizes to both the cytoplasm and mitochondria, where apoptosis-inducing factor (AIF) is cleaved by calpain-1 and translocates to the nucleus to induce DNA fragmentation. The autoinhibitory region of calpain-1 conjugated with the cell-penetrating peptide HIV1-Tat (namely Tat-μCL) specifically prevents the activity of mitochondrial calpain-1 and attenuates neuronal cell death in animal models of retinitis pigmentosa, as well as glutamate-induced cell death in mouse hippocampal HT22 cells. In the present study, we constructed a lentiviral vector expressing the Tat-μCL peptide and evaluated its protective effect against glutamate-induced cell death in HT22 cells. Lentiviral transduction with Tat-μCL significantly suppressed glutamate-induced nuclear translocation of AIF and DNA fragmentation. The findings of the present study suggest that the stable expression of Tat-μCL may be a potential gene therapy modality for neurodegenerative diseases.
Collapse
Affiliation(s)
- Takenori Oikawa
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 020-8550, Japan
| | - Tomokazu Fukuda
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 020-8550, Japan
| | - Hiroshi Tomita
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Taku Ozaki
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| |
Collapse
|
40
|
Ko J, Jang S, Kwon W, Kim SY, Jang S, Kim E, Ji YR, Park S, Kim MO, Choi SK, Cho DH, Lee HS, Lim SG, Ryoo ZY. Protective Effect of GIP against Monosodium Glutamate-Induced Ferroptosis in Mouse Hippocampal HT-22 Cells through the MAPK Signaling Pathway. Antioxidants (Basel) 2022; 11:antiox11020189. [PMID: 35204073 PMCID: PMC8868324 DOI: 10.3390/antiox11020189] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
The effect of glucose-dependent insulinotropic polypeptide (GIP) on cells under oxidative stress induced by glutamate, a neurotransmitter, and the underlying molecular mechanisms were assessed in the present study. We found that in the pre-treatment of HT-22 cells with glutamate in a dose-dependent manner, intracellular ROS were excessively generated, and additional cell damage occurred in the form of lipid peroxidation. The neurotoxicity caused by excessive glutamate was found to be ferroptosis and not apoptosis. Other factors (GPx-4, Nrf2, Nox1 and Hspb1) involved in ferroptosis were also identified. In other words, it was confirmed that GIP increased the activity of sub-signalling molecules in the process of suppressing ferroptosis as an antioxidant and maintained a stable cell cycle even under glutamate-induced neurotoxicity. At the same time, in HT-22 cells exposed to ferroptosis as a result of excessive glutamate accumulation, GIP sustained cell viability by activating the mitogen-activated protein kinase (MAPK) signalling pathway. These results suggest that the overexpression of the GIP gene increases cell viability by regulating mechanisms related to cytotoxicity and reactive oxygen species production in hippocampal neuronal cell lines.
Collapse
Affiliation(s)
- Jiwon Ko
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
| | - Soyoung Jang
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
| | - Wookbong Kwon
- Core Protein Resources Center, DGIST, Daegu 42988, Korea; (W.K.); (S.-K.C.)
- Division of Biotechnology, DGIST, Daegu 42988, Korea
| | - Si-Yong Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
| | - Soyeon Jang
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
| | - Eungyung Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Korea; (E.K.); (M.-O.K.)
| | - Young-Rae Ji
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
- Section on Sensory Cell Regeneration and Development, Laboratory of Molecular Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sijun Park
- School of Life Science, Kyungpook National University, Daegu 42988, Korea;
| | - Myoung-Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Korea; (E.K.); (M.-O.K.)
| | - Seong-Kyoon Choi
- Core Protein Resources Center, DGIST, Daegu 42988, Korea; (W.K.); (S.-K.C.)
- Division of Biotechnology, DGIST, Daegu 42988, Korea
| | - Dong-Hyung Cho
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 42988, Korea
| | - Hyun-Shik Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
| | - Su-Geun Lim
- School of Life Science, Kyungpook National University, Daegu 42988, Korea;
- Correspondence: (S.-G.L.); (Z.-Y.R.)
| | - Zae-Young Ryoo
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.K.); (S.J.); (S.-Y.K.); (S.J.); (Y.-R.J.); (D.-H.C.); (H.-S.L.)
- Correspondence: (S.-G.L.); (Z.-Y.R.)
| |
Collapse
|
41
|
Pokharel S, Gliyazova NS, Dandepally SR, Williams AL, Ibeanu GC. Neuroprotective effects of an in vitro BBB permeable phenoxythiophene sulfonamide small molecule in glutamate-induced oxidative injury. Exp Ther Med 2022; 23:79. [PMID: 34938365 PMCID: PMC8688931 DOI: 10.3892/etm.2021.11002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS) play a central role in oxidative stress-associated neuronal cell death during ischemia. Further investigation into the inhibition of excessive ROS generation post-stroke is urgently required for the treatment of ischemic stroke. In the present study, the neuroprotective properties of the blood-brain barrier (BBB) penetrant B355227 were investigated. B355227 is a chemical analogue of B355252, and the role of the phenoxythiophene sulfonamide compound B355227 was further investigated in a glutamate-induced oxidative injury model. An in vitro model of the BBB was established in the immortalized mouse brain capillary endothelial cell line, bEnd.3. Formation of barrier in Transwell inserts was confirmed using EVOM resistance meter and Caffeine, Imatinib and Axitinib were used to validate the efficacy of the model. The validated BBB assay in combination with high performance liquid chromatography were used to analyse and verify the permeability of B355227 through the barrier. The integrity of the cell junctions after the BBB assays were confirmed using immunofluorescence to visualize the expression of the barrier junction protein zonula occludens-1. Cell survival was measured with Resazurin, a redox indicator dye, in HT22, a hippocampal neuronal cell treated with 5 mM glutamate or co-treated with the B355227 recovered from the BBB permeability experiment. Changes in glutathione levels were detected using a glutathione detection kit, while analyses of ROS, calcium (Ca2+), and mitochondrial membrane potential (MMP) were accomplished with the fluorescent dyes 2',7'-dichlorofluorescein diacetate, Fura-2 AM and MitoTracker Red dyes, respectively. Immunoblotting was also performed to detect the expression and activation of Erk1/2, p-38, JNK, Bax and Bcl-2. The results of the present study demonstrated that B355227 crossed the BBB in vitro and protected HT22 from oxidative injury induced by glutamate exposure. Treatment of cells with B355227 blocked the glutamate-dependent depletion of intracellular glutathione and significantly reduced ROS production. Increased Ca2+ influx and subsequent collapse of the MMP was attenuated by B355227. Furthermore, the results of the present study demonstrated that B355227 protected against oxidative stress via the MAPK pathway, by increasing the activation of Erk1/2, JNK and P38, and restoring anti-apoptotic Bcl-2. Collectively, the results of the present study indicate that B355227 has potent antioxidant and neuroprotective attributes in glutamate-induced neuronal cell death. Further investigation into the role of B355227 in the modulation of glutamate-dependent oxidative stress is required.
Collapse
Affiliation(s)
- Smritee Pokharel
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Nailya S. Gliyazova
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Srinivasa R. Dandepally
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Alfred L. Williams
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
- Department of Pharmaceutical Science, North Carolina Central University, Durham, NC 27707, USA
| | - Gordon C. Ibeanu
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
- Department of Pharmaceutical Science, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
42
|
Kang JB, Park DJ, Shah MA, Koh PO. Quercetin ameliorates glutamate toxicity-induced neuronal cell death by controlling calcium-binding protein parvalbumin. J Vet Sci 2022; 23:e26. [PMID: 35187882 PMCID: PMC8977545 DOI: 10.4142/jvs.21273] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/06/2021] [Accepted: 12/23/2021] [Indexed: 11/24/2022] Open
Abstract
Background Glutamate is the main excitatory neurotransmitter. Excessive glutamate causes excitatory toxicity and increases intracellular calcium, leading to neuronal death. Parvalbumin is a calcium-binding protein that regulates calcium homeostasis. Quercetin is a polyphenol found in plant and has neuroprotective effects against neurodegenerative diseases. Objectives We investigated whether quercetin regulates apoptosis by modulating parvalbumin expression in glutamate induced neuronal damage. Methods Glutamate was treated in hippocampal-derived cell line, and quercetin or vehicle was treated 1 h before glutamate exposure. Cells were collected for experimental procedure 24 h after glutamate treatment and intracellular calcium concentration and parvalbumin expression were examined. Parvalbumin small interfering RNA (siRNA) transfection was performed to detect the relation between parvalbumin and apoptosis. Results Glutamate reduced cell viability and increased intracellular calcium concentration, while quercetin preserved calcium concentration and neuronal damage. Moreover, glutamate reduced parvalbumin expression and quercetin alleviated this reduction. Glutamate increased caspase-3 expression, and quercetin attenuated this increase in both parvalbumin siRNA transfected and non-transfected cells. The alleviative effect of quercetin was statistically significant in non-transfected cells. Moreover, glutamate decreased bcl-2 and increased bax expressions, while quercetin alleviated these changes. The alleviative effect of quercetin in bcl-2 family protein expression was more remarkable in non-transfected cells. Conclusions These results demonstrate that parvalbumin contributes to the maintainace of intracellular calcium concentration and the prevention of apoptosis, and quercetin modulates parvalbumin expression in glutamate-exposed cells. Thus, these findings suggest that quercetin performs neuroprotective function against glutamate toxicity by regulating parvalbumin expression.
Collapse
Affiliation(s)
- Ju-Bin Kang
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Dong-Ju Park
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Murad-Ali Shah
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Phil-Ok Koh
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
43
|
Rosarda J, Baron KR, Nutsch K, Kline GM, Stanton C, Kelly JW, Bollong MJ, Wiseman RL. Metabolically Activated Proteostasis Regulators Protect against Glutamate Toxicity by Activating NRF2. ACS Chem Biol 2021; 16:2852-2863. [PMID: 34797633 PMCID: PMC8689639 DOI: 10.1021/acschembio.1c00810] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022]
Abstract
The extracellular accumulation of glutamate is a pathologic hallmark of numerous neurodegenerative diseases including ischemic stroke and Alzheimer's disease. At high extracellular concentrations, glutamate causes neuronal damage by promoting oxidative stress, which can lead to cellular death. This has led to significant interest in developing pharmacologic approaches to mitigate the oxidative toxicity caused by high levels of glutamate. Here, we show that the small molecule proteostasis regulator AA147 protects against glutamate-induced cell death in a neuronal-derived cell culture model. While originally developed as an activator of the activating transcription factor 6 (ATF6) arm of the unfolded protein response, this AA147-dependent protection against glutamate toxicity is primarily mediated through activation of the NRF2-regulated oxidative stress response. We demonstrate that AA147 activates NRF2 selectively in neuronal-derived cells through a mechanism involving metabolic activation to a reactive electrophile and covalent modification of KEAP1─a mechanism analogous to that involved in the AA147-dependent activation of ATF6. These results define the potential for AA147 to protect against glutamate-induced oxidative toxicity and highlight the potential for metabolically activated proteostasis regulators like AA147 to activate both protective ATF6 and NRF2 stress-responsive signaling pathways to mitigate oxidative damage associated with diverse neurologic diseases.
Collapse
Affiliation(s)
- Jessica
D. Rosarda
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Kelsey R. Baron
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Kayla Nutsch
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Gabriel M. Kline
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
- The
Skaggs Institute for Chemical Biology, The
Scripps Research Institute, La
Jolla, California 92037, United States
| | - Caroline Stanton
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Jeffery W. Kelly
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
- The
Skaggs Institute for Chemical Biology, The
Scripps Research Institute, La
Jolla, California 92037, United States
| | - Michael J. Bollong
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - R. Luke Wiseman
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| |
Collapse
|
44
|
Wang F, He J, Xing R, Sha T, Sun B. Molecular mechanisms of ferroptosis and their role in inflammation. Int Rev Immunol 2021; 42:71-81. [PMID: 34918993 DOI: 10.1080/08830185.2021.2016739] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Ferroptosis is a type of non-apoptotic cell death, which demonstrates a definite iron-dependent expression pattern and is associated with lipid peroxidation. Glutathione peroxidase 4 (GPX4) is a key regulator of ferroptosis. Ferroptosis is involved in the development and progression of various diseases, such as cancer, tissue ischemia-reperfusion injury, neurological diseases, and respiratory diseases. It has been established previously that ferroptotic cells trigger the innate immune system by releasing inflammation-linked damage-related molecules, and immune cells stimulate the inflammatory response by recognizing the operational mechanism of ferroptosis. Some anti-inflammatory drugs have been shown to inhibit ferroptosis in certain cell models. Conversely, some ferroptosis inhibitors also exert anti-inflammatory effects in certain diseases. The present review evaluated the relationship between ferroptosis and inflammation, as well as the underlying internal mechanism, and provided valuable insights into developing novel treatment strategies for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Feng Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, P.R. China
| | - Jingya He
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, P.R. China
| | - Ruxiao Xing
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, P.R. China
| | - Tong Sha
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, P.R. China
| | - Bin Sun
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, P.R. China
| |
Collapse
|
45
|
Cui Y, Xiong Y, Li H, Zeng M, Wang Y, Li Y, Zou X, Lv W, Gao J, Cao R, Meng L, Long J, Liu J, Feng Z. Chalcone-Derived Nrf2 Activator Protects Cognitive Function via Maintaining Neuronal Redox Status. Antioxidants (Basel) 2021; 10:antiox10111811. [PMID: 34829682 PMCID: PMC8615013 DOI: 10.3390/antiox10111811] [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: 10/21/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/05/2022] Open
Abstract
NF-E2-related factor 2 (Nrf2), the key transcription regulator of phase II enzymes, has been considered beneficial for neuronal protection. We previously designed a novel chalcone analog, 1-(2,3,4-trimethoxyphenyl)-2-(3,4,5-trimethoxyphenyl)-acrylketone (Tak), that could specifically activate Nrf2 in vitro. Here, we report that Tak confers significant hippocampal neuronal protection both in vitro and in vivo. Treatment with Tak has no significant toxicity on cultured neuronal cells. Instead, Tak increases cellular ATP production by increasing mitochondrial function and decreases the levels of reactive oxygen species by activating Nrf2-mediated phase II enzyme expression. Tak pretreatment prevents glutamate-induced excitotoxic neuronal death accompanied by suppressed mitochondrial respiration, increased superoxide production, and activation of apoptosis. Further investigation indicates that the protective effect of Tak is mediated by the Akt signaling pathway. Meanwhile, Tak administration in mice can sufficiently abrogate scopolamine-induced cognitive impairment via decreasing hippocampal oxidative stress. In addition, consistent benefits are also observed in an energy stress mouse model under a high-fat diet, as the administration of Tak remarkably increases Akt signaling-mediated antioxidative enzyme expression and prevents hippocampal neuronal apoptosis without significant effect on the mouse metabolic status. Overall, our study demonstrates that Tak protects cognitive function by Akt-mediated Nrf2 activation to maintain redox status both vivo and in vitro, suggesting that Tak is a promising pharmacological candidate for the treatment of oxidative neuronal diseases.
Collapse
Affiliation(s)
- Yuting Cui
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Yue Xiong
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Hua Li
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Mengqi Zeng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Yan Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Yuan Li
- Institute of Basic Medical Science, Xi’an Medical University, Xi’an 710021, China;
| | - Xuan Zou
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Shannxi 710004, China;
- Shaanxi Provincial Clinical Research Center for Hepatic and Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Shannxi 710004, China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Jing Gao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Ruijun Cao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Science, Xi’an Jiaotong University, Xi’an 710049, China; (R.C.); (L.M.)
| | - Lingjie Meng
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Science, Xi’an Jiaotong University, Xi’an 710049, China; (R.C.); (L.M.)
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.C.); (Y.X.); (H.L.); (M.Z.); (Y.W.); (W.L.); (J.G.); (J.L.)
- University of Health and Rehabilitation Sciences, Qingdao 266071, China
- Correspondence: (J.L.); (Z.F.)
| | - Zhihui Feng
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Shannxi 710004, China;
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Correspondence: (J.L.); (Z.F.)
| |
Collapse
|
46
|
Torres VI, Barrera DP, Varas-Godoy M, Arancibia D, Inestrosa NC. Selective Surface and Intraluminal Localization of Wnt Ligands on Small Extracellular Vesicles Released by HT-22 Hippocampal Neurons. Front Cell Dev Biol 2021; 9:735888. [PMID: 34722516 PMCID: PMC8548728 DOI: 10.3389/fcell.2021.735888] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
The Wnt signaling pathway induces various responses underlying the development and maturation of the nervous system. Wnt ligands are highly hydrophobic proteins that limit their diffusion through an aqueous extracellular medium to a target cell. Nevertheless, their attachment to small extracellular vesicles-like exosomes is one of the described mechanisms that allow their transport under this condition. Some Wnt ligands in these vehicles are expected to be dependent on post-translational modifications such as acylation. The mechanisms determining Wnt loading in exosomes and delivery to the target cells are largely unknown. Here, we took advantage of a cell model that secret a highly enriched population of small extracellular vesicles (sEVs), hippocampal HT-22 neurons. First, to establish the cell model, we characterized the morphological and biochemical properties of an enriched fraction of sEVs obtained from hippocampal HT-22 neurons that express NCAM-L1, a specific exosomal neuronal marker. Transmission electron microscopy showed a highly enriched fraction of exosome-like vesicles. Next, the exosomal presence of Wnt3a, Wnt5a, and Wnt7a was confirmed by western blot analysis and electron microscopy combined with immunogold. Also, we studied whether palmitoylation is a necessary post-translational modification for the transport Wnt in these vesicles. We found that proteinase-K treatment of exosomes selectively decreased their Wnt5a and Wnt7a content, suggesting that their expression is delimited to the exterior membrane surface. In contrast, Wnt3a remained attached, suggesting that it is localized within the exosome lumen. On the other hand, Wnt-C59, a specific inhibitor of porcupine O-acyltransferase (PORCN), decreased the association of Wnt with exosomes, suggesting that Wnt ligand acylation is necessary for them to be secreted by exosomes. These findings may help to understand the action of the Wnt ligands in the target cell, which could be defined during the packaging of the ligands in the secretory cell sEVs.
Collapse
Affiliation(s)
- Viviana I Torres
- Departamento Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Daniela P Barrera
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Varas-Godoy
- Cancer Cell Biology Laboratory, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Duxan Arancibia
- Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
47
|
Vongthip W, Sillapachaiyaporn C, Kim KW, Sukprasansap M, Tencomnao T. Thunbergia laurifolia Leaf Extract Inhibits Glutamate-Induced Neurotoxicity and Cell Death through Mitophagy Signaling. Antioxidants (Basel) 2021; 10:antiox10111678. [PMID: 34829549 PMCID: PMC8614718 DOI: 10.3390/antiox10111678] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
Oxidative stress plays a crucial role in neurodegeneration. Therefore, reducing oxidative stress in the brain is an important strategy to prevent neurodegenerative disorders. Thunbergia laurifolia (Rang-jued) is well known as an herbal tea in Thailand. Here, we aimed to determine the protective effects of T. laurifolia leaf extract (TLE) on glutamate-induced oxidative stress toxicity and mitophagy-mediated cell death in mouse hippocampal cells (HT-22). Our results reveal that TLE possesses a high level of bioactive antioxidants by LC–MS technique. We found that the pre-treatment of cells with TLE prevented glutamate-induced neuronal death in a concentration-dependent manner. TLE reduced the intracellular ROS and maintained the mitochondrial membrane potential caused by glutamate. Moreover, TLE upregulated the gene expression of antioxidant enzymes (SOD1, SOD2, CAT, and GPx). Interestingly, glutamate also induced the activation of the mitophagy process. However, TLE could reverse this activity by inhibiting autophagic protein (LC3B-II/LC3B-I) activation and increasing a specific mitochondrial protein (TOM20). Our results suggest that excessive glutamate can cause neuronal death through mitophagy-mediated cell death signaling in HT-22 cells. Our findings indicate that TLE protects cells from neuronal death by stimulating the endogenous antioxidant enzymes and inhibiting glutamate-induced oxidative toxicity via the mitophagy–autophagy pathway. TLE might have potential as an alternative or therapeutic approach in neurodegenerative diseases.
Collapse
Affiliation(s)
- Wudtipong Vongthip
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.V.); (C.S.)
| | - Chanin Sillapachaiyaporn
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.V.); (C.S.)
| | - Kyu-Won Kim
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul 151-742, Korea;
| | - Monruedee Sukprasansap
- Food Toxicology Unit, Institute of Nutrition, Mahidol University, Nakhon Pathom 73170, Thailand
- Correspondence: (M.S.); (T.T.); Tel.: +66-2-800-2380 (M.S.); +66-2-218-1533 (T.T.)
| | - 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
- Correspondence: (M.S.); (T.T.); Tel.: +66-2-800-2380 (M.S.); +66-2-218-1533 (T.T.)
| |
Collapse
|
48
|
Selvaraj B, Woon Kim D, Park JS, Cheol Kwon H, Lee H, Yoo KY, Wook Lee J. Neuroprotective effects of 2-heptyl-3-hydroxy-4-quinolone in HT22 mouse hippocampal neuronal cells. Bioorg Med Chem Lett 2021; 49:128312. [PMID: 34375718 DOI: 10.1016/j.bmcl.2021.128312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
The neuroprotective activity of 2-heptyl-3-hydroxy-4(1H)-quinolone (compound 1) was evaluated using the neurotoxicity of glutamate in the HT22 cell line. Compound 1, known as a signal molecule of the bacterial quorum-sensing system, protects neuronal cells from glutamate-induced neurotoxicity by inhibiting cellular Ca2+ uptake and glutamate-triggered ROS accumulation. MAPK signaling pathway inhibition by compound 1 was evaluated by immunoblotting the phosphorylation status of the proteins. Furthermore, pro-apoptotic protein levels and AIF translocation to the nucleus were found to be reduced by compound 1. In conclusion, compound 1 showed neuroprotective effects by inhibiting apoptotic neuronal cell death.
Collapse
Affiliation(s)
- Baskar Selvaraj
- Natural Product Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangnueng 25451, Republic of Korea; Convergence Research Center of Dementia, Brain Science Institute, Korea Institute of Science and Technology, 02792, Republic of Korea; Division of Bio-medical Science & Technology, University of Science and Technology, Daejun 34113, Republic of Korea
| | - Dae Woon Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Science, College of Dentistry, Gangneung Wonju National University, 25457, Republic of Korea
| | - Jin-Soo Park
- Natural Product Informatics Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangnueng 25451, Republic of Korea
| | - Hak Cheol Kwon
- Natural Product Informatics Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangnueng 25451, Republic of Korea
| | - Heesu Lee
- Department of Oral Anatomy, College of Dentistry, Institute of Oral Science, Gangneung Wonju National University, Gangneung, Republic of Korea
| | - Ki-Yeon Yoo
- Department of Oral Anatomy, College of Dentistry, Institute of Oral Science, Gangneung Wonju National University, Gangneung, Republic of Korea
| | - Jae Wook Lee
- Natural Product Research Center, Institute of Natural Product, Korea Institute of Science and Technology, Gangnueng 25451, Republic of Korea; Convergence Research Center of Dementia, Brain Science Institute, Korea Institute of Science and Technology, 02792, Republic of Korea; Division of Bio-medical Science & Technology, University of Science and Technology, Daejun 34113, Republic of Korea.
| |
Collapse
|
49
|
Calpain-1 C2L domain peptide protects mouse hippocampus-derived neuronal HT22 cells against glutamate-induced oxytosis. Biochem Biophys Rep 2021; 27:101101. [PMID: 34430716 PMCID: PMC8374356 DOI: 10.1016/j.bbrep.2021.101101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 01/13/2023] Open
Abstract
Calpains are Ca2+-dependent cysteine proteases; their aberrant activation is associated with several neurodegenerative diseases. The μ-calpain catalytic subunit, calpain-1, is located in the cytoplasm as well as in the mitochondria. Mitochondrial calpain-1 cleaves apoptosis-inducing factor (AIF), leading to apoptotic cell death. We have previously reported that short peptides of calpain-1 C2-like domain conjugated with cell penetrating peptide HIV-Tat (Tat-μCL) selectively inhibit mitochondrial calpain-1 and effectively prevent neurodegenerative diseases of the eye. In this study, we determined whether mitochondrial calpain-1 mediates oxytosis (oxidative glutamate toxicity) in hippocampal HT22 cells using Tat-μCL and newly generated polyhistidine-conjugated μCL peptide and compared their efficacies in preventing oxytosis. TUNEL assay and single strand DNA staining revealed that both μCL peptides inhibited glutamate-induced oxytosis. Additionally, both the peptides suppressed the mitochondrial AIF translocation into the nucleus. All polyhistidine-μCL peptides (containing 4–16 histidine residues) showed higher cell permeability than Tat-μCL. Notably, tetrahistidine (H4)-μCL exerted the highest cytoprotective activity. Thus, H4-μCL may be a potential peptide drug for calpain-1-mediated neurodegenerative diseases such as Alzheimer's disease. Mitochondrial calpain-1 mediates glutamate-induced oxytosis in HT22 cells. CPP-μCL inhibits AIF translocation and DNA fragmentation in oxytosis. Polyhistidine conjugation enhances intracellular μCL peptide uptake efficiency.
Collapse
Key Words
- AIF, apoptosis-inducing factor
- BBB, blood–brain barrier
- CPP, cell penetrating peptide
- Cell penetrating peptide
- DAPI, 4′,6-diamidino-2-phenylindole
- DMEM, Dulbecco's Modified Eagle Medium
- Hippocampal HT22 cells
- Hn-μCL, polyhistidine-conjugated μCL
- MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium
- Mitochondrial calpain-1
- Neurodegeneration
- Oxytosis
- PBS, phosphate-buffered saline
- ssDNA, single stranded DNA
Collapse
|
50
|
Duangjan C, Rangsinth P, Zhang S, Gu X, Wink M, Tencomnao T. Neuroprotective Effects of Glochidion zeylanicum Leaf Extract against H 2O 2/Glutamate-Induced Toxicity in Cultured Neuronal Cells and Aβ-Induced Toxicity in Caenorhabditis elegans. BIOLOGY 2021; 10:biology10080800. [PMID: 34440032 PMCID: PMC8389654 DOI: 10.3390/biology10080800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022]
Abstract
Simple Summary Antioxidants that are interrelated in the process of overcoming oxidative-stress-induced toxicity and neurite-outgrowth-inducing activity have become the main targets of neuroprotective therapy. The methanol extract of Glochidion zeylanicum (GZM) exhibits neuroprotective properties that are not only limited against H2O2/glutamate/Aβ insults but also promote neurite outgrowth activity. The neuroprotective effects of GZM extract were confirmed in cultured neuronal (HT-22 and Neuro-2a) cells and C. elegans models. To the best of our knowledge, this study is the first to report for the neuroprotective effects of GZM extract, suggesting that G. zeylanicum may be a neuroprotectant applicant for the prevention and alleviation of oxidative stress-induced neurodegenerative disorders, including Alzheimer’s disease. However, additional studies are required to identify the mechanistic pathways involved in neuroprotection and to confirm the efficacy of the extract in more complex model organisms. Abstract Oxidative stress plays a crucial role in the development of age-related neurodegenerative diseases. Previously, Glochidion zeylanicum methanol (GZM) extract has been reported to have antioxidant and anti-aging properties. However, the effect of GZM on neuroprotection has not been reported yet; furthermore, the mechanism involved in its antioxidant properties remains unresolved. The study is aimed to demonstrate the neuroprotective properties of GZM extract and their underlying mechanisms in cultured neuronal (HT-22 and Neuro-2a) cells and Caenorhabditis elegans models. GZM extract exhibited protective effects against glutamate/H2O2-induced toxicity in cultured neuronal cells by suppressing the intracellular reactive oxygen species (ROS) generation and enhancing the expression of endogenous antioxidant enzymes (SODs, GPx, and GSTs). GZM extract also triggered the expression of SIRT1/Nrf2 proteins and mRNA transcription of antioxidant genes (NQO1, GCLM, and EAAT3) which are the master regulators of cellular defense against oxidative stress. Additionally, GZM extract exhibited protective effects to counteract β-amyloid (Aβ)-induced toxicity in C. elegans and promoted neuritogenesis properties in Neuro-2a cells. Our observations suggest that GZM leaf extract has interesting neuritogenesis and neuroprotective potential and can possibly act as potential contender for the treatment of oxidative stress-induced Alzheimer’s disease (AD) and related neurodegenerative conditions; however, this needs to be studied further in other in vivo systems.
Collapse
Affiliation(s)
- Chatrawee Duangjan
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (C.D.); (P.R.)
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Panthakarn Rangsinth
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (C.D.); (P.R.)
| | - Shaoxiong Zhang
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg University, 69120 Heidelberg, Germany;
| | - Xiaojie Gu
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg University, 69120 Heidelberg, Germany;
- Department of Biotechnology, School of Environmental and Chemical Engineering, Dalian Jiaotong University, Dalian 116028, China
| | - Michael Wink
- Department of Biotechnology, School of Environmental and Chemical Engineering, Dalian Jiaotong University, Dalian 116028, China
- Correspondence: (M.W.); (T.T.); Tel.: +49-6221-544881 (M.W.); +66-2181081 (ext. 313) (T.T.)
| | - Tewin Tencomnao
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (C.D.); (P.R.)
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (M.W.); (T.T.); Tel.: +49-6221-544881 (M.W.); +66-2181081 (ext. 313) (T.T.)
| |
Collapse
|