1
|
Sandech N, Yang MC, Juntranggoor P, Rukthong P, Gorelkin P, Savin N, Timoshenko R, Vaneev A, Erofeev A, Wichaiyo S, Pradidarcheep W, Maiuthed A. Benja-ummarit induces ferroptosis with cell ballooning feature through ROS and iron-dependent pathway in hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118672. [PMID: 39127118 DOI: 10.1016/j.jep.2024.118672] [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: 04/10/2024] [Revised: 07/22/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Benja-ummarit (BU), a traditional Thai herbal formula, has been prescribed by traditional Thai practitioners for the treatment of liver cancer. Clinical trials of BU have shown an increase in overall survival in hepatocellular carcinoma (HCC) patients, including stage 1-3 (with or without prior standard chemotherapy) and terminal stage. The clinical outcomes differ from those of other apoptosis-based conventional chemotherapies. The molecular mechanisms underlying the anti-cancer properties of BU remain unclear. AIM OF STUDY To investigate BU-induced ferroptosis through morphological and molecular analyses of HCC cell lines and HCC rat tissues. METHODOLOGY Cytotoxicity of BU extract in HepG2 and HuH-7 cells, with or without LX-2 in 2D and 3D cultures, was determined through MTT assay and by observing spheroid formation, respectively, as compared to sorafenib. Morphological changes and the cellular ultrastructure of the treated cells were evaluated by light microscopy and transmission electron microscopy (TEM), respectively. In addition, alterations in ferroptosis protein markers in both cell lines and rat liver tissue were determined using western blot analysis and immunohistochemical staining, respectively. To investigate the pathways mediating ferroptosis, cells were pretreated with an iron chelator to confirm the iron-dependent ferroptosis induced by the BU extract. Intracellular ROS, a mediator of ferroptosis, was measured using a scanning ion conductance microscope (SICM). SICM was also used to determine cellular stiffness. The lipid profiles of BU-treated cells were studied using LC-MS/MS. RESULTS The BU extract induced cell death under all HCC cell culture conditions. The BU-IC50 in HepG2 and HuH-7 were 31.24 ± 4.46 μg/mL and 23.35 ± 0.27 μg/mL, respectively as determined by MTT assay. In co-culture with LX-2, BU exhibited a similar trend of cytotoxicity in both HepG2 and HuH-7 cells. Light microscopy showed cell ballooning features with intact plasma membranes, and TEM microscopy showed mitochondrial swelling and reduced mitochondrial cristae in BU-treated cells. BU promotes intracellular iron levels by increasing DMT1 and NCOA4 expression and decreasing FTH1 expression. BU also suppressed the cellular antioxidant system by lowering CD98, NRF2, and GPX4 expression, and promoting KEAP1 expression. IHC results of HCC rat liver tissues showed the absence of DMT1 and high expression of GPX4 in the tumor area. Pre-treatment with an iron chelator partially restored cell viability and shifted the mode of cell death to a more apoptosis-like morphology in the BU-treated group. The SICM showed increased intracellular ROS levels and cellular stiffness 24 h after BU treatment. In more detail of BU-mediated ferroptosis, cellular lipid profiling revealed increased expression of 3 polyunsaturated lipids, which are highly susceptible to lipid peroxidation, in BU-treated cells. DISCUSSION Alterations in intracellular iron levels, ROS levels, and cellular lipid composition have been previously reported in cancer cells. Therefore, targeting the iron-dependent ROS pathway and polyunsaturated lipids via BU-induced ferroptosis may be more cancer-specific than apoptosis-based cancer drugs. These observations are in accordance with the clinical outcomes of BU. The ferroptosis-inducing mechanism of BU makes it an extremely promising novel drug candidate for the treatment of HCC.
Collapse
Affiliation(s)
- Nichawadee Sandech
- Doctor of Philosophy Program in Innovative Anatomy, Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand; Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Meng Chieh Yang
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Pichakorn Juntranggoor
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Pattarawit Rukthong
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Srinakharinwirot University, Nakornnayok, 26120, Thailand; Center for Excellence in Plant and Herbal Innovation Research, Strategic Wisdom and Research Institute, Srinakharinwirot University, Nakornnayok, 26120, Thailand
| | - Petr Gorelkin
- ICAPPIC Limited, London, E8 3PN, United Kingdom; Research laboratory of biophysics, National University of Science and Technology (MISIS), Moscow, 119049, Russia
| | - Nikita Savin
- Research laboratory of biophysics, National University of Science and Technology (MISIS), Moscow, 119049, Russia
| | - Roman Timoshenko
- Research laboratory of biophysics, National University of Science and Technology (MISIS), Moscow, 119049, Russia
| | - Alexander Vaneev
- Research laboratory of biophysics, National University of Science and Technology (MISIS), Moscow, 119049, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexander Erofeev
- Research laboratory of biophysics, National University of Science and Technology (MISIS), Moscow, 119049, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Surasak Wichaiyo
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand; Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Wisuit Pradidarcheep
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand.
| | - Arnatchai Maiuthed
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand; Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
2
|
Lyu L, Wen H, Li Y, Wang X, Li J, Zuo C, Yan S, Qi X. PGE2 functions in ovoviviparous teleost black rockfish (Sebastes schlegelii): evolutionary status between parturition and ovulation†. Biol Reprod 2024; 110:140-153. [PMID: 37812450 DOI: 10.1093/biolre/ioad135] [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] [Indexed: 10/10/2023] Open
Abstract
Fish have evolved various reproductive strategies including oviparity, viviparity, and ovoviviparity, which undoubtedly affect the survival of the whole species continuity. As the final step in reproduction, parturition in viviparous vertebrate and ovulation in oviparous teleost seem to share a similar mechanism, when prostaglandins (PGs) act as the trigger to launch the whole process. In the present study, ovoviviparous teleost black rockfish (Sebastes schlegelii) is employed as the research object. Intraperitoneal injection showed that PGE2 (500 μg/kg) could activate the delivery reactions in perinatal black rockfish. RNA-seq data of ovary in perinatal period revealed transcriptional change in cell junction, inflammation, and apoptosis, which is related to mammal parturition and teleost ovulation. Further results proved the positive correlation between ptger EP2 and previous mentioned pathways. Subsequent experiment proved that PGE2 was able to induce the ovulation and spawning in unfertilized individuals, which had a bilayer follicular structure compared to monolayer follicular in perinatal period black rockfish. Both unfertilized and perinatal ovary matrix could response to PGE2 stimulation. In conclusion, the function of PGE2 in activating both parturition and ovulation in a relatively different pathways conserved with viviparity or oviparity provided novel evidence of the evolutionary status of ovoviviparous vertebrates.
Collapse
Affiliation(s)
- Likang Lyu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Haishen Wen
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Yun Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Xiaojie Wang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Jianshuang Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Chenpeng Zuo
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Shaojing Yan
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Xin Qi
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| |
Collapse
|
3
|
Nagano T, Tsuda N, Fujimura K, Ikezawa Y, Higashi Y, Kimura SH. Prostaglandin E 2 increases the expression of cyclooxygenase-2 in cultured rat microglia. J Neuroimmunol 2021; 361:577724. [PMID: 34610503 DOI: 10.1016/j.jneuroim.2021.577724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/21/2022]
Abstract
Prostaglandin E2 (PGE2) plays pivotal roles in controlling microglial activation with the EP2 receptor, a PGE2 receptor subtype. Activated microglia are often reported to increase cyclooxygenase (COX)-2 expression, followed by PGE2 production, but it is unclear whether extracellular PGE2 is involved in microglial PGE2 synthesis. In the present study, we report that PGE2 increases COX-2 protein in microglia. In a culture system, PGE2 at 10-6 M for 3 h increased COX-2 and microsomal PGE synthase (mPGES)-1 mRNA levels, and reduced mPGES-2, but did not affect COX-1 or cytosolic PGE synthase (cPGES) in microglia. PGE2 at 10-6 M for 3 h also increased the COX-2 protein level, but did not affect COX-1, mPGES-1, mPGES-2, or cPGES. An EP2 agonist, ONO-AE1-259-01, also increased COX-2 and mPGES-1 mRNA levels, and reduced mPGES-2, but did not affect COX-1 or cPGES, whereas an EP1 agonist, ONO-DI-004, an EP3 agonist, ONO-AE-248, and an EP4 agonist, ONO-AE1-329, had no effect. Similar to PGE2, ONO-AE1-259-01 increased the COX-2 protein level, but did not affect COX-1, mPGES-1, mPGES-2, or cPGES. In addition, the effects of PGE2 were inhibited by an EP2 antagonist, PF-04418948, but not by an EP1 antagonist, ONO-8713, an EP3 antagonist, ONO-AE3-240, or an EP4 antagonist, ONO-AE3-208, at 10-6 M. On the other hand, lipopolysaccharide (LPS) increased PGE2 production, but the LPS-induced PGE2 production was not affected by ONO-8713, PF-04418948, ONO-AE3-240, or ONO-AE3-208. These results indicate that PGE2 increases COX-2 protein in microglia through the EP2 receptor supporting the idea that extracellular PGE2 has a triggering aspect for microglial activation.
Collapse
Affiliation(s)
- Takayuki Nagano
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| | - Naohiko Tsuda
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Kenichi Fujimura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yuji Ikezawa
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yuki Higashi
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shinya H Kimura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
4
|
Intracellular prostaglandin E2 contributes to hypoxia-induced proximal tubular cell death. Sci Rep 2021; 11:7047. [PMID: 33782420 PMCID: PMC8007803 DOI: 10.1038/s41598-021-86219-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Proximal tubular cells (PTC) are particularly vulnerable to hypoxia-induced apoptosis, a relevant factor for kidney disease. We hypothesized here that PTC death under hypoxia is mediated by cyclo-oxygenase (COX-2)-dependent production of prostaglandin E2 (PGE2), which was confirmed in human proximal tubular HK-2 cells because hypoxia (1% O2)-induced apoptosis (i) was prevented by a COX-2 inhibitor and by antagonists of prostaglandin (EP) receptors and (ii) was associated to an increase in intracellular PGE2 (iPGE2) due to hypoxia-inducible factor-1α-dependent transcriptional up-regulation of COX-2. Apoptosis was also prevented by inhibitors of the prostaglandin uptake transporter PGT, which indicated that iPGE2 contributes to hypoxia-induced apoptosis (on the contrary, hypoxia/reoxygenation-induced PTC death was exclusively due to extracellular PGE2). Thus, iPGE2 is a new actor in the pathogenesis of hypoxia-induced tubular injury and PGT might be a new therapeutic target for the prevention of hypoxia-dependent lesions in renal diseases.
Collapse
|
5
|
Zhu W, Chang L, Zhao T, Wang B, Jiang J. Remarkable metabolic reorganization and altered metabolic requirements in frog metamorphic climax. Front Zool 2020; 17:30. [PMID: 33062031 PMCID: PMC7542913 DOI: 10.1186/s12983-020-00378-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Background Metamorphic climax is the crucial stage of amphibian metamorphosis responsible for the morphological and functional changes necessary for transition to a terrestrial habitat. This developmental period is sensitive to environmental changes and pollution. Understanding its metabolic basis and requirements is significant for ecological and toxicological research. Rana omeimontis tadpoles are a useful model for investigating this stage as their liver is involved in both metabolic regulation and fat storage. Results We used a combined approach of transcriptomics and metabolomics to study the metabolic reorganization during natural and T3-driven metamorphic climax in the liver and tail of Rana omeimontis tadpoles. The metabolic flux from the apoptotic tail replaced hepatic fat storage as metabolic fuel, resulting in increased hepatic amino acid and fat levels. In the liver, amino acid catabolism (transamination and urea cycle) was upregulated along with energy metabolism (TCA cycle and oxidative phosphorylation), while the carbohydrate and lipid catabolism (glycolysis, pentose phosphate pathway (PPP), and β-oxidation) decreased. The hepatic glycogen phosphorylation and gluconeogenesis were upregulated, and the carbohydrate flux was used for synthesis of glycan units (e.g., UDP-glucuronate). In the tail, glycolysis, β-oxidation, and transamination were all downregulated, accompanied by synchronous downregulation of energy production and consumption. Glycogenolysis was maintained in the tail, and the carbohydrate flux likely flowed into both PPP and the synthesis of glycan units (e.g., UDP-glucuronate and UDP-glucosamine). Fatty acid elongation and desaturation, as well as the synthesis of bioactive lipid (e.g., prostaglandins) were encouraged in the tail during metamorphic climax. Protein synthesis was downregulated in both the liver and tail. The significance of these metabolic adjustments and their potential regulation mechanism are discussed. Conclusion The energic strategy and anabolic requirements during metamorphic climax were revealed at the molecular level. Amino acid made an increased contribution to energy metabolism during metamorphic climax. Carbohydrate anabolism was essential for the body construction of the froglets. The tail was critical in anabolism including synthesizing bioactive metabolites. These findings increase our understanding of amphibian metamorphosis and provide background information for ecological, evolutionary, conservation, and developmental studies of amphibians.
Collapse
Affiliation(s)
- Wei Zhu
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, No.9, Section4, South Renmin Road, Chengdu, 610041 Sichuan China
| | - Liming Chang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, No.9, Section4, South Renmin Road, Chengdu, 610041 Sichuan China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Tian Zhao
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, No.9, Section4, South Renmin Road, Chengdu, 610041 Sichuan China
| | - Bin Wang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, No.9, Section4, South Renmin Road, Chengdu, 610041 Sichuan China
| | - Jianping Jiang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, No.9, Section4, South Renmin Road, Chengdu, 610041 Sichuan China
| |
Collapse
|
6
|
Teixeira-Pinheiro LC, Toledo MF, Nascimento-Dos-Santos G, Mendez-Otero R, Mesentier-Louro LA, Santiago MF. Paracrine signaling of human mesenchymal stem cell modulates retinal microglia population number and phenotype in vitro. Exp Eye Res 2020; 200:108212. [PMID: 32910940 DOI: 10.1016/j.exer.2020.108212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/13/2020] [Accepted: 08/31/2020] [Indexed: 01/13/2023]
Abstract
PURPOSE Cellular therapy with mesenchymal stem cells (MSC) is emerging as an effective option to treat optic neuropathies. In models of retinal degeneration, MSC injected in the vitreous body protects injured retinal ganglion cells and stimulate their regeneration, however the mechanism is still unknown. Considering the immunomodulating proprieties of MSC and the controversial role of microglial contribution on retinal regeneration, we developed an in vitro co-culture model to analyze the effect of MSC on retinal microglia population. METHODS We used whole adult rat retinal explants in co-culture with human Wharton's jelly mesenchymal stem cells (hMSC) separated by a transwell membrane and analyzed hMSC effect on both retinal ganglion cells (RGCs) and retinal microglia. RESULTS hMSC in co-culture protected RGCs after 3 days in vitro by paracrine signaling. In addition, hMSC reduced microglia population and inhibited the pro-inflammatory phenotype of the remaining microglia. CONCLUSIONS Using a co-culture model, we demonstrated the paracrine effect of hMSC on RGC survival after injury concomitant with a reduction of microglial population. Paracrine signaling of hMSC also changed microglia phenotype and the expression of antiinflammatory factors in the retina. Our results are consistent with a detrimental effect of microglia on RGC survival and regeneration after injury.
Collapse
Affiliation(s)
- Leandro C Teixeira-Pinheiro
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil.
| | - Maria F Toledo
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil
| | | | - Rosalia Mendez-Otero
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil
| | - Louise A Mesentier-Louro
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Department of Ophthalmology, Stanford University, Palo Alto, USA
| | - Marcelo F Santiago
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Brazil
| |
Collapse
|
7
|
Caspase-11 promotes allergic airway inflammation. Nat Commun 2020; 11:1055. [PMID: 32103022 PMCID: PMC7044193 DOI: 10.1038/s41467-020-14945-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/12/2020] [Indexed: 11/29/2022] Open
Abstract
Activated caspase-1 and caspase-11 induce inflammatory cell death in a process termed pyroptosis. Here we show that Prostaglandin E2 (PGE2) inhibits caspase-11-dependent pyroptosis in murine and human macrophages. PGE2 suppreses caspase-11 expression in murine and human macrophages and in the airways of mice with allergic inflammation. Remarkably, caspase-11-deficient mice are strongly resistant to developing experimental allergic airway inflammation, where PGE2 is known to be protective. Expression of caspase-11 is elevated in the lung of wild type mice with allergic airway inflammation. Blocking PGE2 production with indomethacin enhances, whereas the prostaglandin E1 analog misoprostol inhibits lung caspase-11 expression. Finally, alveolar macrophages from asthma patients exhibit increased expression of caspase-4, a human homologue of caspase-11. Our findings identify PGE2 as a negative regulator of caspase-11-driven pyroptosis and implicate caspase-4/11 as a critical contributor to allergic airway inflammation, with implications for pathophysiology of asthma. Caspase 11 activation involves transcriptional upregulation and proteolytic cleavage. Here the authors show that prostaglandin E2 prevents caspase-11-mediated pyroptosis, blocking caspase-11 mRNA and protein upregulation in macrophages and in vivo, and that mice lacking caspase-11 are strongly protected from allergic airway inflammation.
Collapse
|
8
|
García-Pastor C, Blázquez-Serra R, Bosch RJ, Lucio Cazaña FJ, Fernández-Martínez AB. Apoptosis and cell proliferation in proximal tubular cells exposed to apoptotic bodies. Novel pathophysiological implications in cisplatin-induced renal injury. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2504-2515. [DOI: 10.1016/j.bbadis.2019.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/10/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022]
|
9
|
Ju IG, Choi JG, Kim N, Kwak C, Lee JK, Oh MS. Peucedani Japonici Radix ameliorates lipopolysaccharide-induced neuroinflammation by regulating microglial responses. Neurosci Lett 2018; 686:161-167. [PMID: 30213621 DOI: 10.1016/j.neulet.2018.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/01/2018] [Accepted: 09/05/2018] [Indexed: 11/26/2022]
Abstract
Neuroinflammation is an inflammatory process within the central nervous system that is mediated by microglial activation, which releases pro-inflammatory mediators leading to neurodegeneration. In this study, we investigated the effects of Peucedani Japonici Radix (PJR), a medicinal herb traditionally used in East Asia to treat neuroinflammation both in vitro and in vivo. First, we examined the effects of PJR on pro-inflammatory mediators in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells. The results showed that PJR suppressed the LPS-induced increase of several inflammatory factors, such as nitric oxide, inducible nitric oxide synthase, cyclooxygenase-2, prostaglandin E2, interleukin-1β, and tumor necrosis factor-α. We also revealed that PJR inhibited the nuclear factor kappa B (NF-κB) pathway, which is the upstream modulator of inflammatory processes. Furthermore, to confirm the regulatory effects of PJR on microglia in vivo, we measured the number of ionized calcium-binding adapter molecule 1-positive cells in mouse brains and found that PJR treatment reduced microglial activation. Taken together, these results suggest that PJR inhibits microglia-mediated neuroinflammation through the modulation of NF-κB signaling and has the therapeutic potential to prevent inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- In Gyoung Ju
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jin Gyu Choi
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Namkwon Kim
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chaewon Kwak
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jong Kil Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dozgdaemun-gu, Seoul 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
10
|
Nagano T, Nishiyama R, Sanada A, Mutaguchi Y, Ioku A, Umeki H, Kishimoto S, Yamanaka D, Kimura SH, Takemura M. Prostaglandin E 2 potentiates interferon-γ-induced nitric oxide production in cultured rat microglia. J Neurochem 2017; 140:605-612. [PMID: 27973680 DOI: 10.1111/jnc.13926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/26/2022]
Abstract
Prostaglandin E2 (PGE2 ) plays crucial roles in managing microglial activation through the prostanoid EP2 receptor, a PGE2 receptor subtype. In this study, we report that PGE2 enhances interferon-γ (IFN-γ)-induced nitric oxide production in microglia. IFN-γ increased the release of nitrite, a metabolite of nitric oxide, which was augmented by PGE2 , although PGE2 by itself slightly affects nitrite release. The potentiating effect of PGE2 was positively associated with increased expression of inducible nitric oxide synthase. In contrast to nitrite release induced by IFN-γ, lipopolysaccharide-induced nitrite release was not affected by PGE2 . An EP2 agonist, ONO-AE1-259-01 also augmented IFN-γ-induced nitrite release, while an EP1 agonist, ONO-DI-004, an EP3 agonist, ONO-AE-248, or an EP4 agonist, ONO-AE1-329, did not. In addition, the potentiating effect of PGE2 was inhibited by an EP2 antagonist, PF-04418948, but not by an EP1 antagonist, ONO-8713, an EP3 antagonist, ONO-AE3-240, or an EP4 antagonist, ONO-AE3-208, at 10-6 M. Among the EP agonists, ONO-AE1-259-01 alone was able to accumulate cyclic adenosine monophosphate (AMP), and among the EP antagonists, PF-04418948 was the only one able to inhibit PGE2 -increased intracellular cyclic AMP accumulation. On the other hand, IFN-γ promoted phosphorylation of signal transducer and activator of transcription 1, which was not affected by PGE2 . Furthermore, other prostanoid receptor agonists, PGD2 , PGF2α , iloprost, and U-46119, slightly affected IFN-γ-induced nitrite release. These results indicate that PGE2 potentiates IFN-γ-induced nitric oxide production in microglia through the EP2 receptor, which may shed light on one of the pro-inflammatory aspects of PGE2 .
Collapse
Affiliation(s)
- Takayuki Nagano
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ryo Nishiyama
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ayaka Sanada
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yukiko Mutaguchi
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Anna Ioku
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hirohisa Umeki
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Satoshi Kishimoto
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Daisuke Yamanaka
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shinya H Kimura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Motohiko Takemura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
11
|
Maślanka T, Chrostowska M, Otrocka-Domagała I, Snarska A, Mikiewicz M, Zuśka-Prot M, Jasiecka A, Ziółkowski H, Markiewicz W, Jaroszewski JJ. Prostaglandin E2 exerts the proapoptotic and antiproliferative effects on bovine NK cells. Res Vet Sci 2016; 107:80-87. [DOI: 10.1016/j.rvsc.2016.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/05/2016] [Accepted: 05/22/2016] [Indexed: 12/21/2022]
|
12
|
Przybysz J, Chrostowska M, Ziółkowski H, Jaroszewski JJ, Maślanka T. The influence of prostaglandin E2 on the production of IFN-γ by bovine CD4+, CD8+ and WC1+ T cells. Res Vet Sci 2016; 105:31-5. [DOI: 10.1016/j.rvsc.2016.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/07/2016] [Accepted: 01/13/2016] [Indexed: 01/01/2023]
|
13
|
Intracellular prostaglandin E2 mediates cisplatin-induced proximal tubular cell death. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:293-302. [DOI: 10.1016/j.bbamcr.2015.11.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/11/2015] [Accepted: 11/30/2015] [Indexed: 01/15/2023]
|
14
|
Fu Y, Yang MS, Jiang J, Ganesh T, Joe E, Dingledine R. EP2 Receptor Signaling Regulates Microglia Death. Mol Pharmacol 2015; 88:161-70. [PMID: 25715797 DOI: 10.1124/mol.115.098202] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/20/2015] [Indexed: 11/22/2022] Open
Abstract
The timely resolution of inflammation prevents continued tissue damage after an initial insult. In the brain, the death of activated microglia by apoptosis has been proposed as one mechanism to resolve brain inflammation. How microglial death is regulated after activation is still unclear. We reported that exposure to lipopolysaccharide (LPS) and interleukin (IL)-13 together initially activates and then kills rat microglia in culture by a mechanism dependent on cyclooxygenase-2 (COX-2). We show here that activation of the E prostanoid receptor 2 (EP2, PTGER2) for prostaglandin E2 mediates microglial death induced by LPS/IL-13, and that EP2 activation by agonist alone kills microglia. Both EP2 antagonists and reactive oxygen scavengers block microglial death induced by either LPS/IL-13 or EP2 activation. By contrast, the homeostatic induction of heme oxygenase 1 (Hmox1) by LPS/IL-13 or EP2 activation protects microglia. Both the Hmox1 inducer cobalt protoporphyrin and a compound that releases the Hmox1 product carbon monoxide (CO) attenuated microglial death produced by LPS/IL-13. Whereas CO reduced COX-2 protein expression, EP2 activation increased Hmox1 and COX-2 expression at both the mRNA and protein level. Interestingly, caspase-1 inhibition prevented microglial death induced by either LPS/IL-13 or low (but not high) concentrations of butaprost, suggestive of a predominantly pyroptotic mode of death. Butaprost also caused the expression of activated caspase-3 in microglia, pointing to apoptosis. These results indicate that EP2 activation, which initially promotes microglial activation, later causes delayed death of activated microglia, potentially contributing to the resolution phase of neuroinflammation.
Collapse
Affiliation(s)
- Yujiao Fu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Myung-Soon Yang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Jianxiong Jiang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Thota Ganesh
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Eunhye Joe
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Raymond Dingledine
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| |
Collapse
|