1
|
Chen Y, Liu J, Sun Y, Li M, Fan X, Gu X. Multi-omics study reveals Shuangshen Pingfei formula regulates EETs metabolic reprogramming to exert its therapeutic effect on pulmonary fibrosis. Int Immunopharmacol 2024; 143:113275. [PMID: 39395378 DOI: 10.1016/j.intimp.2024.113275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/05/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024]
Abstract
As a clinical formula derived from Renshen Pingfei San, Shuangshen Pingfei formula (SSPF) has been used to treat pulmonary fibrosis (PF). However, its in-depth mechanism of action remains unknown. In this study, the effect of SSPF was evaluated by applying a rat model of PF caused by intratracheal drip bleomycin. To characterize the molecular changes related to PF and reveal therapeutic targets for SSPF, we performed transcriptomic and metabolomic analyses on rat lung. Finally, western blotting and qPCR experiments were used to validate the multi-omics results. As a result, a significant reduction in inflammation and fibrosis caused by BLM was observed when SSPF was administered. Widespread changes in gene expression and metabolic programming were observed in the lungs of PF rats through RNA-seq and untargeted metabolomic analysis. Combined transcriptomic and metabolomic analyses revealed the involvement of arachidonic acid (AA) metabolism pathways in PF. Further validation of AA metabolite synthase genes and protein levels showed a significant decrease in the levels of epoxyeicosatrienoic acids (EETs) synthases, including Cyp2j2, Cyp2b1, in the PF lungs. SSPF treatment regulated the above changes in gene expression and metabolic programming, particularly the regulation of EETs. This study is the first to investigate the mechanism of action of SSPF in the treatment of PF from the perspective of regulating the synthesis of EETs in AA metabolism.
Collapse
Affiliation(s)
- Yeqing Chen
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jiayi Liu
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yubo Sun
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengwen Li
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinsheng Fan
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xin Gu
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Hwang YJ, Jung GS, Lee KM. Alantolactone alleviates epithelial-mesenchymal transition by regulating the TGF-β/STAT3 signaling pathway in renal fibrosis. Heliyon 2024; 10:e36253. [PMID: 39253189 PMCID: PMC11382038 DOI: 10.1016/j.heliyon.2024.e36253] [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: 11/01/2022] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Objective The epithelial-to-mesenchymal transition (EMT) of renal tubular epithelial cells (RTECs) plays a crucial role in renal interstitial fibrosis and inflammation, which are key components of chronic kidney disease (CKD). Alantolactone, a selective inhibitor of signal transducer and activator of transcription 3 (STAT3), is used in Chinese herbal medicine. Despite its use, the effects of alnatolactone on EMT of RTECs has not been fully elucidated. Methods In this study, we investigated the potential of alantolactone to EMT in vivo and in vitro. Our experiments were performed using a unilateral ureteral obstruction (UUO) models and HK-2 cells, RTECs, treated with transforming growth factor (TGF-β). Results Alantolactone decreased tubular injury and reduced the expression of vimentin, a key EMT marker, while increasing E-cadherin expression in UUO kidneys. Similarly, in RTECs, alantolactone inhibited TGF-β-induced EMT and its markers. Furthermore, alantolactone attenuated UUO- and TGF-β-induced STAT3 phosphorylation both in vivo and in vitro, and inhibited the expression of TWIST, an EMT transcription factor, in both models. Conclusion Alantolactone improves EMT in RTECs by inhibiting STAT3 phosphorylation and Twist expression, suggesting its potential as a therapeutic agent for kidney fibrosis.
Collapse
Affiliation(s)
- Yeo Jin Hwang
- Division of AI, Big Data and Block Chain, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea
| | - Gwon-Soo Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, Republic of Korea
| | - Kyeong-Min Lee
- Division of Biomedical Technology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea
| |
Collapse
|
3
|
Peng Y, Guo Y, Zhang S, Chang Y, Zhang S, Wang X, Zhao W, Ma X. Identification of diverse sesquiterpenoids with anti-fibrotic potential from Inula japonica Thunb. Bioorg Chem 2024; 143:107084. [PMID: 38176376 DOI: 10.1016/j.bioorg.2023.107084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
In the chemical investigation of Inula japonica, a total of 29 sesquiterpenoids (1-29) were obtained, including pseudoguaine-, xanthane-, eudesmane-, and 1,10-secoeudesmane-type compounds, as well as their dimers. Among them, six new dimeric sesquiterpenoids, bisinulains A-F (1-5, 7), characterized by a [4 + 2] biogenetic pathway between different sesquiterpenoid monomers were identified. Additionally, three new monomers named inulaterins A-C (13, 18 and 21) were discovered. The structures of these compounds were determined through analysis of spectroscopic data, X-ray crystallographic data, and ECD experiments. To assess their potential anti-inflammatory activities, the sesquiterpenoid dimers were tested for their ability to inhibit NO production in LPS-stimulated RAW 264.7 cells. Furthermore, the compounds that exhibited anti-inflammatory effects underwent evaluation for their anti-fibrotic potential using a TGF-β-induced epithelial-mesenchymal transition model in A549 cells. As a result, bisinulain B (2) was screened out to significantly inhibit the production of cytokines involved in pulmonary fibrosis such as NO, α-SMA, collagen I and fibronectin.
Collapse
Affiliation(s)
- Yulin Peng
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuxin Guo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Shuyuan Zhang
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China; The 967th Hospital of Joint Logistics Force, Dalian, China
| | - Yibo Chang
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shujing Zhang
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaobo Wang
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China; The 967th Hospital of Joint Logistics Force, Dalian, China.
| | - Wenyu Zhao
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Xiaochi Ma
- College (Institute) of Integrative Medicine, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
4
|
Wei F, Yin Y, Li J, Chang Y, Zhang S, Zhao W, Ma X. Essential oil from Inula japonica Thunb. And its phenolic constituents ameliorate pulmonary injury and fibrosis in bleomycin-treated mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117169. [PMID: 37704119 DOI: 10.1016/j.jep.2023.117169] [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: 07/05/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary injury and fibrosis can be caused by various factors because of their inflammatory nature, both can lead to serious clinical consequences. Inula japonica Thunb. is used in traditional Chinese medicine for the treatment of lung diseases. However, the effect and mechanism of action of the essential oil of I. japonica (EOI) on pulmonary injury and fibrosis are not well understood. AIM OF THE STUDY To investigate the therapeutic effects of EOI on mice with bleomycin (BLM)-induced acute pulmonary injury and chronic fibrosis formation, as well as its potential mechanism. MATERIALS AND METHODS A short-term mouse model of pulmonary injury was established by intratracheal injection of BLM to investigate the anti-inflammatory effect of EOI, and a long-term model of pulmonary fibrosis was used to explore the anti-fibrosis effect of EOI. High-dose EOI (200 mg/kg) was administered intragastrically, and low-dose (50 mg/kg) was administered by intratracheal injection. Gas chromatography-mass spectrometry (GC-MS) was used to identify the ingredients in EOI, and high-performance liquid chromatography (HPLC) was performed for the preparation of EOI compounds. Western blot and real-time qPCR were used to verify the effects of EOI and its active composition on inflammation, oxidative stress and fibrosis signaling pathway. RESULTS Treatment with EOI significantly reduced the inflammation and oxidative stress by reducing the levels of inflammatory and oxidative cytokines such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and malondialdehyde in BLM-treated mice with acute pulmonary injury. EOI treatment could also suppress the formation of fibrous tissue in mice with BLM-induced pulmonary fibrosis through inhibiting TGF-β/Smad and PI3K/Akt pathways. Chromatographic analysis and preparation suggested that fatty acid and phenol derivatives are present in EOI. Based on cellular inflammation and fibrosis models, the phenolic compounds in EOI can represent the anti-inflammatory and anti-fibrotic effects of EOI by regulating pro-inflammatory and pro-fibrotic cytokines such as NO, TNF-α, IL-6, TGF-β1, and α-SMA. CONCLUSION EOI ameliorated BLM-induced pulmonary injury and fibrosis in mice by inhibiting the inflammatory response and regulating the redox equilibrium, as well as by mediating TGFβ/Smad and PI3K/Akt, which suggested that EOI has potential to treat pulmonary diseases.
Collapse
Affiliation(s)
- Fan Wei
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuzhen Yin
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Li
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yibo Chang
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuyuan Zhang
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wenyu Zhao
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.
| | - Xiaochi Ma
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
5
|
Zhang J, Liu J, Liu JW, Zhu QM, Zhang M, Zhang R, Ma XC, Lv X, Yu ZL, Sun CP. Targeting Keap1 with Inulae Herba activated the Nrf2 receptor to alleviate LPS-mediated acute lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117358. [PMID: 37890806 DOI: 10.1016/j.jep.2023.117358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inulae Herba (IH) is known as Jinfeicao recorded in Chinese Pharmacopoeia with effects of lowering qi and eliminating phlegm, and used for the treatment of pulmonary diseases. However, its protective mechanism on pulmonary diseases, especially acute lung injury (ALI), is still undefined. AIM OF THE STUDY This study aimed to explore anti-inflammatory and anti-oxidation effects of IH and its underlying mechanism for treating ALI. MATERIALS AND METHODS We constructed a lipopolysaccharide (LPS)-ALI mouse model to reveal the therapeutical effect of IH. Western blot, real-time quantitative PCR, flow cytometry, small RNA interference, immunohistochemical staining, and the dual-luciferase experiment were performed to study the mechanism of IH for treating ALI. RESULTS IH attenuated LPS-mediated pathological changes (e.g. pneumonedema and pulmonary congestion) through inactivation of macrophages in an ALI mouse model. The result of flow cytometry demonstrated that IH regulated the homeostasis of M1 (CD80+CD206-) and M2 (CD80+CD206+) phenotype macrophages. Furthermore, IH suppressed mRNA expressions of M1 phenotype markers, such as iNOS and IL-6, whereas promoted mRNA expressions of M2 phenotype markers, such as ARG1 and RETNLA in LPS-mediated mice. Notably, IH targeted Keap1 to activate the Nrf2 receptor, exerting its anti-inflammatory and anti-oxidation effects proved by using immunohistochemical staining, dual-luciferase, and Keap1 knockdown technologies. CONCLUSION These findings suggested that targeting Keap1 with IH alleviated LPS-mediated ALI, and it could serve as a herbal agent for developing anti-ALI drugs.
Collapse
Affiliation(s)
- Juan Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China; School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518061, China
| | - Jing Liu
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Jing-Wen Liu
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Qi-Meng Zhu
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Min Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Rui Zhang
- School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiao-Chi Ma
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China.
| | - Xia Lv
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zhen-Long Yu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Cheng-Peng Sun
- Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China; School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
6
|
Jung Y, Yim NH, Lee SM, Cho WK, Cha MH, Ma JY. Anti-Fibrosis Effect of Panax ginseng and Inula japonica Formula in Human Pulmonary Fibroblasts. Nutrients 2024; 16:319. [PMID: 38276557 PMCID: PMC10819838 DOI: 10.3390/nu16020319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Panax ginseng Meyer and Inula japonica Thunb. are well established in traditional medicine and are known for their therapeutic properties in managing a range of ailments such as diabetes, asthma, and cancer. Although P. ginseng and I. japonica can alleviate pulmonary fibrosis (PF), the anti-fibrosis effect on PF by the combination of two herbal medicines remains unexplored. Therefore, this study explores this combined effect. In conditions that were not cytotoxic, MRC-5 cells underwent treatment using the formula combining P. ginseng and I. japonica (ISE081), followed by stimulation with transforming growth factor (TGF)-β1, to explore the fibroblast-to-myofibroblast transition (FMT). After harvesting the cells, mRNA levels and protein expressions associated with inflammation and FMT-related markers were determined to evaluate the antiinflammation activities and antifibrosis effect of ISE081. Additionally, the anti-migratory effects of ISE081 were validated through a wound-healing assay. ISE081 remarkably reduced the mRNA levels of interleukin (IL)-6, IL-8, α-smooth muscle actin (SMA), and TGF-β1 in MRC-5 cells and suppressed the α-SMA and fibronectin expressions, respectively. Furthermore, ISE081 inhibited Smad2/3 phosphorylation and wound migration of MRC-5 cells. Under the same conditions, comparing those of ISE081, P. ginseng did not affect the expression of α-SMA, fibronectin, and Smad2/3 phosphorylation, whereas I. japonica significantly inhibited them but with cytotoxicity. The results indicate that the synergistic application of P. ginseng and I. japonica enhances the anti-fibrotic properties in pulmonary fibroblasts and concurrently diminishes toxicity. Therefore, ISE081 has the potential as a prevention and treatment herbal medicine for PF.
Collapse
Affiliation(s)
- YeonGyun Jung
- Burn Institute, Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07247, Republic of Korea;
| | - Nam-Hui Yim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (W.-K.C.); (M.H.C.)
| | - Sang Myung Lee
- Division of Food and Pharmaceutical Technology, College of Health and Safety Science, Mokwon University, Daejeon 35349, Republic of Korea;
| | - Won-Kyung Cho
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (W.-K.C.); (M.H.C.)
| | - Min Ho Cha
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (W.-K.C.); (M.H.C.)
| | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (W.-K.C.); (M.H.C.)
| |
Collapse
|
7
|
Zhou YM, Dong XR, Xu D, Tang J, Cui YL. Therapeutic potential of traditional Chinese medicine for interstitial lung disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116952. [PMID: 37487964 DOI: 10.1016/j.jep.2023.116952] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/26/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Interstitial lung disease (ILD) is a chronic lung dysfunction disease with a poor prognosis and poor recovery. The clinically used therapeutic drugs, such as glucocorticoids and immunosuppressants, have no significant therapeutic effect and are accompanied with severe side effects. In recent years, considerable progress has been made in exploring and applying natural herb components for treating ILD. Traditional Chinese Medicine (TCM) possesses innate, non-toxic characteristics and offers advantages in preventing and treating pulmonary ailments. However, a comprehensive study of TCM on ILD therapy has not yet been reviewed. AIM OF THE REVIEW This review aimed to provide a comprehensive summary of the monomer components, total extracts, and prescriptions of TCM for ILD therapy, elucidating their molecular mechanisms to serve as a reference in treating ILD. MATERIALS AND METHODS The literature information was searched in the PubMed, Web of Science databases. The search keywords included 'interstitial lung disease', 'lung fibrosis' or 'pulmonary fibrosis', and 'traditional Chinese medicine', 'traditional herbal medicine', or 'herb medicine'. RESULTS The active components of single herbs, such as alkaloids, flavonoids, terpenoids, phenols, and quinones, have potential therapeutic effects on ILD. The active extracts and prescriptions were also summarized and analyzed. The herbs, Glycyrrhiza uralensis Fisch. (Gancao), Astragalus membranaceus Fisch. Bunge. (Huangqi) and Angelicasinensis (Oliv.) Diels (Danggui), play significant roles in the treatment of ILD. The mechanisms involve the inhibition of inflammatory factor release, anti-oxidative injury, and interference with collagen production, etc. CONCLUSION: This review examines the therapeutic potential of TCM for ILD and elucidates its molecular mechanisms, demonstrating that mitigating inflammation and oxidative stress, modulating the immune system, and promoting tissue repair are efficacious strategies for ILD therapy. The depth research will yield both theoretical and practical implications.
Collapse
Affiliation(s)
- Yan-Ming Zhou
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Xin-Ran Dong
- The Second Hospital of Tianjin Medical University, Tianjin, 300211, PR China
| | - Dong Xu
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China.
| | - Jie Tang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
8
|
Hao Y, Li J, Dan L, Wu X, Xiao X, Yang H, Zhou R, Li B, Wang F, Du Q. Chinese medicine as a therapeutic option for pulmonary fibrosis: Clinical efficacies and underlying mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116836. [PMID: 37406748 DOI: 10.1016/j.jep.2023.116836] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023]
Abstract
ETHNIC PHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is a fibrotic interstitial lung disease caused by continuous damage and excessive repair of alveolar epithelial cells, the pathogenesis of which is not fully understood. At present, the incidence of PF has increased significantly around the world. The therapeutic arsenals against PF are relatively limited, with often poor efficacy and many adverse effects. As a conventional and effective therapeutic strategy, traditional Chinese medicine (TCM) has been widely applied in treating lung fibrosis for thousands of years in China. Due to the multi-ingredient, multi-target characteristics, Chinese medicines possess promising clinical benefits for PF treatment. AIM OF THIS REVIEW This review aims to systematically analyze the clinical efficacy of Chinese medicine on PF, and further summarize the relevant mechanisms of Chinese medicine treating PF in preclinical studies, in order to provide a comprehensive insight into the beneficial effects of Chinese medicines on PF. METHODS Eight major Chinese and English databases were searched from database inception up to October 2022, and all randomized clinical trials (RCTs) investigating the effects of Chinese medicine intervention on effectiveness and safety in the treatment of PF patients were included. Subsequently, preclinical studies related to the treatment of PF in Chinese medicine, including Chinese medicine compounds, Chinese herbal materials and extracts, and Chinese herbal formulas (CHFs) were searched through PubMed and Web of science to summarize the related mechanisms of Chinese medicine against PF. RESULTS A total of 56 studies with 4019 patients were included by searching the relevant databases. Total clinical efficacy, pulmonary function, blood gas analysis, lung high resolution CT (HRCT), 6 min walk test (6-MWT), St George's Respiratory Questionnaire (SGRQ) scores, clinical symptom scores, TCM syndrome scores and other outcome indicators related to PF were analyzed. Besides, numerous preclinical studies have shown that many Chinese medicine compounds, Chinese herbal materials and extracts, and CHFs play a preventive and therapeutic role in PF by reducing oxidative stress, ameliorating inflammation, inhibiting epithelial-mesenchymal transition and myofibroblasts activation, and regulating autophagy and apoptosis. CONCLUSION Chinese medicines show potential as supplements or substitutes for treating PF. And studies on Chinese medicines will provide a new approach to better management of PF.
Collapse
Affiliation(s)
- Yanwei Hao
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jiaxin Li
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lijuan Dan
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xuanyu Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiang Xiao
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Han Yang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Rui Zhou
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Bin Li
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Fei Wang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Quanyu Du
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
9
|
Lee KM, Hwang YJ, Jung GS. Alantolactone Attenuates Renal Fibrosis via Inhibition of Transforming Growth Factor β/Smad3 Signaling Pathway. Diabetes Metab J 2024; 48:72-82. [PMID: 38173367 PMCID: PMC10850280 DOI: 10.4093/dmj.2022.0231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 01/25/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGRUOUND Renal fibrosis is characterized by the accumulation of extracellular matrix proteins and interstitial fibrosis. Alantolactone is known to exert anticancer, anti-inflammatory, antimicrobial and antifungal effects; however, its effects on renal fibrosis remains unknown. Here, we investigated whether alantolactone attenuates renal fibrosis in mice unilateral ureteral obstruction (UUO) and evaluated the effect of alantolactone on transforming growth factor (TGF) signaling pathway in renal cells. METHODS To evaluate the therapeutic effect of alantolactone, cell counting kit-8 (CCK-8) assay, histological staining, Western blot analysis, and real-time quantitative polymerase chain reaction were performed in UUO kidneys in vivo and in TGF-β-treated renal cells in vitro. RESULTS Alantolactone (0.25 to 4 µM) did not affect the viability of renal cells. Mice orally administered 5 mg/kg of alantolactone daily for 15 days did not show mortality or liver toxicity. Alantolactone decreased UUO-induced blood urea nitrogen and serum creatinine levels. In addition, it significantly alleviated renal tubulointerstitial damage and fibrosis and decreased collagen type I, fibronectin, and α-smooth muscle actin (α-SMA) expression in UUO kidneys. In NRK-49F cells, alantolactone inhibited TGF-βstimulated expression of fibronectin, collagen type I, plasminogen activator inhibitor-1 (PAI-1), and α-SMA. In HK-2 cells, alantolactone inhibited TGF-β-stimulated expression of collagen type I and PAI-1. Alantolactone inhibited UUO-induced phosphorylation of Smad3 in UUO kidneys. In addition, it not only decreased TGF-β secretion but also Smad3 phosphorylation and translocation to nucleus in both kidney cell lines. CONCLUSION Alantolactone improves renal fibrosis by inhibiting the TGF-β/Smad3 signaling pathway in obstructive nephropathy. Thus, alantolactone is a potential therapeutic agent for chronic kidney disease.
Collapse
Affiliation(s)
- Kyeong-Min Lee
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Yeo Jin Hwang
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Gwon-Soo Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| |
Collapse
|
10
|
Zhang J, Zhang WH, Morisseau C, Zhang M, Dong HJ, Zhu QM, Huo XK, Sun CP, Hammock BD, Ma XC. Genetic deletion or pharmacological inhibition of soluble epoxide hydrolase attenuated particulate matter 2.5 exposure mediated lung injury. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131890. [PMID: 37406527 PMCID: PMC10699546 DOI: 10.1016/j.jhazmat.2023.131890] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/27/2023] [Accepted: 06/17/2023] [Indexed: 07/07/2023]
Abstract
Air pollution represented by particulate matter 2.5 (PM2.5) is closely related to diseases of the respiratory system. Although the understanding of its mechanism is limited, pulmonary inflammation is closely correlated with PM2.5-mediated lung injury. Soluble epoxide hydrolase (sEH) and epoxy fatty acids play a vital role in the inflammation. Herein, we attempted to use the metabolomics of oxidized lipids for analyzing the relationship of oxylipins with lung injury in a PM2.5-mediated mouse model, and found that the cytochrome P450 oxidases/sEH mediated metabolic pathway was involved in lung injury. Furthermore, the sEH overexpression was revealed in lung injury mice. Interestingly, sEH genetic deletion or the selective sEH inhibitor TPPU increased levels of epoxyeicosatrienoic acids (EETs) in lung injury mice, and inactivated pulmonary macrophages based on the MAPK/NF-κB pathway, resulting in protection against PM2.5-mediated lung injury. Additionally, a natural sEH inhibitor luteolin from Inula japonica displayed a pulmonary protective effect towards lung injury mediated by PM2.5 as well. Our results are consistent with the sEH message and protein being both a marker and mechanism for PM2.5-induced inflammation, which suggest its potential as a pharmaceutical target for treating diseases of the respiratory system.
Collapse
Affiliation(s)
- Juan Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China; School of Pharmaceutical Sciences, Medical School, Shenzhen University, Shenzhen 518061, People's Republic of China
| | - Wen-Hao Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China; College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, United States
| | - Min Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China; College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Hong-Jun Dong
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Qi-Meng Zhu
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China; College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xiao-Kui Huo
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Cheng-Peng Sun
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China; College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China; School of Chinese Materia Medica, State Key Laboratory of Component-Based Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China.
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, United States.
| | - Xiao-Chi Ma
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, People's Republic of China.
| |
Collapse
|
11
|
Qin S, Tan P, Xie J, Zhou Y, Zhao J. A systematic review of the research progress of traditional Chinese medicine against pulmonary fibrosis: from a pharmacological perspective. Chin Med 2023; 18:96. [PMID: 37537605 PMCID: PMC10398979 DOI: 10.1186/s13020-023-00797-7] [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: 04/27/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
Pulmonary fibrosis is a chronic progressive interstitial lung disease caused by a variety of etiologies. The disease can eventually lead to irreversible damage to the lung tissue structure, severely affecting respiratory function and posing a serious threat to human health. Currently, glucocorticoids and immunosuppressants are the main drugs used in the clinical treatment of pulmonary fibrosis, but their efficacy is limited and they can cause serious adverse effects. Traditional Chinese medicines have important research value and potential for clinical application in anti-pulmonary fibrosis. In recent years, more and more scientific researches have been conducted on the use of traditional Chinese medicine to improve or reduce pulmonary fibrosis, and some important breakthroughs have been made. This review paper systematically summarized the research progress of pharmacological mechanism of traditional Chinese medicines and their active compounds in improving or reducing pulmonary fibrosis. We conducted a systematic search in several main scientific databases, including PubMed, Web of Science, and Google Scholar, using keywords such as idiopathic pulmonary fibrosis, pulmonary fibrosis, interstitial pneumonia, natural products, herbal medicine, and therapeutic methods. Ultimately, 252 articles were included and systematically evaluated in this analysis. The anti-fibrotic mechanisms of these traditional Chinese medicine studies can be roughly categorized into 5 main aspects, including inhibition of epithelial-mesenchymal transition, anti-inflammatory and antioxidant effects, improvement of extracellular matrix deposition, mediation of apoptosis and autophagy, and inhibition of endoplasmic reticulum stress. The purpose of this article is to provide pharmaceutical researchers with information on the progress of scientific research on improving or reducing Pulmonary fibrosis with traditional Chinese medicine, and to provide reference for further pharmacological research.
Collapse
Affiliation(s)
- Shanbo Qin
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Peng Tan
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| | - Junjie Xie
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Yongfeng Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junning Zhao
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| |
Collapse
|
12
|
Zhang M, Zhang J, Zhu QM, Zhao WY, Lv X, Yi J, Huo XK, Wang MJ, Sun CP. Inula japonica ameliorated the inflammation and oxidative stress in LPS-induced acute lung injury through the MAPK/NF-κB and Keap1/Nrf2 signalling pathways. J Pharm Pharmacol 2023; 75:287-299. [PMID: 36617177 DOI: 10.1093/jpp/rgac084] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/15/2022] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To investigate the protective effect and underlying mechanism of Inula japonica (TEIJ) in the treatment of acute lung injury (ALI). METHODS Protective effects of TEIJ in the inflammation and oxidative stress were studied in lipopolysaccharide (LPS)-induced ALI mice. Meanwhile, Western blot and real-time qPCR were carried out to investigate the underlying mechanism of TEIJ for ALI as well as immunohistochemistry. KEY FINDINGS TEIJ significantly alleviated the course of ALI via suppressing the interstitial infiltrated inflammatory cells, the increase of inflammatory factors and the decrease of anti-oxidative factors. TEIJ inactivated the MAPK/NF-κB signalling pathway to suppress the transcription of its downstream target genes, such as TNF-α, IL-6, etc. Meanwhile, TEIJ activated the Keap1/Nrf2 signalling pathway to regulate expression levels of Nrf2 and its target proteins. The results of LC-QTOF-MS/MS indicated potential active constituents of I. japonica, terpenoids and flavonoids. Additionally, terpenoids and flavonoids synergistically alleviated LPS-induced ALI depending on MAPK/NF-κB and Keap1/Nrf2 signalling pathways. CONCLUSION I. japonica could be considered a potential agent to treat ALI via regulating the MAPK/NF-κB and Keap1/Nrf2 signalling pathways.
Collapse
Affiliation(s)
- Min Zhang
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Juan Zhang
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Qi-Meng Zhu
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wen-Yu Zhao
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xia Lv
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Jing Yi
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Mi-Jia Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Zhang J, Zhang M, Zhang WH, Zhu QM, Ning J, Huo XK, Xiao HT, Sun CP. Total terpenoids of Inula japonica activated the Nrf2 receptor to alleviate the inflammation and oxidative stress in LPS-induced acute lung injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154377. [PMID: 36116200 DOI: 10.1016/j.phymed.2022.154377] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/15/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Acute lung injury (ALI) is a life-threatening lung disease and characterized by pulmonary edema and atelectasis. Inula japonica Thunb. is a commonly used traditional Chinese medicine for the treatment of lung diseases. However, the potential effect and mechanism of total terpenoids of I. japonica (TTIJ) on ALI remain obscure. PURPOSE This study focused on the protective effect of TTIJ on lipopolysaccharide (LPS)-induced ALI in mice and its potential mechanism. STUDY DESIGN AND METHODS A mouse model of ALI was established by intratracheal instillation of LPS to investigate the protective effect of TTIJ. RNA-seq and bioinformatics were then performed to reveal the underlying mechanism. Finally, western blot and real-time qPCR were used to verify the effects of TTIJ on the inflammation and oxidative stress. RESULTS TTIJ notably attenuated LPS-induced histopathological changes of lung. The RNA-seq result suggested that the protective effect of TTIJ on LPS-induced ALI were associated with the Toll-like receptor 4 (TLR4) and nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling pathways. Pretreatment with TTIJ significantly reduced the inflammation and oxidative stress via regulating levels of pro-inflammatory and anti-oxidative cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), superoxide dismutase (SOD), and glutathione (GSH), in LPS-induced ALI mice. TTIJ treatment could suppress the cyclooxygenase-2 (COX-2) expression level and the phosphorylation of p65, p38, ERK, and JNK through the inactivation of the MAPK/NF-κB signaling pathway in a TLR4-independent manner. Meanwhile, TTIJ treatment upregulated expression levels of proteins involved in the Nrf2 signaling pathway, such as heme oxygenase-1 (HO-1), NAD(P)H: quinoneoxidoreductase-1 (NQO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and glutamate-cysteine ligase modifier subunit (GCLM), via activating the Nrf2 receptor, which was confirmed by the luciferase assay. CONCLUSION TTIJ could activate the Nrf2 receptor to alleviate the inflammatory response and oxidative stress in LPS-induced ALI mice, which suggested that TTIJ could serve as the potential agent in the treatment of ALI.
Collapse
Affiliation(s)
- Juan Zhang
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen, China; School of pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China; Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Min Zhang
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Wen-Hao Zhang
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Qi-Meng Zhu
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jing Ning
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Hai-Tao Xiao
- School of pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.
| | - Cheng-Peng Sun
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China.
| |
Collapse
|
14
|
Zhang J, Zhang M, Zhang WH, Zhu QM, Huo XK, Sun CP, Ma XC, Xiao HT. Total flavonoids of Inula japonica alleviated the inflammatory response and oxidative stress in LPS-induced acute lung injury via inhibiting the sEH activity: Insights from lipid metabolomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154380. [PMID: 36150346 DOI: 10.1016/j.phymed.2022.154380] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/04/2022] [Accepted: 08/02/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Acute lung injury (ALI) is a severe respiratory disease characterized by diffuse lung interstitial and respiratory distress and pulmonary edema with a mortality rate of 35%-40%. Inula japonica Thunb., known as "Xuan Fu Hua" in Chinese, is a traditional Chinese medicine Inulae Flos to use for relieving cough, eliminating expectorant, and preventing bacterial infections in the clinic, and possesses an anti-pulmonary fibrosis effect. However, the effect and action mechanism of I. japonica on ALI is still unclear. PURPOSE This study aimed to investigate the protective effect and underlying mechanism of total flavonoids of I. japonica (TFIJ) in the treatment of ALI. STUDY DESIGN AND METHODS A mouse ALI model was established through administration of LPS by the intratracheal instillation. Protective effects of TFIJ in the inflammation and oxidative stress were studied in LPS-induced ALI mice based on inflammatory and oxidative stress factors, including MDA, MPO, SOD, and TNF-α. Lipid metabolomics, bioinformatics, Western blot, quantitative real-time PCR, and immunohistochemistry were performed to reveal the potential mechanism of TFIJ in the treatment of ALI. RESULTS TFIJ significantly alleviated the interstitial infiltration of inflammatory cells and the collapse of the alveoli in LPS-induced ALI mice. Lipid metabolomics demonstrated that TFIJ could significantly affect the CYP2J/sEH-mediated arachidonic acid metabolism, such as 11,12-EET, 14,15-EET, 8,9-DHET, 11,12-DHET, and 14,15-DHET, revealing that sEH was the potential target of TFIJ, which was further supported by the recombinant sEH-mediated the substrate hydrolysis in vitro (IC50 = 1.18 μg/ml). Inhibition of sEH by TFIJ alleviated the inflammatory response and oxidative stress via the MAPK, NF-κB, and Nrf2 signaling pathways. CONCLUSION These results demonstrated that TFIJ could suppress the sEH activity to stabilize the level of EETs, allowing the alleviation of the pathological course of lung injury in LPS-treated mice, which suggested that TFIJ could serve as the potential agents in the treatment of ALI.
Collapse
Affiliation(s)
- Juan Zhang
- School of pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China; Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Min Zhang
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Wen-Hao Zhang
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Qi-Meng Zhu
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Xiao-Chi Ma
- Second Affiliated Hospital, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Hai-Tao Xiao
- School of pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
15
|
Sun CP, Zhou JJ, Yu ZL, Huo XK, Zhang J, Morisseau C, Hammock BD, Ma XC. Kurarinone alleviated Parkinson's disease via stabilization of epoxyeicosatrienoic acids in animal model. Proc Natl Acad Sci U S A 2022; 119:e2118818119. [PMID: 35217618 PMCID: PMC8892522 DOI: 10.1073/pnas.2118818119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders and is characterized by loss of dopaminergic neurons in the substantia nigra (SN), causing bradykinesia and rest tremors. Although the molecular mechanism of PD is still not fully understood, neuroinflammation has a key role in the damage of dopaminergic neurons. Herein, we found that kurarinone, a unique natural product from Sophora flavescens, alleviated the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced behavioral deficits and dopaminergic neurotoxicity, including the losses of neurotransmitters and tyrosine hydroxylase (TH)-positive cells (SN and striatum [STR]). Furthermore, kurarinone attenuated the MPTP-mediated neuroinflammation via suppressing the activation of microglia involved in the nuclear factor kappa B signaling pathway. The proteomics result of the solvent-induced protein precipitation and thermal proteome profiling suggest that the soluble epoxide hydrolase (sEH) enzyme, which is associated with the neuroinflammation of PD, is a promising target of kurarinone. This is supported by the increase of plasma epoxyeicosatrienoic acids (sEH substrates) and the decrease of dihydroxyeicosatrienoic acids (sEH products), and the results of in vitro inhibition kinetics, surface plasmon resonance, and cocrystallization of kurarinone with sEH revealed that this natural compound is an uncompetitive inhibitor. In addition, sEH knockout (KO) attenuated the progression of PD, and sEH KO plus kurarinone did not further reduce the protection of PD in MPTP-induced PD mice. These findings suggest that kurarinone could be a potential natural candidate for the treatment of PD, possibly through sEH inhibition.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- College of Pharmacy, The Second Affiliated Hospital, Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Jun-Jun Zhou
- College of Pharmacy, The Second Affiliated Hospital, Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Zhen-Long Yu
- College of Pharmacy, The Second Affiliated Hospital, Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xiao-Kui Huo
- College of Pharmacy, The Second Affiliated Hospital, Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Juan Zhang
- College of Pharmacy, The Second Affiliated Hospital, Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616
| | - Xiao-Chi Ma
- College of Pharmacy, The Second Affiliated Hospital, Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, People's Republic of China;
| |
Collapse
|
16
|
Bailly C, Vergoten G. Japonicone A and related dimeric sesquiterpene lactones: molecular targets and mechanisms of anticancer activity. Inflamm Res 2022; 71:267-276. [PMID: 35034149 DOI: 10.1007/s00011-021-01538-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE AND DESIGN Japonicone A (Jap-A) is a sesquiterpene lactone (SL) dimer isolated from the plant Inula japonica Thunb. and the leading compound in the japonicone series of SL dimers which comprises 25 members (Jap-A to Jap-Y). We have analyzed the anticancer properties of Jap-A and the associated molecular targets. METHODS All literature data on japonicones and related SL dimers, including inulanolide A (Inu-A) and lineariifolianoid A (Lin-A) have been analyzed. Molecular models of the compound/target interactions were constructed to support our analysis. RESULTS Inulae Flos (Xuan Fu Hua) is used in traditional medicine in China and Korea to treat inflammatory diseases. The plant contains diverse japonicones and structurally related SL dimers. The interactions of Jap-A with the two main proteins, the pro-inflammatory cytokine TNF-α and the ubiquitin ligase MDM2, are at the origin of the anti-inflammatory and anticancer effects. Molecular docking analyses suggest that Inu-A is better adapted than Lin-A and Jap-A to form stable complexes with both TNF-α and MDM2. Jap-A exhibits marked capacities to inhibit cancer cell proliferation and dissemination and to trigger apoptosis, both in vitro and in vivo in several tumor models in mice. Its analogue Inu-A is more potent, functioning as a dual inhibitor of the MDM2-NFAT1 pathway. CONCLUSION This review shed some new light on the molecular targets and potential therapeutic benefits of these SL dimers and should help the design of novel anticancer agents derived from these compounds.
Collapse
Affiliation(s)
| | - Gérard Vergoten
- Inserm, INFINITE-U1286, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, University of Lille, 3 rue du Professeur Laguesse, BP-83, 59006, Lille, France
| |
Collapse
|
17
|
Shen XB, Ding DL, Yu LZ, Ni JZ, Liu Y, Wang W, Liu LM, Nian SH. Total extract of Anemarrhenae Rhizoma attenuates bleomycin-induced pulmonary fibrosis in rats. Bioorg Chem 2021; 119:105546. [PMID: 34954573 DOI: 10.1016/j.bioorg.2021.105546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 11/23/2021] [Accepted: 12/05/2021] [Indexed: 11/02/2022]
Abstract
Pulmonary fibrosis is a progressive interstitial lung disease with poor prognosis. Anemarrhenae Rhizoma is a traditional Chinese herbal medicine and has been applied in clinical practice for a long history. Recently, components of Anemarrhenae Rhizoma were reported to possess anti-inflammatory and immunomodulatory features; however, the effect of them on pulmonary fibrosis remains unknown. In this study, we explored the therapeutic effect of total extract of Anemarrhenae Rhizoma (TEAR) on bleomycin-induced pulmonary fibrosis. Pulmonary fibrosis rat model was established by a single intratracheal instillation of bleomycin, three doses of TEAR were intragastrically administered for consecutive 28 days. Subsequent to sacrificing of rats, pulmonary fibrosis was observed in rats treated with bleomycin, but administration of TEAR attenuated lung fibrosis, as evidenced by the improved lung histopathological damage and decreased weight loss and lung index. Moreover, TEAR treatment inhibited the inflammatory response in lung fibrosis, which was shown by the reduced nitrogen oxide level and myeloperoxidase activity. Furthermore, TEAR modulated the redox balance in lung tissue by alleviated lipid peroxidation and enhanced enzymatic antioxidants activity. Meanwhile, TEAR protected the rats from fibrosis in a dose-dependent manner, and the anti-fibrotic activity of TEAR may be related to the modulation of TGF-β1/Smad signaling pathway. Collectively, TEAR alleviates bleomycin-induced pulmonary fibrosis, indicating perspectives for development of a potential agent for lung fibrosis therapy.
Collapse
Affiliation(s)
- Xue-Bin Shen
- School of Pharmacy, Wannan Medical College, Wuhu 241002, PR China
| | - Da-Li Ding
- School of Pharmacy, Wannan Medical College, Wuhu 241002, PR China
| | - Li-Zhen Yu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, PR China
| | - Jin-Zhong Ni
- School of Basic Medical Sciences, Wannan Medical College, Wuhu 241002, PR China
| | - Yao Liu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, PR China
| | - Wei Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, PR China
| | - Li-Min Liu
- School of Pharmacy, Anhui College of Traditional Chinese Medicine, Wuhu 241003, PR China.
| | - Si-Hui Nian
- School of Pharmacy, Wannan Medical College, Wuhu 241002, PR China; Institute of Modern Chinese Medicine, Wannan Medical College, Wuhu 241002, PR China.
| |
Collapse
|
18
|
Yu B, Jin XQ, Yu WY, Dong YY, Ying HZ, Yu CH. 1β-Hydroxyalantolactone from Inulae Flos alleviated the progression of pulmonary fibrosis via inhibiting JNK/FOXO1/NF-κB pathway. Int Immunopharmacol 2021; 101:108339. [PMID: 34758440 DOI: 10.1016/j.intimp.2021.108339] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/29/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2022]
Abstract
Inulae Flos was widely distributed throughout Europe, Africa, and Asia, and was commonly used as a folk medicine in clinic for treating various respiratory diseases, including cough, asthma, bronchitis, pulmonary fibrosis, and pneumonia. However, the ingredients responsible for the pharmacology effects of I. Flos and the underlying mechanisms remain unclear. In this study, the effects of 16 known sesquiterpene lactones and flavonoids from I. Flos on TGF-β1-induced fibroblast activation were assessed by phenotypic high-content screening. Among those sixteen compounds, 1β-hydroxy alantolactone (HAL), the main characteristic sesquiterpene lactone from I. Flos, exhibited remarkable inhibitory activity. The further studies showed that HAL significantly inhibited the proliferation and induced the apoptosis of human fibroblast cell lines HELF and MRC-5 in a concentration-dependent manner. It also reduced intracellular ROS production, suppressed the mRNA expressions of E-cad, TGF-β1, Smad3, Col I, α-SMA and TNF-α, and downregulated protein expressions of α-SMA and F-actin. Furthermore, HAL significantly reduced the levels of HA, LN, PC-III and IV-C in serum, TNF-α and IL-6 in BALF, and TGF-β1, HYP and Col I in lung tissues of bleomycin (BLM)-treated rats. HAL significantly downregulated the expressions of p-JNK, FOXO1, p-p65, α-SMA, p-smad3 and Col I but upregulated p-FOXO1, which could be reversed by JNK agonist anisomycin. These results demonstrated that HAL induced the apoptosis of lung fibroblast cells activated by TGF-β1 and improved BLM-induced lung fibrosis in rats via inhibiting JNK/FOXO1/NF-κB pathway.
Collapse
Affiliation(s)
- Bing Yu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiao-Qing Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wen-Ying Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Ying-Ying Dong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hua-Zhong Ying
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China; Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou 310018, China; Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China.
| |
Collapse
|
19
|
Sun CP, Chang YB, Wang C, Lv X, Zhou WY, Tian XG, Zhao WY, Ma XC. Bisfischoids A and B, dimeric ent-abietane-type diterpenoids with anti-inflammatory potential from Euphorbia fischeriana Steud. Bioorg Chem 2021; 116:105356. [PMID: 34560562 DOI: 10.1016/j.bioorg.2021.105356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/11/2021] [Indexed: 01/17/2023]
Abstract
Two undescribed ent-abietane-type diterpenoid dimers with nonacyclic backbone formed by intermolecular [4 + 2] cycloaddition into a spirocyclic skeleton, bisfischoids A (1) and B (2), along with a known one fischdiabietane A (3), were identified from Euphorbia fischeriana Steud. Their structures were elucidated by extensive spectroscopic analysis, ECD and NMR calculation combined with DP4+ probability analysis, as well as X-ray diffraction. The anti-inflammatory potential of dimers 1-3 were examined using their inhibitory effects on soluble epoxide hydrolase (sEH), which revealed that 1 and 2 exhibited promising activities with inhibition constant (Ki) of 3.20 and 1.95 μM, respectively. Further studies of molecular docking and molecular dynamics indicated that amino acid residue Tyr343 in the catalytic cavity of sEH was the key site for their inhibitory function.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Yi-Bo Chang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Chao Wang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xia Lv
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Wei-Yu Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiang-Ge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Wen-Yu Zhao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China.
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian 116044, China; Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
20
|
Sun CP, Zhang XY, Zhou JJ, Huo XK, Yu ZL, Morisseau C, Hammock BD, Ma XC. Inhibition of sEH via stabilizing the level of EETs alleviated Alzheimer's disease through GSK3β signaling pathway. Food Chem Toxicol 2021; 156:112516. [PMID: 34411643 DOI: 10.1016/j.fct.2021.112516] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/04/2021] [Accepted: 08/14/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by dementia. Inhibition of soluble epoxide hydrolase (sEH) regulates inflammation involving in central nervous system (CNS) diseases. However, the exactly mechanism of sEH in AD is still unclear. In this study, we evaluated the vital role of sEH in amyloid beta (Aβ)-induced AD mice, and revealed a possible molecular mechanism for inhibition of sEH in the treatment of AD. The results showed that the sEH expression and activity were remarkably increased in the hippocampus of Aβ-induced AD mice. Chemical inhibition of sEH by TPPU, a selective sEH inhibitor, alleviated spatial learning and memory deficits, and elevated levels of neurotransmitters in Aβ-induced AD mice. Furthermore, inhibition of sEH could ameliorate neuroinflammation, neuronal death, and oxidative stress via stabilizing the in vivo level of epoxyeicosatrienoic acids (EETs), especially 8,9-EET and 14,15-EET, further resulting in the anti-AD effect through the regulation of GSK3β-mediated NF-κB, p53, and Nrf2 signaling pathways. These findings revealed the underlying mechanism of sEH as a potential therapeutic target in treatment of AD.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xin-Yue Zhang
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Jun-Jun Zhou
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Zhen-Long Yu
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, United States.
| | - Xiao-Chi Ma
- The Second Affiliated Hospital, College of Pharmacy, Institute of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China.
| |
Collapse
|
21
|
Zhao WY, Yan JJ, Zhang M, Wang C, Feng L, Lv X, Huo XK, Sun CP, Chen LX, Ma XC. Natural soluble epoxide hydrolase inhibitors from Inula britanica and their potential interactions with soluble epoxide hydrolase: Insight from inhibition kinetics and molecular dynamics. Chem Biol Interact 2021; 345:109571. [PMID: 34217688 DOI: 10.1016/j.cbi.2021.109571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/27/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022]
Abstract
Soluble epoxide hydrolase (sEH) is a potential drug target to treat inflammation and neurodegenerative diseases. In this study, we found that the extract of Inula britanica exhibited significantly inhibitory effects against sEH, therefore, we investigated its phytochemical constituents to obtain seven new compounds together with sixteen known ones (1-20), including two pairs of novel enantiomers, (2S,3S)-britanicafanin A (1a), (2R,3R)-britanicafanin A (1b), (2R,3S)-britanicafanin B (2a), and (2S,3R)-britanicafanin B (2b), and three new lignans britanicafanins C-E (3-5). Their structures were determined by HRESIMS, 1D and 2D NMR, and electronic circular dichroism (ECD) spectra as well as quantum chemical computations. All the isolates were evaluated for their inhibitory effects against sEH, compounds 1-3, 5-7, 9, 10, 13, 14, and 17-20 showed significant inhibitory effects against sEH with IC50 values from 3.56 μM to 26.93 μM. The inhibition kinetics results indicated that compounds 9, 10, 13, and 19 were all uncompetitive inhibitors, and their inhibition constants (Ki) values were 7.11, 1.99, 4.06, and 8.78 μM, respectively. Their potential interactions were analyzed by molecular docking and molecular dynamics (MD), which suggested that amino acid residues Asp335 and Asn359, especially Gln384, played an important role in the inhibition of compounds 10 and 13 on sEH, and compounds 10 and 13 could be considered as the potential candidates for the development of sEH inhibitors.
Collapse
Affiliation(s)
- Wen-Yu Zhao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Juan-Juan Yan
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Min Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Chao Wang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Lei Feng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xia Lv
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.
| | - Li-Xia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China.
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
22
|
Zhao WY, Zhang XY, Zhou MR, Tian XG, Lv X, Zhang HL, Deng S, Zhang BJ, Sun CP, Ma XC. Natural soluble epoxide hydrolase inhibitors from Alisma orientale and their potential mechanism with soluble epoxide hydrolase. Int J Biol Macromol 2021; 183:811-817. [PMID: 33957203 DOI: 10.1016/j.ijbiomac.2021.04.187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022]
Abstract
Inhibition of soluble epoxide hydrolase (sEH) is considered to be an effective treatment for inflammation-related diseases, and small molecules origin from natural products show promising activity against sEH. Two undescribed protostanes, 3β-hydroxy-25-anhydro-alisol F (1) and 3β-hydroxy-alisol G (2) were isolated from Alisma orientale and identified as new sEH inhibitors with IC50 values of 10.06 and 30.45 μM, respectively. Potential lead compound 1 was determined as an uncompetitive inhibitor against sEH, which had a Ki value of 5.13 μM. In-depth molecular docking and molecular dynamics simulations revealed that amino acid residue Ser374 plays an important role in the inhibition of 1, which also provides an idea for the development of sEH inhibitors based on protostane-type triterpenoids.
Collapse
Affiliation(s)
- Wen-Yu Zhao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xin-Yue Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Mei-Rong Zhou
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiang-Ge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xia Lv
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Hou-Li Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Sa Deng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Bao-Jing Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
23
|
Huang KC, Li JC, Wang SM, Cheng CH, Yeh CH, Lin LS, Chiu HY, Chang CY, Chuu JJ. The effects of carbon monoxide releasing molecules on paraquat-induced pulmonary interstitial inflammation and fibrosis. Toxicology 2021; 456:152750. [PMID: 33737140 DOI: 10.1016/j.tox.2021.152750] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/10/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023]
Abstract
Paraquat, an herbicide used extensively worldwide, can cause severe toxicity in humans and animals, leading to irreversible, lethal lung fibrosis. The potential of CO-releasing molecules (CORMs), substances that release CO (Carbon monoxide) within animal tissues, for treating paraquat-induced ROS generation and inflammation is investigated here. Our results show that the fast CO releaser CORM-3 (4-20 μM) acts as a potential scavenger of free radicals and decreases fibrosis progression by inhibiting paraquat-induced overexpression of connective tissue growth factor and angiotensin II in MRC-5 cells. The slow CO releaser CORM-A1 (5 mg/kg) clearly decreased expression of the lung profibrogenic cytokines COX-2, TNF-α, and α-SMA and serum hydroxyproline, resulting in a lower mortality rate in paraquat-treated mice. Mice treated with higher-dose CORM-A1 (10 mg/kg) had relatively intact lung lobes and fewer fibrotic patches by gross observation, with less collagen deposition, mesangial matrix accumulation, and pulmonary fibrosis resulting from the mitigation of TGF-β overexpression. In conclusion, our data demonstrate for the first time that CORM-A1 alleviated the development of the fibrotic process and improved survival rate in mice exposed to PQ, would be an attractive therapeutic approach to attenuate the progression of pulmonary fibrosis following PQ exposure.
Collapse
Affiliation(s)
- Kuo-Ching Huang
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Hospital, Liouying, Tainan, Taiwan; Department of Environmental and Occupational Health, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Jui-Chen Li
- Pharmacy Department, Wei-Gong Memorial Hospital, Miaoli, Taiwan
| | - Shu-Mei Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei, Taiwan
| | - Chia-Hui Cheng
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chun-Hsiang Yeh
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Li-Syun Lin
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hsin-Yi Chiu
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chia-Yu Chang
- Department of Neurology, Chi-Mei Medical Center, Tainan, Taiwan; Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| | - Jiunn-Jye Chuu
- Pharmacy Department, Wei-Gong Memorial Hospital, Miaoli, Taiwan; Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|