1
|
Liu T, Zhang R, Jiang L, Zhou L, Zhang H, Liang F, Xiong P, Chen H, Wen T, Shen X, Xie C, Tian L. The potential application and molecular mechanisms of natural products in the treatment of allergic rhinitis: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155663. [PMID: 38759345 DOI: 10.1016/j.phymed.2024.155663] [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: 12/11/2023] [Revised: 03/21/2024] [Accepted: 04/20/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUNDS Allergic rhinitis (AR) is a non-infectious chronic inflammation of the nasal mucosa mainly mediated by immunoglobulin E (IgE) in atopic individuals after exposure to allergens. The application of AR guideline-recommended pharmacotherapies can rapidly relieve symptoms of AR but with poor long-term efficacy, and many of these therapies have side effects. Many natural products and their derivatives have shown potential therapeutic effects on AR with fewer side effects. OBJECTIVES This review aims to expand understanding of the roles and mechanisms of natural compounds in the treatment of AR and to highlight the importance of utilizing natural products in the treatment of AR. MATERIAL AND METHOD We conducted a systematic literature search using PubMed, Web of Science, Google Scholar, and Clinical Trials. The search was performed using keywords including natural products, natural compounds, bioproducts, plant extracts, naturally derived products, natural resources, allergic rhinitis, hay fever, pollinosis, nasal allergy. Comprehensive research and compilation of existing literature were conducted. RESULTS This article provided a comprehensive review of the potential therapeutic effects and mechanisms of natural compounds in the treatment of AR. We emphasized that natural products primarily exert their effects by modulating signalling pathways such as NF-κB, MAPKs, STAT3/ROR-γt/Foxp3, and GATA3/T-bet, thereby inhibiting the activation and expansion of allergic inflammation. We also discussed their toxicity and clinical applications in AR therapy. CONCLUSION Taken together, natural products exhibit great potential in the treatment of AR. This review is also expected to facilitate the application of natural products as candidates for treating AR. Furthermore, drug discovery based on natural products has a promising prospect in AR treatment.
Collapse
Affiliation(s)
- Ting Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Rong Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210004, China
| | - Luyun Jiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Li Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Hai Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Fangqi Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Peizheng Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Hongqing Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Tian Wen
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiaofei Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China.
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China.
| | - Li Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China.
| |
Collapse
|
2
|
Ju S, Tan Y, Wang Q, Zhou L, Wang K, Wen C, Wang M. Antioxidant and anti‑inflammatory effects of esculin and esculetin (Review). Exp Ther Med 2024; 27:248. [PMID: 38682114 PMCID: PMC11046185 DOI: 10.3892/etm.2024.12536] [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: 10/20/2023] [Accepted: 02/19/2024] [Indexed: 05/01/2024] Open
Abstract
Fraxinus chinensis Roxb is a deciduous tree, which is distributed worldwide and has important medicinal value. In Asia, the bark of Fraxinus chinensis Roxb is a commonly used traditional Chinese medicine called Qinpi. Esculetin is a coumarin compound derived from the bark of Fraxinus chinensis Roxb and its glycoside form is called esculin. The aim of the present study was to systematically review relevant literature on the antioxidant and anti-inflammatory effects of esculetin and esculin. Esculetin and esculin can promote the expression of various endogenous antioxidant proteins, such as superoxide dismutase, glutathione peroxidase and glutathione reductase. This is associated with the activation of the nuclear factor erythroid-derived factor 2-related factor 2 signaling pathway. The anti-inflammatory effects of esculetin and esculin are associated with the inhibition of the nuclear factor κ-B and mitogen-activated protein kinase inflammatory signaling pathways. In various inflammatory models, esculetin and esculin can reduce the expression levels of various proinflammatory factors such as tumor necrosis factor-α, interleukin (IL)-1β and IL-6, thereby inhibiting the development of inflammation. In summary, esculetin and esculin may be promising candidates for the treatment of numerous diseases associated with inflammation and oxidative stress, such as ulcerative colitis, acute lung and kidney injury, lung cancer, acute kidney injury.
Collapse
Affiliation(s)
- Shaohua Ju
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| | - Youli Tan
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| | - Qiang Wang
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| | - Ling Zhou
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| | - Kun Wang
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| | - Chenghong Wen
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| | - Mingjian Wang
- Department of Pharmacy, Affiliated Sport Hospital, Chengdu Sport University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
3
|
Leal LE, Moreira ES, Correia BL, Bueno PSA, Comar JF, de Sá-Nakanishi AB, Cuman RKN, Bracht A, Bersani-Amado CA, Bracht L. Comparative study of the antioxidant and anti-inflammatory effects of the natural coumarins 1,2-benzopyrone, umbelliferone and esculetin: in silico, in vitro and in vivo analyses. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:173-187. [PMID: 37395795 DOI: 10.1007/s00210-023-02606-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/25/2023] [Indexed: 07/04/2023]
Abstract
The aim of this work was to compare the anti-inflammatory and antioxidant effects of three natural coumarins: 1,2-benzopyrone, umbelliferone and esculetin. The antioxidant capacity of coumarins was evaluated using both chemical and biological in vitro assays. Chemical assays included DPPH and ABTS∙+ radical scavenging as well as ferric ion reducing ability power (FRAP) assay. Inhibition of mitochondrial ROS generation and lipid peroxidation in brain homogenates were used as biological in vitro assays. The experimental method of carrageenan-induced pleurisy in rats was used for the in vivo investigation of the anti-inflammatory activity. In silico molecular docking analysis was undertaken to predict the affinity of COX-2 to the coumarins. Considering the antioxidant capacity, esculetin was the most efficient one as revealed by all employed assays. Particularly, the mitochondrial ROS generation was totally abolished by the compound at low concentrations (IC50 = 0.57 μM). As for the anti-inflammatory effects, the COX-2 enzyme presented good affinities to the three coumarins, as revealed by the molecular docking analyses. However, considering the in vivo anti-inflammatory effects, 1,2-benzopyrone was the most efficient one in counteracting pleural inflammation and it potentiated the anti-inflammatory actions of dexamethasone. Umbelliferone and esculetin treatments failed to reduce the volume of pleural exudate. Overall, therefore, our results support the notion that this class of plant secondary metabolites displays promising effects in the prevention and/or treatment of inflammation and other diseases associated with oxidative stress, although the singularities regarding the type of the inflammatory process and pharmacokinetics must be taken into account.
Collapse
Affiliation(s)
- Luana Eloísa Leal
- Department of Biochemistry, State University of Maringá, Paraná, Brazil
| | | | | | | | | | | | | | - Adelar Bracht
- Department of Biochemistry, State University of Maringá, Paraná, Brazil
| | | | - Lívia Bracht
- Department of Biochemistry, State University of Maringá, Paraná, Brazil.
| |
Collapse
|
4
|
Ye Z, Zhao TSY, Li SB, Zhou XL, Luo Q, Qin JK, Liang CQ, Wang P, Ge GB. Synthesis and biological evaluation of esculetin derivatives as potential anti-HBV agents. Med Chem Res 2023; 32:899-909. [PMID: 37056462 PMCID: PMC10030075 DOI: 10.1007/s00044-023-03045-7] [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: 11/15/2022] [Accepted: 03/07/2023] [Indexed: 03/24/2023]
Abstract
Previous in vivo and in vitro studies revealed that esculetin (Fig. 1) has anti-hepatitis B virus (anti-HBV) activity as well as a protective effect on liver damage caused by duck hepatitis B virus. We designed and synthesized a series of esculetin derivatives, introduced side chains containing various amino groups into site 7 of the parent structure, and synthesized C-4 and C-8 substituted derivatives with the goal of investigating their anti-HBV activities. In vitro anti-HBV activity was performed against HepG2.2.15 cells by using Enzyme-Linked Immunosorbent Assay(ELISA) kit and cytotoxicity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay with lamivudine as the positive control. The results demonstrated that several compounds showed moderate anti-HBV activity, while the introduction of morpholine groups could significantly inhibit the expression of hepatitis B e antigen (HBeAg) and the introduction of the 2-methylimidazole group could significantly inhibit the expression of Hepatitis B surface antigen (HBsAg). Among all tested compounds, compound 4a demonstrated the best anti-HBeAg activity (IC50 = 15.8 ± 4.2 μM), while compound 6d demonstrated the best anti-HBsAg activity (IC50 = 21.4 ± 2.8 μM). Compounds 6b and 6c showed moderate anti-HBV activity and HBsAg inhibition. Compounds 4b showed moderate anti-HBV activity and an inhibitory effect on HBeAg. In addition, compounds 4a, 4c, 4d, 6b, 6c and 6d showed improved metabolic stability. This study provides useful guidance for the discovery of anti-HBV drugs, which merits further investigation.
Collapse
Affiliation(s)
- Zhen Ye
- grid.443385.d0000 0004 1798 9548College of Pharmacy, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Tong-Shi-Yao Zhao
- grid.443385.d0000 0004 1798 9548College of Biotechnology, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Shan-Bin Li
- grid.443385.d0000 0004 1798 9548College of Biotechnology, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Xian-Li Zhou
- grid.443385.d0000 0004 1798 9548College of Biotechnology, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Qin Luo
- grid.443385.d0000 0004 1798 9548Science Experiment Center, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Jiang-Ke Qin
- grid.459584.10000 0001 2196 0260State Key Laboratory for the Chemistry and MoLecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004 Guangxi China
| | - Cheng-Qin Liang
- grid.443385.d0000 0004 1798 9548College of Pharmacy, Guilin Medical University, Guilin, 541199 Guangxi China
| | - Ping Wang
- grid.412540.60000 0001 2372 7462Shanghai Frontiers Science Center for Traditional Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Guang-Bo Ge
- grid.412540.60000 0001 2372 7462Shanghai Frontiers Science Center for Traditional Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
5
|
Choi J, Yoo MJ, Park SY, Seol JW. Antitumor Effects of Esculetin, a Natural Coumarin Derivative, against Canine Mammary Gland Tumor Cells by Inducing Cell Cycle Arrest and Apoptosis. Vet Sci 2023; 10:vetsci10020084. [PMID: 36851388 PMCID: PMC9961495 DOI: 10.3390/vetsci10020084] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Mammary gland tumors are the most common neoplasms in female dogs, of which 50% are malignant. Esculetin, a coumarin derivative, reportedly induces death in different types of cancer cells. In this study, we explore the anticancer effects of esculetin against CMT-U27 and CF41.mg canine mammary gland tumor cells. Esculetin significantly inhibited the viability and migration of both CMT-U27 and CF41.mg cells in a dose- and time-dependent manner. Flow cytometric analysis and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay revealed increased numbers of annexin-V-positive cells and DNA fragmentation. Furthermore, a cell cycle analysis demonstrated that esculetin blocked the cell progression at the G0/G1 phase and the S phase in CMT-U27 and CF41.mg cells. These results were supported by a Western blot analysis, which revealed upregulated protein expression of cleaved caspase-3, a marker of apoptosis, and downregulated cyclin-dependent kinase 4 and cyclin D1 protein, the cell cycle regulators. In conclusion, this novel study proves that esculetin exerts in vitro antitumor effects by inducing apoptosis and cell cycle arrest in canine mammary gland tumors.
Collapse
Affiliation(s)
- Jawun Choi
- Correspondence: (J.C.); (J.-W.S.); Tel.: +82-63-850-0964 (J.-W.S.)
| | | | | | - Jae-Won Seol
- Correspondence: (J.C.); (J.-W.S.); Tel.: +82-63-850-0964 (J.-W.S.)
| |
Collapse
|
6
|
Febuxostat Alleviates Allergic Rhinitis by Inhibiting Inflammation and Monocyte Adhesion in Human Nasal Epithelial Cells via Regulating KLF6. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9092311. [PMID: 36118091 PMCID: PMC9477640 DOI: 10.1155/2022/9092311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022]
Abstract
Introduction Febuxostat is a novel inhibitor of xanthine oxidase that suppresses cell adhesion molecules-mediated (CAMs) inflammation by activating KLF6. In this study, we explored the therapeutic function and potential mechanisms of febuxostat against allergic rhinitis (AR). Methods We investigated the role of febuxostat through in vitro cell and in vivo animal experiments. Human nasal epithelial cells (hNECs) were cultured with histamine as an in vitro model. To establish the AR animal model, rats were exposed to ovalbumin. Rats were randomly grouped into control, model, 7.5 mg/kg febuxostat, and 15 mg/kg febuxostat groups. Results In the in vitro study, we found significantly increased release of lactate dehydrogenase, elevated production of inflammatory factors and chemokines, and upregulated CAMs in histamine-treated hNECs. However, these results were significantly reversed for the 10 and 20 μM febuxostat treatments. The enhanced adhesion between hNECs and monocytes induced by histamine was dramatically repressed by febuxostat. In the vivo experiments, we observed that febuxostat ameliorated the increased sneezing times, the number of nose scratching episodes, and elevated HE pathological scores as well as alleviated the inflammation in nasal mucous tissues of AR mice. We found that KLF6, which was downregulated in histamine-treated hNECs, was significantly upregulated by febuxostat. The inhibitory effects of febuxostat on the expression levels of CAMs and adhesion between histamine-treated hNECs and monocytes were significantly abolished by the knockdown of KLF6. Conclusion Febuxostat alleviates AR by inhibiting inflammation and monocyte adhesion in human nasal epithelial cells through the regulation of KLF6.
Collapse
|
7
|
Alshawaf E, Hammad MM, Marafie SK, Ali H, Al-Mulla F, Abubaker J, Mohammad A. Discovery of natural products to block SARS-CoV-2 S-protein interaction with Neuropilin-1 receptor: A molecular dynamics simulation approach. Microb Pathog 2022; 170:105701. [PMID: 35963279 PMCID: PMC9364730 DOI: 10.1016/j.micpath.2022.105701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 10/26/2022]
Abstract
Neuropilin-1 (NRP1) is a widely expressed cell surface receptor protein characterized by its pleiotropic function. Recent reports highlighted NRP1 as an additional entry point of the SARS-CoV-2 virus, enhancing viral infectivity by interacting with the S-protein of SARS-CoV-2. The ubiquitous distribution and mechanism of action of NRP1 enable the SARS-CoV-2 virus to attack multiple organs in the body simultaneously. Therefore, blocking NRP1 is a potential therapeutic approach against SARS-CoV-2 infection. The current study screened the South African natural compounds database (SANCDB) for molecules that can disrupt the SARS-CoV-2 S protein-NRP1 interaction as a potential antiviral target for SARS-CoV-2 cellular entry. Following excessive screening and validation analysis 3-O-Methylquercetin and Esculetin were identified as potential compounds to disrupt the S-protein-NRP1 interaction. Furthermore, to understand the conformational stability and dynamic features between NRP1 interaction with the selected natural products, we performed 200 ns molecular dynamics (MD) simulations. In addition, molecular mechanics-generalized Born surface area (MM/GBSA) was utilized to calculate the free binding energies of the natural products interacting with NRP1. 3-O-methylquercetin showed an inhibitory effect with binding energies ΔG of -25.52 ± 0.04 kcal/mol to NRP1, indicating the possible disruption of the NRP1-S-protein interaction. Our analysis demonstrated that 3-O-methylquercetin presents a potential antiviral compound against SARS-CoV-2 infectivity. These results set the path for future functional in-vitro and in-vivo studies in SARS-CoV-2 research.
Collapse
Affiliation(s)
- Eman Alshawaf
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait, 15462
| | - Maha M Hammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait, 15462
| | - Sulaiman K Marafie
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait, 15462
| | - Hamad Ali
- Faculty of Department of Medical Laboratory Sciences 2, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait University, Kuwait; Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait, 15462
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait, 15462
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait, 15462
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait, 15462.
| |
Collapse
|
8
|
Zhang L, Xie Q, Li X. Esculetin: A review of its pharmacology and pharmacokinetics. Phytother Res 2021; 36:279-298. [PMID: 34808701 DOI: 10.1002/ptr.7311] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/12/2021] [Accepted: 09/30/2021] [Indexed: 12/30/2022]
Abstract
Esculetin is a natural dihydroxy coumarin; it is mainly extracted from twig skin and the trunk bark of the Chinese herbal medicine Fraxinus rhynchophylla Hance. Emerging evidence suggests that esculetin has a wide range of pharmacological activities. Based on its fundamental properties, including antioxidant, antiinflammatory, antiapoptotic, anticancer, antidiabetic, neuroprotective, and cardiovascular protective activities, as well as antibacterial activity, among others, esculetin is expected to be a therapeutic drug for specific disease indications, such as cancer, diabetes, atherosclerosis, Alzheimer's disease (AD), Parkinson's disease (PD), nonalcoholic fatty liver disease (NAFLD), and other diseases. The oral bioavailability of esculetin was shown by studies to be low. The extensive glucuronidation was described to be the main metabolic pathway of esculetin and C-7 phenolic hydroxyl to be its major metabolic site. With the development of scientific research technology, the pharmacological effects of esculetin are identified and its potential for the treatment of diseases is demonstrated. The underlining mechanisms of action and biological activities as well as the pharmacokinetic data of the analyzed compound reported so far are highlighted in this review with the aim of becoming a proven, and applicable insight and reference for further studies on the utilization of esculetin.
Collapse
Affiliation(s)
- Linlin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingxuan Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Huang J, Chen X, Xie A. Formononetin ameliorates IL‑13‑induced inflammation and mucus formation in human nasal epithelial cells by activating the SIRT1/Nrf2 signaling pathway. Mol Med Rep 2021; 24:832. [PMID: 34590155 PMCID: PMC8503736 DOI: 10.3892/mmr.2021.12472] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/28/2021] [Indexed: 01/18/2023] Open
Abstract
Formononetin has proven to be anti‑inflammatory and able to alleviate symptoms of certain allergic diseases. The present study aimed to determine and elucidate the potential effects of formononetin in allergic rhinitis. JME/CF15 cells were pretreated with formononetin at different doses, followed by stimulation with IL‑13. Cell Counting Kit‑8 assay was performed to determine the cytotoxicity of formononetin. The expression levels of inflammation‑related proteins, histamine, IgE, TNF‑α, IL‑1β, IL‑6, granulocyte‑macrophage colony‑stimulating factor and eotaxin in IL‑13‑stimulated JME/CF15 cells were detected using ELISAs. The expression levels of phosphorylated‑NF‑κB p65, NF‑κB p65 and cyclooxygenase‑2 (Cox‑2) were analyzed using western blotting. Reverse transcription‑quantitative PCR, western blotting and immunofluorescence were performed to measure the levels of mucin 5AC oligomeric mucus/gel‑forming. Expression levels of sirtuin 1 (SIRT1) and nuclear erythroid factor 2‑related factor 2 (Nrf2) proteins were also measured using western blotting. The results of the present study revealed that formononetin exerted no cytotoxic effect on the viability of JME/CF15 cells. Following stimulation of JME/CF15 cells with IL‑13, formononetin suppressed the upregulated expression levels of proinflammatory cytokines. IL‑13‑induced formation of mucus was also attenuated by formononetin treatment. Furthermore, it was found that the SIRT1/Nrf2 signaling pathway was activated in formononetin‑treated JME/CF15 cells, whereas treatment with the SIRT1 inhibitor, EX527, reversed the effects of formononetin on IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. In conclusion, the findings of the current study indicated that formononetin may activate the SIRT1/Nrf2 signaling pathway, thereby inhibiting IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. These results suggested that formononetin may represent a promising agent for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Juanjuan Huang
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xianfeng Chen
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Aihua Xie
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
10
|
Guo J, Xu S. Astragaloside IV suppresses histamine-induced inflammatory factors and mucin 5 subtype AC overproduction in nasal epithelial cells via regulation of inflammation-related genes. Bioengineered 2021; 12:6045-6056. [PMID: 34482800 PMCID: PMC8806810 DOI: 10.1080/21655979.2021.1965813] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Allergic rhinitis (AR) is a symptomatic allergic disease that leads to severe inflammation. Astragaloside IV (AS-IV) is a primary active component of Astragalus membranaceus and exerts immune-regulation and anti-inflammatory effects. However, the pharmacological effect of AS-IV in the nasal epithelial cells (NECs) has not been reported. The present study aimed to assess the effect of AS-IV on inflammatory cytokines and mucin 5 subtype AC (MUC5AC) overproduction in histamine (His)-stimulated NECs and its underlying mechanism. NECs were stimulated with or without His for 24 h in the absence or presence of AS-IV. The levels of inflammatory cytokines including IL-6, IL-8, MCP-1, IL-1β, granulocyte-macrophage colony-stimulating factor (GM-CSF), eotaxin, and MUC5AC were assayed. Our findings indicated that AS-IV inhibited His-evoked release and expression of inflammatory cytokines and MUC5AC in NECs. RNA-seq analyses indicated the significant changes in expression levels involved in inflammation genes upon treatment of His-induced NECs with AS-IV. Our findings indicated that AS-IV inhibited His-evoked inflammatory cytokines secretion and MUC5AC overproduction in NECs, which were partly mediated by regulation of inflammation-related genes. Therefore, our findings provided a scientific basis for the development of AS-IV as an effective agent for clinical therapeutic strategy in the treatment of AR.
Collapse
Affiliation(s)
- Jie Guo
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang Henan, China
| | - Shuai Xu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang Henan, China
| |
Collapse
|
11
|
Guo J, Peng L, Zeng J, Zhang M, Xu F, Zhang X, Wei Q. Paeoniflorin suppresses allergic and inflammatory responses by promoting autophagy in rats with urticaria. Exp Ther Med 2021; 21:590. [PMID: 33884028 PMCID: PMC8056118 DOI: 10.3892/etm.2021.10022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/01/2020] [Indexed: 01/02/2023] Open
Abstract
Paeoniflorin (PF) has been reported to be effective against several skin disorders, such as allergic contact dermatitis and psoriasis; however, it remains unclear whether PF can protect against urticarial lesions. Herein, the effects of PF on rats with urticarial lesions and the possible underlying mechanism were investigated. The effects of PF administration on a rat model of ovalbumin-induced urticarial-like lesions were evaluated via pathological analysis using hematoxylin-eosin staining. Toluidine blue staining was performed to detect mast cells and ELISA was performed to determine serum histamine levels. PF-induced regulatory effects on autophagic activity and the potential underlying mechanism of this were also investigated using transmission electron microscopy, immunohistochemistry and reverse transcription-quantitative PCR. It was demonstrated that PF suppressed allergic and inflammatory responses to improve urticarial lesions, as evidenced by the attenuation of pathological abnormalities, mast cell infiltration and histamine secretion. Mechanistically, PF treatment was found to markedly limit the production and release of inflammatory cytokine interleukin (IL)-23, while the levels of IL-17 remained unchanged. PF intervention led to an increased number of autophagosomes, along with higher levels of light chain 3B (LC3B) and Beclin-1, and lower levels of P62, indicating that PF could augment autophagic activity in urticarial lesions. PF treatment increased the expression of liver kinase B1 (LKB1) and AMP-activated protein kinase-α (AMPK-α), contributing to the PF-enhanced autophagic activity. In conclusion, PF could effectively improve urticarial lesions by inhibiting inflammatory cytokine IL-23 and increasing the autophagic activity via the LKB1/AMPK-α pathway.
Collapse
Affiliation(s)
- Jing Guo
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Li Peng
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Jinhao Zeng
- Geriatric Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China.,TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Meiheng Zhang
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Feng Xu
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiaotong Zhang
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Qin Wei
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
12
|
Boudreau A, Richard AJ, Harvey I, Stephens JM. Artemisia scoparia and Metabolic Health: Untapped Potential of an Ancient Remedy for Modern Use. Front Endocrinol (Lausanne) 2021; 12:727061. [PMID: 35211087 PMCID: PMC8861327 DOI: 10.3389/fendo.2021.727061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022] Open
Abstract
Botanicals have a long history of medicinal use for a multitude of ailments, and many modern pharmaceuticals were originally isolated from plants or derived from phytochemicals. Among these, artemisinin, first isolated from Artemisia annua, is the foundation for standard anti-malarial therapies. Plants of the genus Artemisia are among the most common herbal remedies across Asia and Central Europe. The species Artemisia scoparia (SCOPA) is widely used in traditional folk medicine for various liver diseases and inflammatory conditions, as well as for infections, fever, pain, cancer, and diabetes. Modern in vivo and in vitro studies have now investigated SCOPA's effects on these pathologies and its ability to mitigate hepatotoxicity, oxidative stress, obesity, diabetes, and other disease states. This review focuses on the effects of SCOPA that are particularly relevant to metabolic health. Indeed, in recent years, an ethanolic extract of SCOPA has been shown to enhance differentiation of cultured adipocytes and to share some properties of thiazolidinediones (TZDs), a class of insulin-sensitizing agonists of the adipogenic transcription factor PPARγ. In a mouse model of diet-induced obesity, SCOPA diet supplementation lowered fasting insulin and glucose levels, while inducing metabolically favorable changes in adipose tissue and liver. These observations are consistent with many lines of evidence from various tissues and cell types known to contribute to metabolic homeostasis, including immune cells, hepatocytes, and pancreatic beta-cells. Compounds belonging to several classes of phytochemicals have been implicated in these effects, and we provide an overview of these bioactives. The ongoing global epidemics of obesity and metabolic disease clearly require novel therapeutic approaches. While the mechanisms involved in SCOPA's effects on metabolic, anti-inflammatory, and oxidative stress pathways are not fully characterized, current data support further investigation of this plant and its bioactives as potential therapeutic agents in obesity-related metabolic dysfunction and many other conditions.
Collapse
Affiliation(s)
- Anik Boudreau
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Allison J. Richard
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Innocence Harvey
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Jacqueline M. Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
- *Correspondence: Jacqueline M. Stephens,
| |
Collapse
|