1
|
Kim U, Kim DH, Oh DK, Shin HY, Lee CH. Gene Expression and Metabolome Analysis Reveals Anti-Inflammatory Impacts of 11,17diHDoPE on PM10-Induced Mouse Lung Inflammation. Int J Mol Sci 2024; 25:5360. [PMID: 38791399 PMCID: PMC11121355 DOI: 10.3390/ijms25105360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Oxylipins, the metabolites of polyunsaturated fatty acids, are vital in regulating cell proliferation and inflammation. Among these oxylipins, specialized pro-resolving mediators notably contribute to inflammation resolution. Previously, we showed that the specialized pro-resolving mediators isomer 11,17dihydroxy docosapentaenoic acid (11,17diHDoPE) can be synthesized in bacterial cells and exhibits anti-inflammatory effects in mammalian cells. This study investigates the in vivo impact of 11,17diHDoPE in mice exposed to particulate matter 10 (PM10). Our results indicate that 11,17diHDoPE significantly mitigates PM10-induced lung inflammation in mice, as evidenced by reduced pro-inflammatory cytokines and pulmonary inflammation-related gene expression. Metabolomic analysis reveals that 11,17diHDoPE modulates inflammation-related metabolites such as threonine, 2-keto gluconic acid, butanoic acid, and methyl oleate in lung tissues. In addition, 11,17diHDoPE upregulates the LA-derived oxylipin pathway and downregulates arachidonic acid- and docosahexaenoic acid-derived oxylipin pathways in serum. Correlation analyses between gene expression and metabolite changes suggest that 11,17diHDoPE alleviates inflammation by interfering with macrophage differentiation. These findings underscore the in vivo role of 11,17diHDoPE in reducing pulmonary inflammation, highlighting its potential as a therapeutic agent for respiratory diseases.
Collapse
Affiliation(s)
- Uijin Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea;
| | - Dong-Hyuk Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.); (D.-K.O.)
| | - Deok-Kun Oh
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.); (D.-K.O.)
| | - Ha Youn Shin
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea;
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.); (D.-K.O.)
| |
Collapse
|
2
|
Zhou Q, He M, Jin Q, Gao S, Yang Z, Zhu P, Tan W, Liu L. Mechanism of action of Taohong Siwu decoction in the alleviation of primary dysmenorrhea. Front Med (Lausanne) 2024; 11:1343179. [PMID: 38751973 PMCID: PMC11095111 DOI: 10.3389/fmed.2024.1343179] [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: 11/23/2023] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
Background As one of the most common gynecological disorders, PD significantly impacts the quality of life for women. TSD, a well-known traditional Chinese medical prescription, has gained popularity for its use in treating gynecological cold coagulation and blood stasis syndromes such as PD. However, the lack of comprehensive data hinders our understanding of its molecular mechanism. Purpose The objective of the present study is to investigate the therapeutic effects of TSD on PD and elucidate its plausible mechanism. Methods HPLC was employed to confirm the presence of the principal metabolites of TSD. The rat model of PD was induced by OT exposure following IWM and EB pretreatment, and subsequently treated with TSD via gastric gavage. The effects and potential mechanisms of TSD on PD rats were explored, encompassing general behavior, morphological alterations in the uterus and ovaries, biochemical indicators in the uterus and serum, and levels of proteins related to the PI3K/AKT signaling pathway. Results Gallic acid, hydroxysafflower yellow A, albiflorin, paeoniflorin, and ferulic acid were determined to be the primary active metabolites of TSD. The pharmacological studies yielded results indicating the successful establishment of the PD model in rats. Additionally, TSD demonstrated its ability to protect PD rats by ameliorating general behavior, mitigating pathological damage to uterine and ovarian tissues, and modulating the expression levels of correlated factors (PGE2, PGF2α, Ca2+, TXB2, IL-6, TNF-α, NO, and COX-2) as well as p-PI3K/PI3K and p-AKT/AKT proteins. Conclusion TSD exhibited protective effects against PD in rats through its interaction with multiple targets including P13K/AKT signaling pathway, indicating that TSD holds therapeutic potential for PD treatment and providing evidence supporting the rational utilization of TSD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wenhong Tan
- Yunnan Yunzhong Institute of Nutrition and health, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Lu Liu
- Yunnan Yunzhong Institute of Nutrition and health, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
3
|
Tajbakhsh A, Yousefi F, Farahani N, Savardashtaki A, Reiner Ž, Jamialahmadi T, Sahebkar A. Molecular Mechanisms and Therapeutic Potential of Resolvins in Cancer - Current Status and Perspectives. Curr Med Chem 2024; 31:5898-5917. [PMID: 37497711 DOI: 10.2174/0929867331666230727100123] [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: 12/04/2022] [Revised: 05/26/2023] [Accepted: 06/15/2023] [Indexed: 07/28/2023]
Abstract
Resolvins are specialized pro-resolving mediators derived from omega-3 fatty acids that can suppress several cancer-related molecular pathways, including important activation of transcription parameters in the tumor cells and their microenvironment, inflammatory cell infiltration, cytokines as well as chemokines. Recently, an association between resolvins and an important anti-inflammatory process in apoptotic tumor cell clearance (efferocytosis) was shown. The inflammation status or the oncogene activation increases the risk of cancer development via triggering the transcriptional agents, including nuclear factor kappa-light-chain-enhancer of activated B cells by generating the pro-inflammatory lipid molecules and infiltrating the tumor cells along with the high level of pro-inflammatory signaling. These events can cause an inflammatory microenvironment. Resolvins might decrease the leukocyte influx into the inflamed tissues. It is widely accepted that resolvins prohibit the development of debris-triggered cancer via increasing the clearance of debris, especially by macrophage phagocytosis in tumors without any side effects. Resolvins D2, D1, and E1 might suppress tumor-growing inflammation by activation of macrophages clearance of cell debris in the tumor. Resolvin D5 can assist patients with pain during treatment. However, the effects of resolvins as anti-inflammatory mediators in cancers are not completely explained. Thus, based on the most recent studies, we tried to summarize the most recent knowledge on resolvins in cancers.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, School of Medicine, University of Zagreb, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Jeon KB, Park HM, Kim S, Kim NY, Lee TE, Oh DK, Yoon DY. Phorbal-12-mysristate-13-acetate-induced inflammation is restored by protectin DX through PPARγ in human promonocytic U937 cells. Life Sci 2024; 336:122288. [PMID: 38007146 DOI: 10.1016/j.lfs.2023.122288] [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: 08/27/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
AIMS Protectin DX (PDX), a specialized pro-resolving mediator, is an important pharmaceutical compound with potential antioxidant and inflammation-resolving effects. However, the fundamental mechanism by which PDX's ameliorate chronic inflammatory diseases has not yet been elucidated. This study aims to evaluate the anti-inflammatory properties and PPARγ-mediated mechanisms of PDX in phorbal-12-mysristate-13-acetate (PMA)-stimulated human promonocytic U937 cells. MAIN METHODS We confirmed the effects of PDX on expressions of pro-inflammatory cytokines, mediators, and CD14 using conventional PCR, RT-qPCR, ELISA, and flow cytometry. Using western blotting, immunofluorescence, and reactive oxygen species (ROS) determination, we observed that PDX regulated PMA-induced signaling cascades. Molecular docking analysis and a cellular thermal shift assay were conducted to verify the interaction between PDX and the proliferator-activated receptor-γ (PPARγ) ligand binding domain. Western blotting was then employed to explore the alterations in PPARγ expression levels and validate PDX as a PPARγ full agonist. KEY FINDINGS PDX attenuated protein and mRNA expression levels of interleukin-6, tumor necrosis factor-α, and cyclooxygenase-2 in PMA-treated U937 cells. PDX acts as a PPARγ agonist, exerting a modulating effect on the ROS/JNK/c-Fos signaling pathways. Furthermore, PDX reduced human monocyte differentiation antigen CD14 expression levels. SIGNIFICANCE PPARγ exhibits pro-resolving effects to regulate the excessive inflammation. These results suggest that PDX demonstrates the resolution of inflammation, indicating the potential for therapeutic targeting of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Kyeong-Bae Jeon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seonhwa Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Tae-Eui Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Deok-Kun Oh
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
5
|
Jeong M, Ju Y, Kwon H, Kim Y, Hyun KY, Choi GE. Protocatechuic Acid and Syringin from Saussurea neoserrata Nakai Attenuate Prostaglandin Production in Human Keratinocytes Exposed to Airborne Particulate Matter. Curr Issues Mol Biol 2023; 45:5950-5966. [PMID: 37504292 PMCID: PMC10378452 DOI: 10.3390/cimb45070376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023] Open
Abstract
Saussurea neoserrata Nakai offers a reliable and efficient source of antioxidants that can help alleviate adverse skin reactions triggered by air pollutants. Air pollutants, such as particulate matter (PM), have the ability to infiltrate the skin and contribute to the higher occurrence of cardiovascular, cerebrovascular, and respiratory ailments. Individuals with compromised skin barriers are particularly susceptible to the impact of PM since it can be absorbed more readily through the skin. This study investigated the impact of protocatechuic acid and syringin, obtained from the n-BuOH extract of S. neoserrata Nakai, on the release of PGE2 and PGD2 induced by PM10. Additionally, it examined the gene expression of the synthesis of PGE2 and PGD2 in human keratinocytes. The findings of this research highlight the potential of utilizing safe and efficient plant-derived antioxidants in dermatological and cosmetic applications to mitigate the negative skin reactions caused by exposure to air pollution.
Collapse
Affiliation(s)
- Myeongguk Jeong
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yeongdon Ju
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
- Medical Science Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hyeokjin Kwon
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yeeun Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Kyung-Yae Hyun
- Department of Clinical Laboratory Science, Dong-Eui University, Busan 47340, Republic of Korea
| | - Go-Eun Choi
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| |
Collapse
|
6
|
Lv J, Chang S, Chen HY, Zhou XY, Wang XY, Chen ZC, Chen BB, Qian RC, Li DW. A multi-channel responsive AuNP@COF core-shell nanoprobe for simultaneous subcellular profiling of multiple cancer biomarkers. Biosens Bioelectron 2023; 234:115325. [PMID: 37148801 DOI: 10.1016/j.bios.2023.115325] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/10/2023] [Accepted: 04/16/2023] [Indexed: 05/08/2023]
Abstract
The abnormal change in the expression profile of multiple cancer biomarkers is closely related to tumor progression and therapeutic effect. Due to their low abundance in living cells and the limitations of existing imaging techniques, simultaneous imaging of multiple cancer biomarkers has remained a significant challenge. Here, we proposed a multi-modal imaging strategy to detect the correlated expression of multiple cancer biomarkers, MUC1, microRNA-21 (miRNA-21) and reactive oxygen (ROS) in living cells, based on a porous covalent organic framework (COF) wrapped gold nanoparticles (AuNPs) core-shell nanoprobe. The nanoprobe is functionalized with Cy5-labeled MUC1 aptamer, a ROS-responsive molecule (2-MHQ), and a miRNA-21-response hairpin DNA tagged by FITC as the reporters for different biomarkers. The target-specific recognition can induce the orthogonal molecular change of these reporters, producing fluorescence and Raman signals for imaging the expression profiles of membrane MUC1 (red fluorescence channel), intracellular miRNA-21 (green fluorescence channel), and intracellular ROS (SERS channel). We further demonstrate the capability of the cooperative expression of these biomarkers, along with the activation of NF-κB pathway. Our research provides a robust platform for imaging multiple cancer biomarkers, with broad potential applications in cancer clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Jian Lv
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shuai Chang
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Hua-Ying Chen
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xin-Yue Zhou
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xiao-Yuan Wang
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zhen-Chi Chen
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Bin-Bin Chen
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Ruo-Can Qian
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Da-Wei Li
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology, Dynamic Chemistry School of Chemistry, Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
7
|
Bian Z, Cao C, Ding J, Ding L, Yu S, Zhang C, Liu Q, Zhu L, Li J, Zhang Y, Liu Y. Neuroprotective effects of PRG on Aβ 25-35-induced cytotoxicity through activation of the ERK1/2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116550. [PMID: 37120057 DOI: 10.1016/j.jep.2023.116550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phylloporia ribis (Schumach:Fr.)Ryvarden is a genus of needle Phellinus medicinal fungi, parasitic on the living rhizomes of hawthorn and pear trees. As a traditional Chinese medicine, Phylloporia ribis was used in folklore for long-term illness, weakness and memory loss in old age. Previous studies have shown that polysaccharides from Phylloporia ribis (PRG) significantly promoted synaptic growth in PC12 cells in a dose-dependent manner, exhibiting "NGF"-like neurotrophic activity. Aβ25-35 damage to PC12 cells produced neurotoxicity and decreased cell survival, and PRG reduced the apoptosis rate, suggesting that PRG has neuroprotective effects. The studies confirmed that PRG had the potential to be a neuroprotective agent, but its neuroprotective mechanism remained unclear. AIM OF THE STUDY We aimed to elucidate the neuroprotective effects of PRG in an Aβ25-35-induced Alzheimer's disease (AD) model. MATERIALS AND METHODS Highly-differentiated PC12 cells were treated with Aβ25-35 (AD model) and PRG, and were assessed for cellular apoptosis, inflammatory factors, oxidative stress, and kinase phosphorylation. RESULTS The results showed that the PRG groups effectively inhibited the neurotoxicity, mainly manifested by inhibiting mitochondrial oxidative stress, attenuating neuroinflammatory responses, and improving mitochondrial energy metabolism, eventually resulting in higher cell survival. The expression of p-ERK, p-CREB and BDNF proteins was increased in the PRG groups compared to the model group, which confirmed that PRG reversed the inhibition of the ERK pathway. CONCLUSION We provide evidence for neuroprotection conferred by PRG and its mechanism by inhibiting ERK1/2 hyper-phosphorylation, prevention of mitochondrial stress, and subsequent prevention of apoptosis. The study highlights PRG as a promising candidate with neuroprotective effects, the potential of which can be harnessed for identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Zhiying Bian
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chenzhen Cao
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; Health Surveillance Section, Junan County Center for Disease Control and Prevention, Linyi, 276600, China
| | - Jie Ding
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Liang Ding
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shuai Yu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chuanxiang Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Qian Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lihao Zhu
- Sishui Siheyuan Culture and Tourism Development Company, Ltd, Sishui, 273200, China
| | - Jing Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yongqing Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yuhong Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
8
|
Kim NY, Kim S, Park HM, Lim CM, Kim J, Park JY, Jeon KB, Poudel A, Lee HP, Oh SR, Ahn J, Yoon DY. Cinnamomum verum extract inhibits NOX2/ROS and PKCδ/JNK/AP-1/NF-κB pathway-mediated inflammatory response in PMA-stimulated THP-1 monocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154685. [PMID: 36753827 DOI: 10.1016/j.phymed.2023.154685] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/08/2022] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Cinnamomum verum J. Presl (Cinnamon) is widely used in the food and pharmaceutical industries. C. verum exhibits various biological activities. However, it is unclear whether C. verum can inhibit NOX, a major source of ROS generation, and exert anti-inflammatory and antioxidant effects in PMA-stimulated THP-1 cells. PURPOSE This study investigates the anti-inflammatory and antioxidant effects of C. verum in PMA-stimulated THP-1 cells. METHODS The MeOH extract of C. verum was analyzed using UPLC-QTOF/MS. Anti-inflammatory and antioxidant effects of C. verum extract were examined by DCF-DA staining, immunofluorescence staining, RT-PCR, and immunoblotting in PMA-stimulated THP-1 cells. RESULTS C. verum and its components, cinnamic acid and coumarin, significantly attenuated the expression of IL-1β, IL-8, CCL5, and COX-2 in PMA-stimulated THP-1. C. verum decreased ROS levels via NOX2 downregulation, as well as ameliorated plasma membrane translocation of PKCδ and decreased JNK phosphorylation. Besides, C. verum suppressed the nuclear translocation of AP-1 and NF-κB, which modulates diverse pro-inflammatory genes. CONCLUSION C. verum effectively inhibits inflammation and oxidative stress during monocyte-macrophage differentiation and downregulates inflammatory mediators via NOX2/ROS and PKCδ/JNK/AP-1/NF-κB signaling.
Collapse
Affiliation(s)
- Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Seonhwa Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Chae-Min Lim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jinju Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jae-Young Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyeong-Bae Jeon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Amrit Poudel
- Panchamrit Research Center, Pokhara-07, Kaski 33700, Nepal
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea.
| | - Jongmin Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea.
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
9
|
Kono M, Okuda T, Ishihara N, Hagino H, Tani Y, Okochi H, Tokoro C, Takaishi M, Ikeda H, Ishihara Y. Chemokine expression in human 3-dimensional cultured epidermis exposed to PM2.5 collected by cyclonic separation. Toxicol Res 2023; 39:1-13. [PMID: 36726829 PMCID: PMC9839915 DOI: 10.1007/s43188-022-00142-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
Fine particulate matter (PM2.5) exposure has a risk of inducing several health problems, especially in the respiratory tract. The skin is the largest organ of the human body and is therefore the primary target of PM2.5. In this study, we examined the effects of PM2.5 on the skin using a human 3-dimensional cultured epidermis model. PM2.5 was collected by cyclonic separation in Yokohama, Japan. Global analysis of 34 proteins released from the epidermis revealed that the chemokines, chemokine C-X-C motif ligand 1 (CXCL1) and interleukin 8 (IL-8), were significantly increased in response to PM2.5 exposure. These chemokines stimulated neutrophil chemotaxis in a C-X-C motif chemokine receptor 2-dependent manner. The oxidative stress and signal transducer and activator of transcription 3 pathways may be involved in the increased expression of CXCL1 and IL-8 in the human epidermis model. Interestingly, in the HaCaT human keratinocyte cell line, PM2.5 did not affect chemokine expression but did induce IL-6 expression, suggesting a different effect of PM2.5 between the epidermis model and HaCaT cells. Overall, PM2.5 could induce the epidermis to release chemokines, followed by neutrophil activation, which might cause an unregulated inflammatory reaction in the skin.
Collapse
Affiliation(s)
- Maori Kono
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871 Japan
- Product Assurance Division, Mandom Corporation, Osaka, 540-8530 Japan
| | - Tomoaki Okuda
- Faculty of Science and Technology, Keio University, Kanagawa, 223-8522 Japan
| | - Nami Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521 Japan
| | - Hiroyuki Hagino
- Japan Automobile Research Institute, Ibaraki, 305-0822 Japan
| | - Yuto Tani
- School of Creative Science and Engineering, Waseda University, Tokyo, 169-8555 Japan
| | - Hiroshi Okochi
- School of Creative Science and Engineering, Waseda University, Tokyo, 169-8555 Japan
| | - Chiharu Tokoro
- School of Creative Science and Engineering, Waseda University, Tokyo, 169-8555 Japan
| | - Masayuki Takaishi
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871 Japan
- Product Assurance Division, Mandom Corporation, Osaka, 540-8530 Japan
| | - Hidefumi Ikeda
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871 Japan
- Product Assurance Division, Mandom Corporation, Osaka, 540-8530 Japan
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521 Japan
| |
Collapse
|
10
|
Liu X, Meng X, Su X, Ren K, Ning C, Qi X, Zhang S. The mechanism of ginger and its processed products in the treatment of estradiol valerate coupled with oxytocin-induced dysmenorrhea in mice via regulating the TRP ion channel-mediated ERK 1/2/NF-κB signaling pathway. Food Funct 2022; 13:11236-11248. [PMID: 36222424 DOI: 10.1039/d2fo01845d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Ginger (Rhizoma zingiberis, RZ) has been used as a food, spice, supplement, flavoring agent, and as an edible herbal medicine. It is characterized by its pungency and aroma, and is rich in nutrients with remarkable pharmacological effects. It is used in traditional medicine clinics to treat diseases and symptoms, such as colds, headache, and primary dysmenorrhea (PD). In China, a variety of processed products of RZ are used as herbal medicines, such as baked ginger (BG) or ginger charcoal (GC) to treat different diseases and symptoms. However, the molecular mechanism of the therapeutic effect of RZ and its processed products (RZPPs, including BG or GC) against PD has not been well characterized. Moreover, whether the transient receptor potential (TRP) ion channels are involved in this process is not clear. In the present study, UHPLC-Q-TOF MS was adopted to analyse the differential quality markers (DQMs) between RZ and RZPPs. In addition, differential metabolomics (DMs) was acquired between RZ- and RZPPs-treated estradiol valerate coupled with an oxytocin-induced PD mouse uterus using untargeted metabolomics (UM). A correlation analysis between DQMs and DMs was also conducted. Benzenoids, lipids, and lipid-like molecules were the main DQMs between RZ and RZPPs. RZ and RZPPs were found to improve the pathological status of the uterus of a PD mouse, with significantly decreased serum levels of E2, PGF2α, TXB2 and remarkably increased levels of PROG and 6-keto-PGF1α. Moreover, RZ and RZPPs alleviated PD in mice via regulating the TRP ion channel-mediated ERK1/2/NF-κB signaling pathway. Our results indicate that the therapeutic effect of RZ and RZPPs against PD may be mediated by regulating the TRP ion channel-mediated ERK1/2/NF-κB signaling pathway, and provide a reference for the development of new dietary supplements or medicines.
Collapse
Affiliation(s)
- Xiaoqin Liu
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Xianglong Meng
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Xiaojuan Su
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Kele Ren
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Chenxu Ning
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Xiaoming Qi
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| | - Shuosheng Zhang
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030619, Shanxi, China.
- Shanxi Key Laboratory of Traditional Herbal Medicines Processing, Jinzhong 030619, Shanxi, China
| |
Collapse
|
11
|
Kim NY, Lim CM, Park HM, Kim J, Pham TH, Yang Y, Lee HP, Hong JT, Yoon DY. MMPP promotes adipogenesis and glucose uptake via binding to the PPARγ ligand binding domain in 3T3-L1 MBX cells. Front Pharmacol 2022; 13:994584. [PMID: 36339572 PMCID: PMC9634037 DOI: 10.3389/fphar.2022.994584] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/05/2022] [Indexed: 08/13/2023] Open
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARγ) is a transcription factor involved in adipogenesis, and its transcriptional activity depends on its ligands. Thiazolidinediones (TZDs), well-known PPARγ agonists, are drugs that improve insulin resistance in type 2 diabetes. However, TZDs are associated with severe adverse effects. As current therapies are not well designed, novel PPARγ agonists have been investigated in adipocytes. (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP) is known to have anti-arthritic, anti-inflammatory, and anti-cancer effects. In this study, we demonstrated the adipogenic effects of MMPP on the regulation of PPARγ transcriptional activity during adipocyte differentiation in vitro. MMPP treatment increased PPARγ transcriptional activity, and molecular docking studies revealed that MMPP binds directly to the PPARγ ligand binding domain. MMPP and rosiglitazone showed similar binding affinities to the PPARγ. MMPP significantly promoted lipid accumulation in adipocyte cells and increased the expression of C/EBPβ and the levels of p-AKT, p-GSK3, and p-AMPKα at an early stage. MMPP enhanced the expression of adipogenic markers such as PPARγ, C/EBPα, FAS, ACC, GLUT4, FABP4 and adiponectin in the late stage. MMPP also improved insulin sensitivity by increasing glucose uptake. Thus, MMPP, as a PPARγ agonist, may be a potential drug for type 2 diabetes and metabolic disorders, which may help increase adipogenesis and insulin sensitivity.
Collapse
Affiliation(s)
- Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Chae-Min Lim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Jinju Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Thu-Huyen Pham
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Young Yang
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| |
Collapse
|