1
|
Lu D, Tian X, Cao T, Chen S, Liu C, Zheng L, Zhou M, Peng X, Li Y, Liu T. Emodin mitigates rheumatoid arthritis through direct binding to TNF-α. Front Pharmacol 2025; 16:1520281. [PMID: 40078278 PMCID: PMC11896996 DOI: 10.3389/fphar.2025.1520281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Emodin has shown certain anti-rheumatoid arthritis (RA) activity in preliminary studies. However, the precise mechanisms of emodin's anti-RA effects, particularly its direct targets, remain unclear. This study aimed to evaluate the anti-RA activity of emodin and elucidate its potential mechanisms, with a specific focus on identifying its molecular targets. Employing a collagen-induced arthritis (CIA) rat model, along with transcriptomic analysis, thermal proteome profiling (TPP) and TNF-α-induced L929 cell model, the anti-RA activity of emodin was confirmed, identifying TNF-α as a potential target. Techniques such as drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), Affinity ultrafiltration-liquid chromatography/mass spectrometry (AUF-LC/MS), surface plasmon resonance (SPR) and bio-layer interferometry (BLI) validated the direct binding of emodin to TNF-α. Molecular dynamics simulation, ELISA and BLI further revealed that emodin stabilizes the asymmetric trimeric structure of TNF-α, disrupting the TNF-α-TNFR1 interaction. In vitro assays, including luciferase reporter gene assay and TNF-α-induced MH7A cell model, demonstrated that this disruption inhibits TNF-α-induced NF-κB activation, leading to the downregulation of inflammatory mediators such as IL-6, IL-1β, and COX2. In conclusion, emodin directly targets TNF-α, stabilizing its structure and blocking TNF-α-TNFR1 interaction, which subsequently suppresses downstream NF-κB pathway activation and contributes to its potent anti-RA properties.
Collapse
Affiliation(s)
- Dingyan Lu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Xudong Tian
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Taotao Cao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Shuaishuai Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Chunhua Liu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Lin Zheng
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
- National Engineering Research Center of Miao’s Medicines, Guizhou Medical University, Guiyang, China
| | - Meng Zhou
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Xiaoyan Peng
- School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, China
| | - Yongjun Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
- National Engineering Research Center of Miao’s Medicines, Guizhou Medical University, Guiyang, China
| | - Ting Liu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
- National Engineering Research Center of Miao’s Medicines, Guizhou Medical University, Guiyang, China
| |
Collapse
|
2
|
Memari E, Helfield B. Shear stress preconditioning and microbubble flow pattern modulate ultrasound-assisted plasma membrane permeabilization. Mater Today Bio 2024; 27:101128. [PMID: 38988819 PMCID: PMC11234154 DOI: 10.1016/j.mtbio.2024.101128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
The recent and exciting success of anti-inflammatory therapies for ischemic heart disease (e.g. atherosclerosis) is hindered by the lack of site-specific and targeted therapeutic deposition. Microbubble-mediated focused ultrasound, which uses circulating, lipid-encapsulated intravascular microbubbles to locally enhance endothelial permeability, offers an exciting approach. Atherosclerotic plaques preferentially develop in regions with disturbed blood flow, and microbubble-endothelial cell membrane interactions under such flow conditions are not well understood. Here, using an acoustically-coupled microscopy system, endothelial cells were sonicated (1 MHz, 20 cycle bursts, 1 ms PRI, 4 s duration, 300 kPa peak-negative pressure) under perfusion with Definity™ bubbles to examine microbubble-mediated endothelial permeabilization under a range of physiological conditions. Endothelial preconditioning under prolonged shear influenced physiology and the secretome, inducing increased expression of pro-angiogenesis analytes, decreasing levels of pro-inflammatory ones, and increasing the susceptibility of ultrasound therapy. Ultrasound treatment efficiency was positively correlated with concentrations of pro-angiogenic cytokines (e.g. VEGF-A, EGF, FGF-2), and negatively correlated with pro-inflammatory chemokines (e.g. MCP-1, GCP-2, SDF-1). Furthermore, ultrasound therapy under non-reversing pulsatile flow (∼4-8 dyne/cm2, 0.5-1 Hz) increased permeabilization up to 2.4-fold compared to shear-matched laminar flow, yet treatment under reversing oscillatory flow resulted in more heterogeneous modulation. This study provides insight into the role of vascular physiology, including endothelial biology, into the design of a localized ultrasound drug delivery system for ischemic heart disease.
Collapse
Affiliation(s)
- Elahe Memari
- Department of Physics, Concordia University, Montreal, H4B 1R6, Canada
| | - Brandon Helfield
- Department of Physics, Concordia University, Montreal, H4B 1R6, Canada
- Department of Biology, Concordia University, Montreal, H4B 1R6, Canada
| |
Collapse
|
3
|
Alkafaas SS, Abdallah AM, Hassan MH, Hussien AM, Elkafas SS, Loutfy SA, Mikhail A, Murad OG, Elsalahaty MI, Hessien M, Elshazli RM, Alsaeed FA, Ahmed AE, Kamal HK, Hafez W, El-Saadony MT, El-Tarabily KA, Ghosh S. Molecular docking as a tool for the discovery of novel insight about the role of acid sphingomyelinase inhibitors in SARS- CoV-2 infectivity. BMC Public Health 2024; 24:395. [PMID: 38321448 PMCID: PMC10848368 DOI: 10.1186/s12889-024-17747-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Recently, COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, caused > 6 million deaths. Symptoms included respiratory strain and complications, leading to severe pneumonia. SARS-CoV-2 attaches to the ACE-2 receptor of the host cell membrane to enter. Targeting the SARS-CoV-2 entry may effectively inhibit infection. Acid sphingomyelinase (ASMase) is a lysosomal protein that catalyzes the conversion of sphingolipid (sphingomyelin) to ceramide. Ceramide molecules aggregate/assemble on the plasma membrane to form "platforms" that facilitate the viral intake into the cell. Impairing the ASMase activity will eventually disrupt viral entry into the cell. In this review, we identified the metabolism of sphingolipids, sphingolipids' role in cell signal transduction cascades, and viral infection mechanisms. Also, we outlined ASMase structure and underlying mechanisms inhibiting viral entry 40 with the aid of inhibitors of acid sphingomyelinase (FIASMAs). In silico molecular docking analyses of FIASMAs with inhibitors revealed that dilazep (S = - 12.58 kcal/mol), emetine (S = - 11.65 kcal/mol), pimozide (S = - 11.29 kcal/mol), carvedilol (S = - 11.28 kcal/mol), mebeverine (S = - 11.14 kcal/mol), cepharanthine (S = - 11.06 kcal/mol), hydroxyzin (S = - 10.96 kcal/mol), astemizole (S = - 10.81 kcal/mol), sertindole (S = - 10.55 kcal/mol), and bepridil (S = - 10.47 kcal/mol) have higher inhibition activity than the candidate drug amiodarone (S = - 10.43 kcal/mol), making them better options for inhibition.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Abanoub Mosaad Abdallah
- Narcotic Research Department, National Center for Social and Criminological Research (NCSCR), Giza, 11561, Egypt
| | - Mai H Hassan
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Aya Misbah Hussien
- Biotechnology department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Abanoub Mikhail
- Department of Physics, Faculty of Science, Minia University, Minia, Egypt
- Faculty of Physics, ITMO University, Saint Petersburg, Russia
| | - Omnia G Murad
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed I Elsalahaty
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University - Egypt, New Damietta, 34517, Egypt
| | - Fatimah A Alsaeed
- Department of Biology, College of Science, King Khalid University, Muhayl, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16Th Street, 35233, Khalifa City, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, 12622, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate, Egypt
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, 15551, United Arab Emirates
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa
- Natural & Medical Science Research Center, University of Nizwa, Nizwa, Oman
| |
Collapse
|
4
|
Cheng YW, Anzell AR, Morosky SA, Schwartze TA, Hinck CS, Hinck AP, Roman BL, Davidson LA. Shear Stress and Sub-Femtomolar Levels of Ligand Synergize to Activate ALK1 Signaling in Endothelial Cells. Cells 2024; 13:285. [PMID: 38334677 PMCID: PMC10854672 DOI: 10.3390/cells13030285] [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: 10/25/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Endothelial cells (ECs) respond to concurrent stimulation by biochemical factors and wall shear stress (SS) exerted by blood flow. Disruptions in flow-induced responses can result in remodeling issues and cardiovascular diseases, but the detailed mechanisms linking flow-mechanical cues and biochemical signaling remain unclear. Activin receptor-like kinase 1 (ALK1) integrates SS and ALK1-ligand cues in ECs; ALK1 mutations cause hereditary hemorrhagic telangiectasia (HHT), marked by arteriovenous malformation (AVM) development. However, the mechanistic underpinnings of ALK1 signaling modulation by fluid flow and the link to AVMs remain uncertain. We recorded EC responses under varying SS magnitudes and ALK1 ligand concentrations by assaying pSMAD1/5/9 nuclear localization using a custom multi-SS microfluidic device and a custom image analysis pipeline. We extended the previously reported synergy between SS and BMP9 to include BMP10 and BMP9/10. Moreover, we demonstrated that this synergy is effective even at extremely low SS magnitudes (0.4 dyn/cm2) and ALK1 ligand range (femtogram/mL). The synergistic response to ALK1 ligands and SS requires the kinase activity of ALK1. Moreover, ALK1's basal activity and response to minimal ligand levels depend on endocytosis, distinct from cell-cell junctions, cytoskeleton-mediated mechanosensing, or cholesterol-enriched microdomains. However, an in-depth analysis of ALK1 receptor trafficking's molecular mechanisms requires further investigation.
Collapse
Affiliation(s)
- Ya-Wen Cheng
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Anthony R. Anzell
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Stefanie A. Morosky
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tristin A. Schwartze
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cynthia S. Hinck
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrew P. Hinck
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Beth L. Roman
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lance A. Davidson
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
5
|
Zheng Z, Song X, Shi Y, Long X, Li J, Zhang M. Recent Advances in Biologically Active Ingredients from Natural Drugs for Sepsis Treatment. Comb Chem High Throughput Screen 2024; 27:688-700. [PMID: 37254548 DOI: 10.2174/1386207326666230529101918] [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: 07/25/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 06/01/2023]
Abstract
Sepsis refers to the dysregulated host response to infection; its incidence and mortality rates are high. It is a worldwide medical problem but there is no specific drug for it. In recent years, clinical and experimental studies have found that many monomer components of traditional Chinese medicine have certain effects on the treatment of sepsis. This paper reviews the advances in research on the active ingredients of traditional Chinese medicine involved in the treatment of sepsis in recent years according to their chemical structure; it could provide ideas and references for further research and development in Chinese materia medica for the treatment of sepsis.
Collapse
Affiliation(s)
- Zhenzhen Zheng
- Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Xiayinan Song
- Innovation Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yanmei Shi
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaofeng Long
- Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Jie Li
- Innovation Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Min Zhang
- Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| |
Collapse
|
6
|
Wang X, Yang S, Li Y, Jin X, Lu J, Wu M. Role of emodin in atherosclerosis and other cardiovascular diseases: Pharmacological effects, mechanisms, and potential therapeutic target as a phytochemical. Biomed Pharmacother 2023; 161:114539. [PMID: 36933375 DOI: 10.1016/j.biopha.2023.114539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/20/2023] Open
Abstract
The morbidity and mortality of cardiovascular diseases (CVDs) are increasing in recent years, and atherosclerosis (AS), a major CVD, becomes a disorder that afflicts human beings severely, especially the elders. AS is recognized as the primary cause and pathological basis of some other CVDs. The active constituents of Chinese herbal medicines have garnered increasing interest in recent researches owing to their influence on AS and other CVDs. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is a naturally occurring anthraquinone derivative found in some Chinese herbal medicines such as Rhei radix et rhizome, Polygoni cuspidati rhizoma et radix and Polygoni multiflori root. In this paper, we first review the latest researches about emodin's pharmacology, metabolism and toxicity. Meanwhile, it has been shown to be effective in treating CVDs caused by AS in dozens of previous studies. Therefore, we systematically reviewed the mechanisms by which emodin treats AS. In summary, these mechanisms include anti-inflammatory activity, lipid metabolism regulation, anti-oxidative stress, anti-apoptosis and vascular protection. The mechanisms of emodin in other CVDs are also discussed, such as vasodilation, inhibition of myocardial fibrosis, inhibition of cardiac valve calcification and antiviral properties. We have further summarized the potential clinical applications of emodin. Through this review, we hope to provide guidance for clinical and preclinical drug development.
Collapse
Affiliation(s)
- Xinyue Wang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujuan Li
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Min Wu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Emodin Alleviates Lupus Nephritis in Rats by Regulating M1/M2 Macrophage Polarization. J CHEM-NY 2023. [DOI: 10.1155/2023/5224921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Lupus nephritis (LN) is one of the most common clinical manifestations of systemic lupus erythematosus (SLE), causing death and disability. The current research study explored whether there was any improvement effect on LN after emodin administration. Network pharmacology was used to screen the target genes of emodin for the treatment of LN. LPS and IL-4 were employed for RAW264.7 macrophage M1/M2 polarization induction, and 0.1% HgCl2 was used for the LN rat model’s establishment. Flow cytometry was performed to detect the effect of 20, 40, and 80 µM emodin on RAW264.7 macrophage polarization. HE and PAS staining were subsequently conducted to detect 70 mg/kg emodin action on renal injury in LN rats. The effect of emodin on the content of urinary proteins and dsDNA antibodies was also determined. The results indicated that peroxisome proliferators-activated receptors gamma (PPARG) may be a target gene of emodin in LN, and emodin had no significant toxicity to macrophages at different concentrations. Compared with the control, emodin significantly inhibited LPS-induced polarization in M1 macrophages and improved that of IL-4-induced M2 macrophages. Besides, emodin alleviated kidney injury and markedly reduced the levels of urinary protein and dsDNA antibodies in rats. Moreover, after targeting interference with the PPARG expression, the improvement effect of emodin on LN is significantly reduced, indicating that emodin may relieve the symptoms of LN by activating the PPARG expression. Our study revealed that PPARG may be applied as a new therapy for LN.
Collapse
|
8
|
Gu L, Ju Y, Hu M, Zheng M, Li Q, Zhang X. Research progress of PPARγ regulation of cholesterol and inflammation in Alzheimer's disease. Metab Brain Dis 2023; 38:839-854. [PMID: 36723831 DOI: 10.1007/s11011-022-01139-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/29/2022] [Indexed: 02/02/2023]
Abstract
Peroxidase proliferator receptors (PPARs) are defined as key sensors and regulators of cell metabolism, transcription factors activated by ligands, involved in lipid, glucose and amino acid metabolism, participating in the processes of cell differentiation, apoptosis, inflammation regulation, and acute and chronic nerve damage. Among them, PPARγ is expressed in different brain regions and can regulate lipid metabolism, mitochondrial disorders, oxidative stress, and cell apoptosis. It has anti-inflammatory activity and shows neuroprotection. The regulation of Aβ levels in Alzheimer's disease involves cholesterol metabolism and inflammation, so this article first analyzes the biological functions of PPARγ, then mainly focuses on the relationship between cholesterol and inflammation and Aβ, and elaborates on the regulation of PPARγ on key proteins and the corresponding molecules, which provides new ideas for the treatment of AD.
Collapse
Affiliation(s)
- Lili Gu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Yue Ju
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Min Hu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Miao Zheng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Qin Li
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Xinyue Zhang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China.
| |
Collapse
|
9
|
Cao S, Xue J, Chen L, Hao Y, Lu M, Feng M, Wang H, Zhou J, Yao C. Effects of the Chinese herbal medicine Hong Huang decoction, on myocardial injury in breast cancer patients who underwent anthracycline-based chemotherapy. Front Cardiovasc Med 2022; 9:921753. [PMID: 35935647 PMCID: PMC9353583 DOI: 10.3389/fcvm.2022.921753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 06/24/2022] [Indexed: 12/24/2022] Open
Abstract
Objective To assess the effects of Hong Huang Decoction (HHD), a Chinese herbal medicine, on myocardial injury in breast cancer patients who underwent anthracycline (ANT)-based chemotherapy. Methods A total of 51 patients with breast cancer who underwent an ANT-based chemotherapy program and met the inclusion/exclusion criteria were allocated to the treatment or placebo groups using a random number generation process. Patients in the treatment group received liquid HHD twice a day. Treatment was given from 1 day prior to chemotherapy up to the end of chemotherapy (after 6 months). Participants in the placebo group received a placebo over the same schedule. Left ventricular ejection fraction (LVEF), global longitudinal strain (GLS), diagnostic markers of acute myocardial infarction [e.g., lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and B-type natriuretic peptide (BNP)], nitric oxide (NO), superoxide dismutase (SOD), as well as pro-inflammatory cytokines [e.g., tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and human C-reactive protein (CRP)], and anti-inflammatory cytokine interleukin-10 (IL-10), were outcome measures assessed before chemotherapy, 3 and 6 months after chemotherapy. Results Compared to the placebo group, the GLS value was significantly higher in the treatment group (19.95 ± 1.16 vs. 19.06 ± 1.64, P ≤ 0.001). Significant differences were also noted for levels of SOD (689.71 ± 203.60 vs. 807.88 ± 182.10, P < 0.05), IL-6 (58.04 ± 22.06 vs. 194.20 ± 40.14, P ≤ 0.001), IL-10 (237.90 ± 94.98 vs. 68.81 ± 32.92, P ≤ 0.001), NO (75.05 ± 26.39 vs. 55.83 ± 19.37, P ≤ 0.005), and TNF-α (301.80 ± 134.20 vs. 680.30 ± 199.60, P ≤ 0.001) in the patients before chemotherapy compared to 6 months after initiating chemotherapy. Conclusion HHD regulated the levels of IL-6, IL-10, SOD, NO, and TNF-α. The results demonstrated that GLS is a better indicator of early myocardial injury compared to LVEF, and HHD could modulate oxidative stress to protect against ANT cardio toxicity. Clinical trial registration Chinese Clinical Trial Registry, identifier ChiCTR1900022394. Date of registration: 2019-04-09.
Collapse
Affiliation(s)
- Sihan Cao
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingxian Xue
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Chen
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Hao
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meijuan Lu
- Department of Echocardiography, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Feng
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huanhuan Wang
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Zhou
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Yao
- Department of Breast Disease, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
10
|
Zhang S, Zhu N, Gu J, Li HF, Qiu Y, Liao DF, Qin L. Crosstalk between Lipid Rafts and Aging: New Frontiers for Delaying Aging. Aging Dis 2022; 13:1042-1055. [PMID: 35855333 PMCID: PMC9286918 DOI: 10.14336/ad.2022.0116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/16/2022] [Indexed: 12/15/2022] Open
Abstract
With the rapid aging in the global population, delay of aging has become a hot research topic. Lipid rafts (LRs) are microdomains in the plasma membrane that contain sphingolipids and cholesterol. Emerging evidence indicates an interesting interplay between LRs and aging. LRs and their components are altered with aging. Further, the aging process is strongly influenced by LRs. In recent years, LRs and their component signaling molecules have been recognized to affect aging by interfering with its hallmarks. Therefore, targeting LRs is a promising strategy to delay aging.
Collapse
Affiliation(s)
- Shuo Zhang
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Neng Zhu
- 2Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jia Gu
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Hong-Fang Li
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yun Qiu
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Li Qin
- 1Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China.,3Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
11
|
Recent Progress in Traditional Chinese Medicines and Their Mechanism in the Treatment of Allergic Rhinitis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3594210. [PMID: 35444784 PMCID: PMC9015857 DOI: 10.1155/2022/3594210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
Objective To conduct a systematic review on the mechanism of action and use of traditional Chinese medicines (TCM) in allergic rhinitis treatment. Background Allergic rhinitis (AR) is a type I allergic disease of the immune system induced by immunoglobulin E mediated inflammation and is characterized by sneezing, nasal itching, paroxysmal nasal obstruction, mucosal edema, cough, and rhinorrhea. More than 500 million people have been affected by rhinitis worldwide in the past 20 years, leading to negative effects on health, quality of life, and social relationships. Currently, the trending medicines used in the case of AR include intranasal corticosteroids and oral H1 antihistamines, which are given as combinatorial medicines supplemented with immune therapy. These medications have been found to be very effective in either the short term or long term; however, they have been found to possess some serious side effects. Search Methodology. The information in this article on classical and traditional Chinese medications used to treat AR was derived from original papers and reviews published in Chinese and English language journals. Two Chinese databases (Wanfang and CNKI) and three English databases (Cochrane Library, PubMed, and Embase) were utilized for data gathering. Results Traditional Chinese remedies have been identified to influence the production of cytokines such as IL-5 and IL-6, which are key mediators of eosinophilic inflammation, TNF-α, which stimulates TH2 cells at the site of inflammation, and NF-кB, which is required for cytokine and IgE antibody production. TCM has also been shown to be successful in lowering histamine levels, preserving histological changes by decreasing the thickness of the lamina propria, and downregulating the expression of Orai1, STIM1, and TRYC1, showing low expression of Ca+2 channel proteins. Conclusion In this review, we discussed a series of classical, traditional Chinese medications, including Centipeda minima, Scutellaria baicalensis, licorice root (Glycyrrhiza uralensis), and others, as potential antiallergic agents and investigate their in vivo effect upon the production of cytokines and release of histamines for allergic rhinitis treatment.
Collapse
|
12
|
Ruan L, Jiang L, Zhao W, Meng H, Zheng Q, Wang J. Hepatotoxicity or hepatoprotection of emodin? Two sides of the same coin by 1H-NMR metabolomics profiling. Toxicol Appl Pharmacol 2021; 431:115734. [PMID: 34606778 DOI: 10.1016/j.taap.2021.115734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
Emodin is the major anthraquinone component of many important traditional Chinese herbs, such as Rheum palmatum L. and Polygonum multiflorum Thunb. They have been popular health products but recently aroused concerns about their hepatotoxicity, which are believed to be arising from the contained anthraquinones, such as emodin. However, emodin exerts potent hepatoprotective ability, such as anti-fibrotic, anti-oxidative, and anti-inflammatory effects. In this study, 1H NMR based metabolomics approach, complemented with histopathological observation, biochemical measurements, western blotting analysis and real-time quantitative PCR (RT-qPCR), was applied to interpret the paradox of emodin (30 mg/kg, 10 mg/kg BW) using both healthy mice (male, ICR) and chronic CCl4-injured mice (0.1 mL/kg, 0.35% CCl4, 3 times a week for a month). Emodin exerted a weight loss property associated with its lipid-lowing effects, which helped alleviate CCl4-induced steatosis. Emodin effectively ameliorated CCl4-induced oxidative stress and energy metabolism dysfunction in mice liver via regulating glucose, lipid and amino acid metabolism, and inhibited excessive inflammatory response. In healthy mice, emodin only exhibited hepatoxicity on high-dosage by disturbing hepatic anti-oxidant homeostasis, especially GSH and xanthine metabolism. This integrated metabolomics approach identified the bidirectional potential of emodin, which are important for its rational use.
Collapse
Affiliation(s)
- Lingyu Ruan
- Center for Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Lei Jiang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Wenlong Zhao
- Center for Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Huihui Meng
- Center for Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Qi Zheng
- Center for Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Junsong Wang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| |
Collapse
|
13
|
Semwal RB, Semwal DK, Combrinck S, Viljoen A. Emodin - A natural anthraquinone derivative with diverse pharmacological activities. PHYTOCHEMISTRY 2021; 190:112854. [PMID: 34311280 DOI: 10.1016/j.phytochem.2021.112854] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/19/2021] [Accepted: 06/20/2021] [Indexed: 06/13/2023]
Abstract
Emodin (1,3,8-trihydroxy-6-methyl-anthraquinone) is a natural anthraquinone derivative that is present in numerous globally renowned herbal medicines. It is recognised as a protein tyrosine kinase inhibitor and as an anticancer drug, active against various tumour cells, including lung, breast, liver, and ovarian cancer cells. Recently, its role in combination chemotherapy with various allopathic medicines, to minimize their toxicity and to enhance their efficacy, has been studied. The use of emodin in these therapies is gaining popularity, due to fewer associated side effects compared with standard anticancer drugs. Emodin has a broad therapeutic window, and in addition to its antineoplastic activity, it displays anti-ulcer, anti-inflammatory, hepatoprotective, neuroprotective, antimicrobial, muscle relaxant, immunosuppressive and antifibrotic activities, in both in vitro and in vivo models. Although reviews on the anticancer activity of emodin have been published, none coherently unite all the pharmacological properties of emodin, particularly the anti-oxidant, antimicrobial, antidiabetic, immunosuppressive and hepatoprotective activities of the compound. Hence, in this review, all of the available data regarding the pharmacological properties of emodin are explored, with particular emphasis on the modes of action of the molecule. In addition, the manuscript details the occurrence, biosynthesis and chemical synthesis of the compound, as well as its toxic effects on biotic systems.
Collapse
Affiliation(s)
- Ruchi Badoni Semwal
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; Department of Chemistry, Pt. Lalit Mohan Sharma Govt. Post Graduate College, Rishikesh, 249201, India
| | - Deepak Kumar Semwal
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; Department of Phytochemistry, Faculty of Biomedical Sciences, Uttarakhand Ayurved University, Harrawala, Dehradun, 248001, India
| | - Sandra Combrinck
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - Alvaro Viljoen
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; SAMRC Herbal Drugs Research Unit, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa.
| |
Collapse
|
14
|
Xu J, Feng H, Ma L, Tan H, Yan S, Fang C. Bakkenolide‑IIIa ameliorates lipopolysaccharide‑induced inflammatory injury in human umbilical vein endothelial cells by upregulating LINC00294. Mol Med Rep 2021; 23:377. [PMID: 33760129 PMCID: PMC7986008 DOI: 10.3892/mmr.2021.12016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/04/2021] [Indexed: 01/21/2023] Open
Abstract
Inflammation, which causes injury to vascular endothelial cells, is one of the major factors associated with atherosclerosis (AS); therefore, inhibition of endothelial inflammation is a key step toward preventing AS. The present study aimed to investigate the effects of bakkenolide-IIIa (Bak-IIIa), an important active component of bakkenolides, on endothelial inflammation, as well as the mechanisms underlying such effects. Lipopolysaccharide (LPS)-damaged human umbilical vein endothelial cells (HUVECs) were treated with Bak-IIIa. The results of the MTT assay and enzyme-linked immunosorbent assay indicated that Bak-IIIa significantly alleviated survival inhibition, and decreased the levels of LPS-induced TNF-α, interleukin (IL)-1β, IL-8, and IL-6. Furthermore, long noncoding RNA (lncRNA) microarray analyses revealed 70 differentially expressed lncRNAs (DELs) in LPS-damaged HUVECs treated with Bak-IIIa. lncRNA target prediction results revealed that 44 DELs had 52 cis-targets, whereas 12 DELs covered 386 trans-targets. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses of the trans-targets indicated that three GO terms were associated with inflammation. Therefore, 17 targets involved in these GO terms and six relevant DELs were screened out. Validation via reverse transcription-quantitative PCR indicated that the fold change of NR_015451 (LINC00294) was the highest among the six candidates and that overexpression of LINC00294 significantly alleviated LPS-induced survival inhibition and inflammatory damage in HUVECs. In conclusion, Bak-IIIa ameliorated LPS-induced inflammatory damage in HUVECs by upregulating LINC00294. Thus, Bak-IIIa exhibited potential for preventing vascular inflammation.
Collapse
Affiliation(s)
- Jichong Xu
- Department of Interventional Radiology, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Hao Feng
- Department of Interventional Radiology, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Lin Ma
- Department of Interventional Radiology, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Huaqiao Tan
- Department of Interventional Radiology, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Shuo Yan
- Department of Interventional Radiology, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Chun Fang
- Department of Interventional Radiology, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| |
Collapse
|
15
|
Interaction of drugs with lipid raft membrane domains as a possible target. Drug Target Insights 2021; 14:34-47. [PMID: 33510571 PMCID: PMC7832984 DOI: 10.33393/dti.2020.2185] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/11/2020] [Indexed: 01/23/2023] Open
Abstract
Introduction Plasma membranes are not the homogeneous bilayers of uniformly distributed lipids but the lipid complex with laterally separated lipid raft membrane domains, which provide receptor, ion channel and enzyme proteins with a platform. The aim of this article is to review the mechanistic interaction of drugs with membrane lipid rafts and address the question whether drugs induce physicochemical changes in raft-constituting and raft-surrounding membranes. Methods Literature searches of PubMed/MEDLINE and Google Scholar databases from 2000 to 2020 were conducted to include articles published in English in internationally recognized journals. Collected articles were independently reviewed by title, abstract and text for relevance. Results The literature search indicated that pharmacologically diverse drugs interact with raft model membranes and cellular membrane lipid rafts. They could physicochemically modify functional protein-localizing membrane lipid rafts and the membranes surrounding such domains, affecting the raft organizational integrity with the resultant exhibition of pharmacological activity. Raft-acting drugs were characterized as ones to decrease membrane fluidity, induce liquid-ordered phase or order plasma membranes, leading to lipid raft formation; and ones to increase membrane fluidity, induce liquid-disordered phase or reduce phase transition temperature, leading to lipid raft disruption. Conclusion Targeting lipid raft membrane domains would open a new way for drug design and development. Since angiotensin-converting enzyme 2 receptors which are a cell-specific target of and responsible for the cellular entry of novel coronavirus are localized in lipid rafts, agents that specifically disrupt the relevant rafts may be a drug against coronavirus disease 2019.
Collapse
|
16
|
Emodin in atherosclerosis prevention: Pharmacological actions and therapeutic potential. Eur J Pharmacol 2020; 890:173617. [PMID: 33010303 DOI: 10.1016/j.ejphar.2020.173617] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/12/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerotic plaque formation, destabilization and eventual rupture leads to the acute cardiovascular events including myocardial infarction and stroke. Emodin (PubChem CID#3220), (1,3,8-trihydroxy-6-methylanthracene-9,10-dione) is a pharmacologically bioactive constituent isolated from the traditional Chinese medicinal herb Radix rhizoma Rhei. This molecule has anti-oxidant, anti-inflammatory, anti-proliferative, anti-apoptotic and lipid-modulating effects. Experimental studies have demonstrated that emodin attenuates and stabilizes atherosclerotic plaques. In this mini-review, we provide a summary of the pharmacological actions of emodin in regulating vascular function and atherosclerosis, highlighting the therapeutic potential of this phytochemical in patients with cardiovascular disease.
Collapse
|
17
|
Lee J, Kim HJ, Nguyen TTH, Kim SC, Ree J, Choi TG, Sohng JK, Park YI. Emodin 8-O-glucoside primes macrophages more strongly than emodin aglycone via activation of phagocytic activity and TLR-2/MAPK/NF-κB signalling pathway. Int Immunopharmacol 2020; 88:106936. [PMID: 32871479 DOI: 10.1016/j.intimp.2020.106936] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/03/2020] [Accepted: 08/23/2020] [Indexed: 01/09/2023]
Abstract
Emodin (Emo) is a natural plant anthraquinone derivative with a wide spectrum of pharmacological properties, including anticancer, antioxidant, and hepatoprotective activities. Glycosylation of natural anthraquinones with various sugar moieties can affect their physical, chemical, and biological functions. In this study, the potential immunomodulatory activities of Emo and its glycosylated derivative, emodin 8-O-glucoside (E8G), were evaluated and compared using murine macrophage RAW264.7 cells and human monocytic THP-1 cells. The results showed that E8G (20 μM) induced the secretion of TNF-α and IL-6 from RAW264.7 cells more effectively than unglycosylated Emo aglycone, by 4.9- and 1.6-fold, respectively, with no significant cytotoxicity in the concentration range tested (up to 20 μM). E8G (2.5-20 μM) significantly and dose-dependently induced inducible nitric oxide synthase (iNOS) expression by up to 3.2-fold compared to that of untreated control following a remarkable increase in nitric oxide (NO) production. E8G also significantly increased the expression of TLR-2 mRNA and the phosphorylation of MAPKs (JNK and p38). The activation and subsequent nuclear translocation of NF-κB was substantially enhanced upon treatment with E8G (2.5-20 μM). Moreover, E8G markedly induced macrophage-mediated phagocytosis of apoptotic Jurkat T cells. These results demonstrated that E8G far more strongly stimulates the secretion of proinflammatory cytokines, such as TNF-α and IL-6, and NO production from macrophages through upregulation of the TLR-2/MAPK/NF-κB signalling pathway than its nonglycosylated form, Emo aglycone. These results suggest for the first time that E8G may represent a novel immunomodulator, enhancing the early innate immunity.
Collapse
Affiliation(s)
- Jisun Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea
| | - Hyeon Jeong Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea
| | - Trang Thi Huyen Nguyen
- Department of Life Science and Biochemical Engineering, Sun Moon University, Chungnam 31460, Republic of Korea
| | - Seong Cheol Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea
| | - Jin Ree
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Kyung Sohng
- Department of Life Science and Biochemical Engineering, Sun Moon University, Chungnam 31460, Republic of Korea
| | - Yong Il Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea.
| |
Collapse
|
18
|
Tu YJ, Tan B, Jiang L, Wu ZH, Yu HJ, Li XQ, Yang AD. Emodin Inhibits Lipopolysaccharide-Induced Inflammation by Activating Autophagy in RAW 264.7 Cells. Chin J Integr Med 2020; 27:345-352. [PMID: 32840732 DOI: 10.1007/s11655-020-3477-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2019] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the effects of emodin on inflammation and autophagy in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages and reveal its underlying mechanism. METHODS 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay was conducted to find the appropriate dose for emodin. RAW264.7 cells pretreated with different concentrations (0-50 μmol/L) of emodin or vehicle for 2 h prior to exposure to LPS for 16 h. Cell morphology was examined and propidium iodide staining was used to examine cell cycle. Expressions of inflammation-related proteins [nuclear factor-kappaB (NF-κ B) and I-kappaB (I κ B)α] and autophagy-related proteins [light chain (LC)3, P62/sequestosome 1, mammalian target of rapamycin (mTOR), and p-mTOR] were examined using Western blot analysis. Expression of inflammation-related cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 were detected by enzyme-linked immunosorbent assay. Autophagy was examined with LC3B fluorescence intensity and aggregation. The effect of emodin on autophagy was conducted with an autophagy inhibitor, 3-methyladenine (3-MA). RESULTS The expression of NF-κ B in LPS-induced cells was significantly increased (P<0.01) and simultaneously I κ B α decreased compared with the normal cell (P<0.05). The expressions of TNF-α, IL-β, and IL-6 proteins in the LPS-induced RAW264.7 cells were significantly higher than in the normal cell (P<0.05 or P<0.01). LPS increased the percentage of cells in the G0/G1 phase, which was recovered by emodin at different doses (12.5, 25, and 50μ mol/L, P<0.05 or P<0.01). The medium-dose (25 μ ml/L) emodin decreased the expressions of NF-κ B, P62 and p-mTOR (P<0.01) and increased I κ B α expression, LC3B II/I ratio as well as LC3B fluorescence intensity (P<0.05 or P<0.01). Meanwhile, the enhanced autophagic effects of emodin, such as the increment of LC3B II/ratio and the decrement of P62 expression, were suppressed by autophagy inhibitor 3-MA. CONCLUSION Emodin could inhibit inflammation of mice RAW264.7 macrophages induced by LPS, possibly through activating autophagy.
Collapse
Affiliation(s)
- Yan-Jie Tu
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lei Jiang
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhong-Hua Wu
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong-Ji Yu
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Qian Li
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ai-Dong Yang
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
19
|
Neuroprotective Effects of Emodin against Ischemia/Reperfusion Injury through Activating ERK-1/2 Signaling Pathway. Int J Mol Sci 2020; 21:ijms21082899. [PMID: 32326191 PMCID: PMC7215870 DOI: 10.3390/ijms21082899] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/11/2020] [Accepted: 04/16/2020] [Indexed: 01/28/2023] Open
Abstract
Background: Stroke is one of the leading causes of death and disability worldwide and places a heavy burden on the economy in our society. Current treatments, such as the use of thrombolytic agents, are often limited by a narrow therapeutic time window. However, the regeneration of the brain after damage is still active days, even weeks, after stroke occurs, which might provide a second window for treatment. Emodin, a traditional Chinese medicinal herb widely used to treat acute hepatitis, has been reported to possess antioxidative capabilities and protective effects against myocardial ischemia/reperfusion injury. However, the underlying mechanisms and neuroprotective functions of Emodin in a rat middle cerebral artery occlusion (MCAO) model of ischemic stroke remain unknown. This study investigates neuroprotective effects of Emodin in ischemia both in vitro and in vivo. Methods: PC12 cells were exposed to oxygen-glucose deprivation to simulate hypoxic injury, and the involved signaling pathways and results of Emodin treatment were evaluated. The therapeutic effects of Emodin in ischemia animals were further investigated. Results: Emodin reduced infarct volume and cell death following focal cerebral ischemia injury. Emodin treatment restored PC12 cell viability and reduced reactive oxygen species (ROS) production and glutamate release under conditions of ischemia/hypoxia. Emodin increased Bcl-2 and glutamate transporter-1 (GLT-l) expression but suppressed activated-caspase 3 levels through activating the extracellular signal-regulated kinase (ERK)-1/2 signaling pathway. Conclusion: Emodin induced Bcl-2 and GLT-1 expression to inhibit neuronal apoptosis and ROS generation while reducing glutamate toxicity via the ERK-1/2 signaling pathway. Furthermore, Emodin alleviated nerve cell injury following ischemia/reperfusion in a rat MCAO model. Emodin has neuroprotective effects against ischemia/reperfusion injury both in vitro and in vivo, which may be through activating the ERK-1/2 signaling pathway.
Collapse
|
20
|
Paudel P, Seong SH, Fauzi FM, Bender A, Jung HA, Choi JS. Establishing GPCR Targets of hMAO Active Anthraquinones from Cassia obtusifolia Linn Seeds Using In Silico and In Vitro Methods. ACS OMEGA 2020; 5:7705-7715. [PMID: 32280914 PMCID: PMC7144155 DOI: 10.1021/acsomega.0c00684] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 03/16/2020] [Indexed: 05/08/2023]
Abstract
The present study examines the effect of human monoamine oxidase active anthraquinones emodin, alaternin (=7-hydroxyemodin), aloe-emodin, and questin from Cassia obtusifolia Linn seeds in modulating human dopamine (hD1R, hD3R, and hD4R), serotonin (h5-HT1AR), and vasopressin (hV1AR) receptors that were predicted as prime targets from proteocheminformatics modeling via in vitro cell-based functional assays, and explores the possible mechanisms of action via in silico modeling. Emodin and alaternin showed a concentration-dependent agonist effect on hD3R with EC50 values of 21.85 ± 2.66 and 56.85 ± 4.59 μM, respectively. On hV1AR, emodin and alaternin showed an antagonist effect with IC50 values of 10.25 ± 1.97 and 11.51 ± 1.08 μM, respectively. Interestingly, questin and aloe-emodin did not have any observable effect on hV1AR. Only alaternin was effective in antagonizing h5-HT1AR (IC50: 84.23 ± 4.12 μM). In silico studies revealed that a hydroxyl group at C1, C3, and C8 and a methyl group at C6 of anthraquinone structure are essential for hD3R agonist and hV1AR antagonist effects, as well as for the H-bond interaction of 1-OH group with Ser192 at a proximity of 2.0 Å. Thus, based on in silico and in vitro results, hV1AR, hD3R, and h5-HT1AR appear to be prime targets of the tested anthraquinones.
Collapse
Affiliation(s)
- Pradeep Paudel
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
| | - Su Hui Seong
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
| | - Fazlin Mohd Fauzi
- Department
of Pharmacology and Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Andreas Bender
- Center
for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2
1EW Cambridge, United Kingdom
| | - Hyun Ah Jung
- Department
of Food Science and Human Nutrition, Jeonbuk
National University, Jeonju 54896, Republic of Korea
- . Tel: 82-63-270-4882. Fax: 82-63-270-3854
| | - Jae Sue Choi
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
- . Tel: +82-51-629-5845. Fax: +82-51-629 5842
| |
Collapse
|
21
|
Mei J, Kong H, Zhao Z, Chen Z, Wang Y, Yang J. Shufengjiedu capsules protect against neuronal loss in olfactory epithelium and lung injury by enhancing autophagy in rats with allergic rhinitis. Biosci Trends 2019; 13:530-538. [PMID: 31866616 DOI: 10.5582/bst.2019.01332] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Shufengjiedu capsules (SFJDCs), a traditional Chinese medicine, have been widely used as an antiviral, antibacterial, antitumor, and anti-inflammatory drug. However, the roles of SFJDCs in allergic rhinitis remain unclear. The purpose of this study was to investigate the effects of SFJDCs in olfaction and lung injury in rats with allergic rhinitis. An animal model of allergic rhinitis was created by intraperitoneal injection and intranasal administration of ovalbumin to rats. All rats were divided into seven groups: a model group, a low-dose SFJDC group, a medium-dose SFJDC group, a high-dose SFJDC group, a cetirizine group, and a control group. Hematoxylin and eosin (HE) staining was used to observe pathological changes in rat lung and olfactory epithelium (OE) tissue, and peripheral blood was collected and subjected to an enzyme-linked immunosorbent assay (ELISA) to detect IgE, tumor necrosis factor alpha (TNF-α), and IL-1ꞵ levels. Western blotting, immunohistochemistry staining, and immunofluorescence staining were performed to detect inflammatory cytokines and levels of the autophagy biomarker beclin1 and the apoptosis biomarker cleaeved-caspased3 in lung and OE tissue. ELISA indicated that SFJDCs significantly decreased IgE, TNF-α, and IL-1ꞵ levels in peripheral blood, the lungs, and OE tissue. In addition, Western blotting and staining indicated that SFJDCs repair lung injury, protect against neuronal apoptosis in OE, and rescue impaired autophagy in the lungs and OE tissue. In conclusion, results indicated that SFJDCs might protect against neuronal loss in the OE and lung injury by enhancing autophagy and decreasing apoptosis in rats with allergic rhinitis. Therefore, SFJDCs might serve as an alternative treatment for allergic rhinitis.
Collapse
Affiliation(s)
- Jinyu Mei
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hua Kong
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhentao Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ziyu Chen
- The Second Clinical College of Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Yatang Wang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jianming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
22
|
Song Y, Cui X, Zhao R, Hu L, Li Y, Liu C. Emodin protects against lipopolysaccharide-induced inflammatory injury in HaCaT cells through upregulation of miR-21. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2654-2661. [PMID: 31250665 DOI: 10.1080/21691401.2019.1629951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background/aim: Pressure ulcers are a disastrous health issue in which inflammation is involved. Emodin possesses biological properties in inflammation. Our study investigated functions of emodin in lipopolysaccharide (LPS)-treated HaCaT cells. Methods: LPS was used to induce cell inflammation. MTT and flow cytometry were applied for cell viability and apoptosis assays, respectively. Moreover, apoptotic proteins were detected by western blot. Similarly, inflammatory factors and signalling related proteins were also determined by western blot. Results: Emodin increased cell viability and diminished apoptosis in LPS-treated HaCaT cells. Moreover, cleaved-PARP, cleaved-caspase-3 and cleaved-caspase-9 were all downregulated by emodin. Furthermore, inflammatory factors IL-1β, IL-6, Cox-2 and iNOS were inhibited by emodin in LPS-treated cells. In addition, emodin decreased phosphorylation of p65 and IκBα and the level of PTEN while enhanced phosphorylation of PI3K and AKT. Importantly, emodin increased expression of miR-21 suppressed by LPS and miR-21 downregulation negated the protective functions of emodin. Conclusions: Emodin promoted cell growth presented by increasing viability and blocking apoptosis process with inflammation inhibition. The protective activity of emodin was mediated by miR-21 up-regulation.
Collapse
Affiliation(s)
- Yanping Song
- a Department of Health Management, Heze Medical College , Heze , China
| | - Xueling Cui
- b Department of Breast and Thyroid Surgery, Heze Municipal Hospital , Heze , China
| | - Ruilan Zhao
- c Department of General Medicine, Heze Municipal Hospital , Heze , China
| | - Lanying Hu
- d Department of Joint Surgery, Heze Municipal Hospital , Heze , China
| | - Yanjun Li
- e e Department of Nursing, Heze Medical College , Heze , China
| | - Cuiling Liu
- b Department of Breast and Thyroid Surgery, Heze Municipal Hospital , Heze , China
| |
Collapse
|
23
|
Woller SA, Choi SH, An EJ, Low H, Schneider DA, Ramachandran R, Kim J, Bae YS, Sviridov D, Corr M, Yaksh TL, Miller YI. Inhibition of Neuroinflammation by AIBP: Spinal Effects upon Facilitated Pain States. Cell Rep 2019; 23:2667-2677. [PMID: 29847797 DOI: 10.1016/j.celrep.2018.04.110] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 04/02/2018] [Accepted: 04/25/2018] [Indexed: 12/17/2022] Open
Abstract
Apolipoprotein A-I binding protein (AIBP) reduces lipid raft abundance by augmenting the removal of excess cholesterol from the plasma membrane. Here, we report that AIBP prevents and reverses processes associated with neuroinflammatory-mediated spinal nociceptive processing. The mechanism involves AIBP binding to Toll-like receptor-4 (TLR4) and increased binding of AIBP to activated microglia, which mediates selective regulation of lipid rafts in inflammatory cells. AIBP-mediated lipid raft reductions downregulate LPS-induced TLR4 dimerization, inflammatory signaling, and expression of cytokines in microglia. In mice, intrathecal injections of AIBP reduce spinal myeloid cell lipid rafts, TLR4 dimerization, neuroinflammation, and glial activation. Intrathecal AIBP reverses established allodynia in mice in which pain states were induced by the chemotherapeutic cisplatin, intraplantar formalin, or intrathecal LPS, all of which are pro-nociceptive interventions known to be regulated by TLR4 signaling. These findings demonstrate a mechanism by which AIBP regulates neuroinflammation and suggest the therapeutic potential of AIBP in treating preexisting pain states.
Collapse
Affiliation(s)
- Sarah A Woller
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Eun Jung An
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Hann Low
- Department of Lipoproteins and Atherosclerosis, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Dina A Schneider
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Roshni Ramachandran
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Jungsu Kim
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Dmitri Sviridov
- Department of Lipoproteins and Atherosclerosis, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Kim YG, Park YH, Yang EY, Park WS, Park KS. Inhibition of tamoxifen's therapeutic effects by emodin in estrogen receptor-positive breast cancer cell lines. Ann Surg Treat Res 2019; 97:230-238. [PMID: 31742207 PMCID: PMC6848006 DOI: 10.4174/astr.2019.97.5.230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/08/2019] [Accepted: 10/13/2019] [Indexed: 01/22/2023] Open
Abstract
Purpose This study was aimed to investigate the combination effect of endoxifen and emodin on estrogen receptor (ER) positive breast cancer cell lines and to explain the mechanism of the combination effect. Methods We conducted this study on MCF-7 (ER+/human epidermal growth factor receptor-2 [HER2]−), T47D (ER+/HER2−), ZR-75-1 (ER+/HER2+), and BT474 (ER+/HER2+) cell lines, which confirmed combination effect of endoxifen and emodin. Optimal concentrations for combination were determined to study the effects on proliferation of MCF-7 and ZR-75-1 cells. Analysis of the combination effect was carried out in the CompuSyn software. The combination of downstream mechanisms, and combined effects of other similar compounds were tested on the MCF-7 and ZR 75-1 cell lines. Protein expression was confirmed by western blot. Results The combination of endoxifen and emodin had antagonistic effects on MCF-7 and ZR-75-1cell lines (combination index > 1). We validated the antagonistic effect in T47D and BT474 cell lines. During the combined treatment, the results showed elevated amounts of cyclin D1 and phosphorylated extracellular signal-regulated kinase (pERK). Analysis of drug interactions showed antagonistic effect between endoxifen and chemical compounds similar to emodin, such as chrysophanol or rhein, in MCF-7 and ZR-75-1 cells. Conclusion Addition of emodin attenuated tamoxifen's treatment effect via cyclin D1 and pERK up-regulation in ER-positive breast cancer cell lines.
Collapse
Affiliation(s)
- Yun Gyoung Kim
- Department of Surgery, Bundang Jesang General Hospital, Seongnam, Korea.,Clinical Science, Department of Medicine, The Graduate School of Konkuk University, Seoul, Korea
| | - Yoon Hwa Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Eun Yoel Yang
- Ewha Womans University Mokdong Hospital/Cancer Center for Women, Breast and Thyroid Cancer Center, Seoul, Korea
| | - Won Seo Park
- Department of Surgery, Kyung Hee University School of Medicine, Seoul, Korea
| | - Kyoung Sik Park
- Department of Surgery, Konkuk University School of Medicine, Seoul, Korea.,Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Recent studies demonstrate an important role of the secreted apolipoprotein A-I binding protein (AIBP) in regulation of cholesterol efflux and lipid rafts. The article discusses these findings in the context of angiogenesis and inflammation. RECENT FINDINGS Lipid rafts are cholesterol-rich and sphingomyelin-rich membrane domains in which many receptor complexes assemble upon activation. AIBP mediates selective cholesterol efflux, in part via binding to toll-like receptor-4 (TLR4) in activated macrophages and microglia, and thus reverses lipid raft increases in activated cells. Recent articles report AIBP regulation of vascular endothelial growth factor receptor-2, Notch1 and TLR4 function. In zebrafish and mouse animal models, AIBP deficiency results in accelerated angiogenesis, increased inflammation and exacerbated atherosclerosis. Spinal delivery of recombinant AIBP reduces neuraxial inflammation and reverses persistent pain state in a mouse model of chemotherapy-induced polyneuropathy. Inhalation of recombinant AIBP reduces lipopolysaccharide-induced acute lung injury in mice. These findings are discussed in the perspective of AIBP's proposed other function, as an NAD(P)H hydrate epimerase, evolving into a regulator of cholesterol trafficking and lipid rafts. SUMMARY Novel findings of AIBP regulatory circuitry affecting lipid rafts and related cellular processes may provide new therapeutic avenues for angiogenic and inflammatory diseases.
Collapse
Affiliation(s)
- Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist, 6550 Fannin St, TX77030
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 407 E 61st St, New York, NY 10065
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| |
Collapse
|
26
|
Ghaffari S, Roshanravan N. Saffron; An updated review on biological properties with special focus on cardiovascular effects. Biomed Pharmacother 2019; 109:21-27. [PMID: 30391705 DOI: 10.1016/j.biopha.2018.10.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Saffron as a natural product has long been used to impede and treat different disorders including cardiovascular disease (CVDs). Stigma is the most principal part of saffron. Various compounds such as carotenoids and flavonoids are the essential components of saffron stigma. The health benefits of saffron have been shown in previous studies; however, there is a lack of comprehensive data on the mechanistic aspects of its cardiovascular-health properties. This current comprehensive review focuses on the medicinal applications of saffron, and then the new findings regarding its cardiovascular-health effects and various cellular and molecular mechanisms of action will be debated. METHODS The literature search of MEDLINE, Embase, PubMed, Google Scholar and Cochrane Library was performed for all comparative studies since 2000-2018 with the limitations of the English language. RESULTS The results provided new evidence about antioxidant, anti-inflammatory, anti- atherogenic, anti- apoptotic, anti- hypertensive, and hypolipidemic effects of saffron. Pharmacological effects of saffron are due to a number of ingredients contained within this spice, including safranal, crocetin and crocins. CONCLUSIONS Our study concludes that saffron with wide range of usefulness in medicine may be the potent candidate in the process of new drug production for the treatment of CVDs.
Collapse
Affiliation(s)
- Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Liu Z, Lang Y, Li L, Liang Z, Deng Y, Fang R, Meng Q. Effect of emodin on chondrocyte viability in an in vitro model of osteoarthritis. Exp Ther Med 2018; 16:5384-5389. [PMID: 30542499 DOI: 10.3892/etm.2018.6877] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/16/2018] [Indexed: 02/06/2023] Open
Abstract
Emodin is an anthraquinone isolated from the Chinese herb Radix et Rhizoma Rhei and has been used to treat various diseases for centuries. The aim of the present study was to investigate the effect of emodin on the inflammatory mediators in rat chondrocytes. In the present study, chondrocytes were isolated from rats, cultured and harvested when they reached generation P3. Cells were treated with different doses of emodin (10, 20, and 30 µg/ml) followed by interleukin 1β (IL-1β, 10 ng/ml). Control cells were either untreated or treated with IL-1β alone. An enzyme-linked immunosorbent assay was used to measure levels of nitric oxide (NO) and prostaglandin E2 (PGE2). Reverse transcription-quantitative polymerase chain was performed to measure levels of matrix metallopeptidase (MMP)-3 and -13 mRNA. The expression of MMP-3, MMP-13, extracellular-signal regulatory kinase (ERK)1/2, phosphorylated ERK1/2 and β-catenin proteins were detected by western-blot analysis. The results demonstrated that treatment with emodin treatment reduced the cytotoxicity of IL-1β and inhibited the expression of NO and PGE2 in rat chondrocytes. Furthermore, emodin inhibited the expression of MMP3 and MMP13, and the phosphorylation of various proteins involved in the ERK and Wnt/β-catenin pathway. Therefore, emodin is able to promote the proliferation of chondrocytes by inhibiting the ERK and Wnt/β-catenin pathway and downregulating the expression of a series of inflammatory mediators in chondrocytes.
Collapse
Affiliation(s)
- Zhenfeng Liu
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| | - Yi Lang
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| | - Li Li
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| | - Zhiquan Liang
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| | - Yingjie Deng
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| | - Rui Fang
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| | - Qingcai Meng
- Department of Orthopedics, Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830000, P.R. China
| |
Collapse
|
28
|
Schink A, Neumann J, Leifke AL, Ziegler K, Fröhlich-Nowoisky J, Cremer C, Thines E, Weber B, Pöschl U, Schuppan D, Lucas K. Screening of herbal extracts for TLR2- and TLR4-dependent anti-inflammatory effects. PLoS One 2018; 13:e0203907. [PMID: 30307962 PMCID: PMC6181297 DOI: 10.1371/journal.pone.0203907] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/29/2018] [Indexed: 12/18/2022] Open
Abstract
Herbal extracts represent an ample source of natural compounds, with potential to be used in improving human health. There is a growing interest in using natural extracts as possible new treatment strategies for inflammatory diseases. We therefore aimed at identifying herbal extracts that affect inflammatory signaling pathways through toll-like receptors (TLRs), TLR2 and TLR4. Ninety-nine ethanolic extracts were screened in THP-1 monocytes and HeLa-TLR4 transfected reporter cells for their effects on stimulated TLR2 and TLR4 signaling pathways. The 28 identified anti-inflammatory extracts were tested in comparative assays of stimulated HEK-TLR2 and HEK-TLR4 transfected reporter cells to differentiate between direct TLR4 antagonistic effects and interference with downstream signaling cascades. Furthermore, the ten most effective anti-inflammatory extracts were tested on their ability to inhibit nuclear factor-κB (NF-κB) translocation in HeLa-TLR4 transfected reporter cell lines and for their ability to repolarize M1-type macrophages. Ethanolic extracts which showed the highest anti-inflammatory potential, up to a complete inhibition of pro-inflammatory cytokine production were Castanea sativa leaves, Cinchona pubescens bark, Cinnamomum verum bark, Salix alba bark, Rheum palmatum root, Alchemilla vulgaris plant, Humulus lupulus cones, Vaccinium myrtillus berries, Curcuma longa root and Arctostaphylos uva-ursi leaves. Moreover, all tested extracts mitigated not only TLR4, but also TLR2 signaling pathways. Seven of them additionally inhibited translocation of NF-κB into the nucleus. Two of the extracts showed impact on repolarization of pro-inflammatory M1-type to anti-inflammatory M2-type macrophages. Several promising anti-inflammatory herbal extracts were identified in this study, including extracts with previously unknown influence on key TLR signaling pathways and macrophage repolarization, serving as a basis for novel lead compound identification.
Collapse
Affiliation(s)
- Anne Schink
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
| | - Jan Neumann
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Anna Lena Leifke
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
| | - Kira Ziegler
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
| | | | - Christoph Cremer
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Eckhard Thines
- Institut für Biotechnologie und Wirkstoff-Forschung gGmbH, Kaiserslautern, Germany
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Bettina Weber
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
| | - Ulrich Pöschl
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University of Mainz Medical Center, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Kurt Lucas
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
- * E-mail:
| |
Collapse
|
29
|
Bruno K, Woller SA, Miller YI, Yaksh TL, Wallace M, Beaton G, Chakravarthy K. Targeting toll-like receptor-4 (TLR4)-an emerging therapeutic target for persistent pain states. Pain 2018; 159:1908-1915. [PMID: 29889119 PMCID: PMC7890571 DOI: 10.1097/j.pain.0000000000001306] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors that initiate signaling in innate and adaptive immune pathways. The highly conserved family of transmembrane proteins comprises an extracellular domain that recognizes exogenous and endogenous danger molecules and an ectodomain that activates downstream pathways in response. Recent studies suggest that continuous activation or dysregulation of TLR signaling may contribute to chronic disease states. The receptor is located not only on inflammatory cells (meningeal and peripheral macrophages) but on neuraxial glia (microglia and astrocytes), Schwann cells, fibroblasts, dorsal root ganglia, and dorsal horn neurons. Procedures blocking TLR functionality have shown pronounced effects on pain behavior otherwise observed in models of chronic inflammation and nerve injury. This review addresses the role of TLR4 as an emerging therapeutic target for the evolution of persistent pain and its role in noncanonical signaling, mediating anomalous pro-algesic actions of opiates. Accordingly, molecules targeting inhibition of this receptor have promise as disease-modifying and opioid-sparing alternatives for persistent pain states.
Collapse
Affiliation(s)
- Kelly Bruno
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Center for Excellence in Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Sarah A. Woller
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
| | - Yury I. Miller
- Department of Medicine, University of California San Diego Health Science, La Jolla, CA, USA
| | - Tony L. Yaksh
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| | - Mark Wallace
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| | - Graham Beaton
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| | - Krishnan Chakravarthy
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| |
Collapse
|
30
|
Choi SH, Wallace AM, Schneider DA, Burg E, Kim J, Alekseeva E, Ubags ND, Cool CD, Fang L, Suratt BT, Miller YI. AIBP augments cholesterol efflux from alveolar macrophages to surfactant and reduces acute lung inflammation. JCI Insight 2018; 3:120519. [PMID: 30135304 DOI: 10.1172/jci.insight.120519] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by an excessive pulmonary inflammatory response. Removal of excess cholesterol from the plasma membrane of inflammatory cells helps reduce their activation. The secreted apolipoprotein A-I binding protein (AIBP) has been shown to augment cholesterol efflux from endothelial cells to the plasma lipoprotein HDL. Here, we find that AIBP was expressed in inflammatory cells in the human lung and was secreted into the bronchoalveolar space in mice subjected to inhalation of LPS. AIBP bound surfactant protein B and increased cholesterol efflux from alveolar macrophages to calfactant, a therapeutic surfactant formulation. In vitro, AIBP in the presence of surfactant reduced LPS-induced p65, ERK1/2 and p38 phosphorylation, and IL-6 secretion by alveolar macrophages. In vivo, inhalation of AIBP significantly reduced LPS-induced airspace neutrophilia, alveolar capillary leak, and secretion of IL-6. These results suggest that, similar to HDL in plasma, surfactant serves as a cholesterol acceptor in the lung. Furthermore, lung injury increases pulmonary AIBP expression, which likely serves to promote cholesterol efflux to surfactant and reduce inflammation.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Aaron M Wallace
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | | | - Elianne Burg
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Jungsu Kim
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | - Niki Dj Ubags
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Carlyne D Cool
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, and.,Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist, Houston, Texas, USA
| | - Benjamin T Suratt
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Yury I Miller
- Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
31
|
Ding Y, Liu P, Chen ZL, Zhang SJ, Wang YQ, Cai X, Luo L, Zhou X, Zhao L. Emodin Attenuates Lipopolysaccharide-Induced Acute Liver Injury via Inhibiting the TLR4 Signaling Pathway in vitro and in vivo. Front Pharmacol 2018; 9:962. [PMID: 30186181 PMCID: PMC6113398 DOI: 10.3389/fphar.2018.00962] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/03/2018] [Indexed: 12/14/2022] Open
Abstract
Aims: Emodin is an anthraquinone with potential anti-inflammatory properties. However, the possible molecular mechanisms and protective effects of emodin are not clear. The objective of this study was to investigate the possible molecular mechanisms and protective effects of emodin on lipopolysaccharide (LPS)-induced acute liver injury (ALI) via the Toll-like receptor 4 (TLR4) signaling pathway in the Raw264.7 cell line and in Balb/c mice. Methods: This study established an inflammatory cellular model and induced an ALI animal model. TLR4 was overexpressed by lentivirus and downregulated by small interfering RNA (siRNA) technology. The mRNA and protein levels of TLR4 and downstream molecules were detected in cells and liver tissue. The tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 levels in supernatant and serum were determined by ELISA. The distribution and expression of mannose receptor C type 1 (CD206) and arginase 1 (ARG1) in the liver were tested by immunofluorescence. Mouse liver function and histopathological observations were assessed. Results: Administration of emodin reduced the protein and/or mRNA levels of TLR4 and its downstream molecules following LPS challenge in Raw264.7 cells and in an animal model. Additionally, emodin suppressed the expression of TNF-α and IL-6 in cell culture supernatant and serum. The inhibitory effect of emodin was also confirmed in RAW264.7 cells, in which TLR4 was overexpressed or knocked down. Additionally, ARG1 and CD206 were elevated in the emodin groups. Emodin also decreased serum ALT and AST levels and alleviated the liver histopathological damage induced by LPS. Conclusion: Emodin showed excellent hepatoprotective effects against LPS-induced ALI, possibly by inhibiting TLR4 signaling pathways.
Collapse
Affiliation(s)
- Yan Ding
- Department of Infectious Diseases and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Liu
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhi-Lin Chen
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Jun Zhang
- National and Local Joint Engineering Research Center for High-throughput Drug Screening Technology, Hubei Collaborative Innovation Center for Green Transformation of Bio-resources, Hubei University, Wuhan, China
| | - You-Qin Wang
- Graduate School of Jinzhou Medical University, Department of Pediatrics, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Xin Cai
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Lei Luo
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuan Zhou
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Song YD, Li XZ, Wu YX, Shen Y, Liu FF, Gao PP, Sun L, Qian F. Emodin alleviates alternatively activated macrophage and asthmatic airway inflammation in a murine asthma model. Acta Pharmacol Sin 2018; 39:1317-1325. [PMID: 29417945 DOI: 10.1038/aps.2017.147] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
Alternatively activated macrophages (AAMs) are not only associated with asthma but also lead to asthmatic airway inflammation and remodeling. Inhibition of AAMs is an alternative therapeutic strategy for treating asthma. In this study we investigated whether emodin (1,3,8-trihydroxy-6-methylanthraquinone), isolated from the rhizome of Rheum palmatum, alleviated asthmatic airway inflammation and reduced AAM polarization in a murine asthma model. Mice were sensitized with a triple allergen mix containing dust mite, ragweed and aspergillus (DRA). In mice with DRA-induced asthma, asthmatic inflammation was significantly enhanced. Intraperitoneal injection of emodin (20 mg·kg-1·d-1, ip) 1 h prior to DRA challenge on days 12-14 significantly decreased pulmonary eosinophil and lymphocyte infiltration, mucus secretion and serum IgE production, as well as IL-4 and IL-5 production in bronchoalveolar lavage fluid. In response to emodin treatment, activated markers of AAM Ym-1, Fizz-1 and arginase-1 in the lung tissues were remarkably decreased. In mouse bone marrow-derived macrophages (BMDMs) in vitro, emodin (2-50 μmol/L) dose-dependently inhibited IL-4-induced AAM polarization and STAT6 phosphorylation. Collectively, our results suggest that emodin effectively ameliorates asthmatic airway inflammation and AAM polarization, and it may therefore become a potential agent for the treatment of asthma.
Collapse
|
33
|
Zhao Y, Li N, Li Z, Zhang D, Chen L, Yao Z, Niu W. Conditioned medium from contracting skeletal muscle cells reverses insulin resistance and dysfunction of endothelial cells. Metabolism 2018; 82:36-46. [PMID: 29289515 DOI: 10.1016/j.metabol.2017.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/07/2017] [Accepted: 12/23/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Obese adipose tissue has been characterized with chronic inflammation associated with elevated secretion of inflammatory cytokines and declined secretion of anti-inflammatory cytokines which can impair endothelial function in an endocrine manner. Adipose tissue hypoxia plays a role in the changes of cytokines. Physical exercise/muscle contraction may help preventing cardiovascular disease through improving insulin resistance and endothelium function. However the mechanism is unclear. Skeletal muscle is an endocrine tissue. Contracting muscles secrete myokines which may play roles in the beneficial effect of exercise. In this study, the conditioned medium from electrical pulse stimulation (EPS) regulated skeletal muscle cells was used to explore the mechanism of contraction on endothelial dysfunction and insulin resistance induced by conditioned medium from hypoxic adipocytes. METHODS 3T3-L1 adipocytes were incubated under normoxia or hypoxia condition, respectively. The supernatant was collected as adipocyte conditioned medium (CM-N and CM-H). C2C12 mouse skeletal muscle cells were stimulated with EPS for 12 h. The supernatant was collected as muscle cells conditioned medium (CM-EPS). Human umbilical vein endothelial cells (HUVECs) were incubated with adipocyte CM and muscle cells CM together. Macrophages migration to HUVECs was detected with transwell system. The mRNA expressions of E-selectin, ICAM-1, MCP-1 and IL-6 were measured by real-time PCR. The phosphorylation of IKKα/β, NF-κB, Akt, AMPK, eNOS and SOCS3 protein levels were detected by Western blot. Concentration of NO was measured by ELISA kit. HUVECs apoptosis was detected by flow cytometry. RESULTS CM-EPS reduced the increase of mRNA expressions of E-selectin, ICAM-1, MCP-1 and IL-6 in HUVECs induced by CN-H. The phosphorylations of IKKα/β and NF-κB, SOCS3 protein level and endothelial cells apoptosis, which were raised by CM-H, were significantly reduced by CM-EPS. CM-EPS reversed the effects of CM-H on Akt and eNOS phosphorylations and NO production in HUVECs. CM-EPS directly stimulated the phosphorylation of AMPK, which caused the following phosphorylation of eNOS in HUVECs. CONCLUSION In summary, CM-EPS reversed endothelial cells inflammation, apoptosis, insulin resistance and dysfunction induced by CM-H.
Collapse
Affiliation(s)
- Yihe Zhao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Nana Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Zhu Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Da Zhang
- Department of Ultrasound, Tianjin Hospital, Tianjin 300211, China
| | - Liming Chen
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Zhi Yao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Wenyan Niu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
34
|
Li J, Li Y, Peng X, Li B, Yuan X, chen Y. Emodin attenuates titanium particle-induced osteolysis and RANKL-mediated osteoclastogenesis through the suppression of IKK phosphorylation. Mol Immunol 2018; 96:8-18. [PMID: 29455094 DOI: 10.1016/j.molimm.2018.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/07/2018] [Indexed: 12/15/2022]
|
35
|
Cheng J, Chen T, Li P, Wen J, Pang N, Zhang L, Wang L. Sodium tanshinone IIA sulfonate prevents lipopolysaccharide-induced inflammation via suppressing nuclear factor-κB signaling pathway in human umbilical vein endothelial cells. Can J Physiol Pharmacol 2017; 96:26-31. [PMID: 28658584 DOI: 10.1139/cjpp-2017-0023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, has been demonstrated to have potent anti-inflammatory properties. However, the protective effects of STS on lipopolysaccharide (LPS)-induced inflammation in endothelial cells remain to be elucidated. In the present study, human umbilical vein endothelial cells (HUVECs) were used to explore the effects of STS on LPS-induced inflammation and the molecular mechanism involved. HUVECs were pretreated with STS for 2 h, followed by stimulation with LPS. Then expression and secretion of tumor necrosis factor (TNF)-α and interleukin (IL)-1β, and the activation of nuclear factor-κB (NF-κB) were assessed. The results demonstrated that STS significantly decreased LPS-induced TNF-α and IL-1β protein expression in HUVECs. Similarly, the increased levels of TNF-α and IL-1β in cell supernatants stimulated by LPS were also significantly inhibited by STS. Furthermore, STS inhibited LPS-induced NF-κB p65 phosphorylation and nuclear translocation. All the results suggest that STS prevents LPS-induced inflammation through suppressing NF-κB signaling pathway in endothelial cells, indicating the potential utility of STS for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Jun Cheng
- a Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Tangting Chen
- a Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Pengyun Li
- a Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jing Wen
- a Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Ningbo Pang
- b Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, Sichuan, China.,c Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Liping Zhang
- b Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, Sichuan, China.,c Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Liqun Wang
- b Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, Sichuan, China.,c Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| |
Collapse
|
36
|
Rojas JM, Arán-Sekul T, Cortés E, Jaldín R, Ordenes K, Orrego PR, González J, Araya JE, Catalán A. Phospholipase D from Loxosceles laeta Spider Venom Induces IL-6, IL-8, CXCL1/GRO-α, and CCL2/MCP-1 Production in Human Skin Fibroblasts and Stimulates Monocytes Migration. Toxins (Basel) 2017; 9:toxins9040125. [PMID: 28379166 PMCID: PMC5408199 DOI: 10.3390/toxins9040125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/16/2017] [Accepted: 03/28/2017] [Indexed: 11/24/2022] Open
Abstract
Cutaneous loxoscelism envenomation by Loxosceles spiders is characterized by the development of a dermonecrotic lesion, strong inflammatory response, the production of pro-inflammatory mediators, and leukocyte migration to the bite site. The role of phospholipase D (PLD) from Loxosceles in the recruitment and migration of monocytes to the envenomation site has not yet been described. This study reports on the expression and production profiles of cytokines and chemokines in human skin fibroblasts treated with catalytically active and inactive recombinant PLDs from Loxosceles laeta (rLlPLD) and lipid inflammatory mediators ceramide 1-phosphate (C1P) and lysophosphatidic acid (LPA), and the evaluation of their roles in monocyte migration. Recombinant rLlPLD1 (active) and rLlPLD2 (inactive) isoforms induce interleukin (IL)-6, IL-8, CXCL1/GRO-α, and CCL2/monocyte chemoattractant protein-1 (MCP-1) expression and secretion in fibroblasts. Meanwhile, C1P and LPA only exhibited a minor effect on the expression and secretion of these cytokines and chemokines. Moreover, neutralization of both enzymes with anti-rLlPLD1 antibodies completely inhibited the secretion of these cytokines and chemokines. Importantly, conditioned media from fibroblasts, treated with rLlPLDs, stimulated the transmigration of THP-1 monocytes. Our data demonstrate the direct role of PLDs in chemotactic mediator synthesis for monocytes in human skin fibroblasts and indicate that inflammatory processes play an important role during loxoscelism.
Collapse
Affiliation(s)
- José M Rojas
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Tomás Arán-Sekul
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Emmanuel Cortés
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Romina Jaldín
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Kely Ordenes
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Patricio R Orrego
- Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Jorge González
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Jorge E Araya
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| | - Alejandro Catalán
- Laboratorio de Parasitología Molecular, Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, CP 1270300, Chile.
| |
Collapse
|
37
|
Emodin induces apoptosis of concanavalin A-stimulated murine splenocytes. Food Sci Biotechnol 2016; 25:1693-1700. [PMID: 30263464 DOI: 10.1007/s10068-016-0260-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/09/2016] [Accepted: 08/22/2016] [Indexed: 01/08/2023] Open
Abstract
Emodin, one of the major compounds in the herb Reynoutria elliptica, is known to maintain immunosuppressive, anti-allergic, anti-cancer, and anti-inflammatory effects. In this study, we assessed the possibility of using emodin to induce apoptosis in stimulated immune cells in vitro. After treatment with emodin and concanavalin A (Con A), we observed DNA damage-induced apoptosis in splenocytes. Moreover, treatment with emodin and Con A increased the number of apoptotic splenocytes compared with untreated controls. Emodin also diminished the size of CD45R/B220+ cells, CD19+CD69+ cells, and cDC populations. These results indicate that emodin-induced apoptosis was involved in attenuating the immune activity promoted by DNA damage and in decreasing the number of CD45R/B220+ B cells and CD19+CD69+ activating B cells. This demonstration of emodin inducing apoptosis of Con A-stimulated immune cells indicates its potential utility as a therapy for diseases caused by abnormally activated immune cells.
Collapse
|
38
|
Wu J, Hu Y, Xiang L, Li S, Yuan Y, Chen X, Zhang Y, Huang W, Meng X, Wang P. San-Huang-Xie-Xin-Tang Constituents Exert Drug-Drug Interaction of Mutual Reinforcement at Both Pharmacodynamics and Pharmacokinetic Level: A Review. Front Pharmacol 2016; 7:448. [PMID: 27965575 PMCID: PMC5124576 DOI: 10.3389/fphar.2016.00448] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/09/2016] [Indexed: 12/29/2022] Open
Abstract
Inflammatory disorders underlie varieties of human diseases. San-Huang-Xie-xin-Tang (SHXXT), composed with Rhizoma Rhei (Rheum palmatum L.), Rhizoma Coptidis (Coptis chinensis Franch), and Radix Scutellaria (Scutellaria baicalensis Georgi), is a famous formula which has been widely used in the fight against inflammatory abnormalities. Mutual reinforcement is one of the basic theories of traditional Chinese medicine. Here this article reviewed and analyzed the recent research on (1) How the main constituents of SHXXT impact on inflammation-associated signaling pathway molecules. (2) The interaction between the main constituents and efflux pumps or intestinal transporters. The goal of this work was to, (1) Provide evidence to support the theory of mutual reinforcement. (2) Clarify the key targets of SHXXT and suggest which targets need further investigation. (3) Give advice for the clinical use of SHXXT to elevated the absorption of main constituents and eventually promote oral bioavailability. We search literatures in scientific databases with key words of “each main SHXXT constituent,” in combination with “each main inflammatory pathway target molecule” or each main intestinal transporter, respectively. We report the effect of five main constituents on target molecules which lies in three main inflammatory signaling pathways, we as well investigate the interaction between constituents and intestinal transporter. We conclude, (1) The synergistic effect of constituents at both levels confirm the mutual reinforcement theory of TCM as it is proven in this work. (2) The effect of main constituents on downstream targets in nuclear need more further investigation. (3) Drug elevating the absorption of rhein, berberine and baicalein can be employed to promote oral bioavailability of SHXXT.
Collapse
Affiliation(s)
- Jiasi Wu
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Yingfan Hu
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Li Xiang
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Sheng Li
- Chengdu Institute of Biology, Chinese Academy of Sciences Chengdu, China
| | - Yi Yuan
- Chengdu University of Traditional Chinese MedicineChengdu, China; Chengdu Institute of Biology, Chinese Academy of SciencesChengdu, China
| | | | - Yan Zhang
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Wenge Huang
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Xianli Meng
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Ping Wang
- Chengdu University of Traditional Chinese Medicine Chengdu, China
| |
Collapse
|
39
|
Pan Y, Liu B, Deng Z, Fan Y, Li J, Li H. Lipid Rafts Promote trans Fatty Acid-Induced Inflammation in Human Umbilical Vein Endothelial Cells. Lipids 2016; 52:27-35. [DOI: 10.1007/s11745-016-4213-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/03/2016] [Indexed: 01/08/2023]
|
40
|
Linghu K, Lin D, Yang H, Xu Y, Zhang Y, Tao L, Chen Y, Shen X. Ameliorating effects of 1,8-cineole on LPS-induced human umbilical vein endothelial cell injury by suppressing NF-κB signaling in vitro. Eur J Pharmacol 2016; 789:195-201. [PMID: 27455900 DOI: 10.1016/j.ejphar.2016.07.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/20/2016] [Accepted: 07/21/2016] [Indexed: 11/18/2022]
Abstract
1,8-Cineole (also known as eucalyptol) is a monoterpene that occurs naturally in many aromatic plants, 1,8-cineole has been reported to ameliorate dysfunction of endothelial cells. However, the mechanism of action of 1,8-cineole is incompletely understood. We investigated the protective effect of 1,8-cineole on lipopolysaccharide (LPS)-induced human umbilical vein endothelial cell (HUVEC) injury and the underlying mechanisms. HUVECs were preincubated with 1,8-cineole for 1.5h, then exposed to LPS for 12h. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase leakage assays showed 1,8-cineole reduced LPS-induced HUVEC injury significantly. Results from enzyme linked immunosorbent assays revealed that 1,8-cineole suppressed LPS-induced secretion of interleukin-6 and interleukin-8, and recovered nitric oxide to normal levels. 1,8-Cineole decreased phosphorylation of nuclear factor-kappa B (NF-κB) p65 and expression of inducible nitric oxide synthase, and simultaneously improved protein levels of endothelial nitric oxide synthase. Immunofluorescence confirmed 1,8-cineole moderates nuclear translocation of NF-κB. These results suggest that 1,8-cineole ameliorates HUVEC dysfunction significantly, and that this effect at least involves NF-κB suppression.
Collapse
Affiliation(s)
- Kegang Linghu
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Dan Lin
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Hong Yang
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Yini Xu
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Yanyan Zhang
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Ling Tao
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Yan Chen
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China; Department of Pharmacology of Materia Medica, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China
| | - Xiangchun Shen
- The Key Lab of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China; Department of Pharmacology of Materia Medica, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China.
| |
Collapse
|
41
|
Jung HA, Ali MY, Jung HJ, Jeong HO, Chung HY, Choi JS. Inhibitory activities of major anthraquinones and other constituents from Cassia obtusifolia against β-secretase and cholinesterases. JOURNAL OF ETHNOPHARMACOLOGY 2016; 191:152-160. [PMID: 27321278 DOI: 10.1016/j.jep.2016.06.037] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/26/2016] [Accepted: 06/13/2016] [Indexed: 05/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Semen Cassiae has been traditionally used as an herbal remedy for liver, eye, and acute inflammatory diseases. Recent pharmacological reports have indicated that Cassiae semen has neuroprotective effects, attributable to its anti-inflammatory actions, in ischemic stroke and Alzheimer's disease (AD) models. AIM OF THE STUDY The basic goal of this study was to evaluate the anti-AD activities of C. obtusifolia and its major constituents. Previously, the extract of C. obtusifolia seeds, was reported to have memory enhancing properties and anti-AD activity to ameliorate amyloid β-induced synaptic dysfunction. However, the responsible components of C. obtusifolia seeds in an AD are currently still unknown. In this study, we investigated the inhibitory effects of C. obtusifolia and its constituents against acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) enzyme activity. MATERIALS AND METHODS In vitro cholinesterase enzyme assays by using AChE, BChE, and BACE1 were performed. We also scrutinized the potentials of Cassiae semen active component as BACE1 inhibitors via enzyme kinetics and molecular docking simulation. RESULTS In vitro enzyme assays demonstrated that C. obtusifolia and its major constituents have promising inhibitory potential against AChE, BChE, and BACE1. All Cassiae semen constituents exhibited potent inhibitory activities against AChE and BACE1 with IC50 values of 6.29-109µg/mL and 0.94-190µg/mL, whereas alaternin, questin, and toralactone gentiobioside exhibited significant inhibitory activities against BChE with IC50 values of 113.10-137.74µg/mL. Kinetic study revealed that alaternin noncompetitively inhibited, whereas cassiaside and emodin showed mixed-type inhibition against BACE1. Furthermore, molecular docking simulation results demonstrated that hydroxyl group of alaternin and emodin tightly interacted with the active site residues of BACE1 and their relevant binding energies (-6.62 and -6.89kcal/mol), indicating a higher affinity and tighter binding capacity of these compounds for the active site of BACE1. CONCLUSION The findings of the present study suggest the potential of C. obtusifolia and its major constituents for use in the development of therapeutic or preventive agents for AD, especially through inhibition of AChE, BChE and BACE1 activities.
Collapse
Affiliation(s)
- Hyun Ah Jung
- Department of Food Science and Human Nutrition, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Md Yousof Ali
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Hee Jin Jung
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Hyong Oh Jeong
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Hae Young Chung
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 608-737, Republic of Korea.
| |
Collapse
|
42
|
Ali AAA, Lee YR, Chen TC, Chen CL, Lee CC, Shiau CY, Chiang CH, Huang HS. Novel Anthra[1,2-c][1,2,5]Thiadiazole-6,11-Diones as Promising Anticancer Lead Compounds: Biological Evaluation, Characterization & Molecular Targets Determination. PLoS One 2016; 11:e0154278. [PMID: 27100886 PMCID: PMC4839570 DOI: 10.1371/journal.pone.0154278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/10/2016] [Indexed: 12/29/2022] Open
Abstract
The novel compounds NSC745885 and NSC757963 developed at our laboratory were tested against a panel of 60 cancer cell lines at the National Cancer Institute, USA, and a panel of 39 cancer cell lines at the Japanese Foundation of Cancer Research. Both compounds demonstrated selective unique multi-log differential patterns of activity, with GI50 values in the sub-micro molar range against cancer cells rather than normal cardiac cells. NSC757963 showed high selectivity towards the leukemia subpanel. Activities of both compounds strongly correlated to expression of NFKB1 and CSNK2B genes, implying that they may inhibit the NF-κB pathway. Immunocytochemical microscopy of OVCAR-3 cells showed clear cytosolic accumulation of the NF-κB p65 subunit following treatment. Western blotting showed dose dependent inhibition of the nuclear expression of the NF-κB p65 subunit with subsequent accumulation in the cytosol following treatment. Docking experiments showed binding of both compounds to the NF-κB activator IKKβ subunit preventing its translocation to the nucleus. Collectively, these results confirm the ability of our compounds to inhibit the constitutively active NF-κB pathway of OVCAR-3 cells. Furthermore, COMPARE analysis indicated that the activity of NSC757963 is similar to the antituberculosis agent rifamycin SV, this was confirmed by testing the antimycobacterial activity of NSC757963 against Mycobacterium tuberculosis, results revealed potent activity suitable for use in clinical practice. Molecular properties and Lipinski’s parameters predicted acceptable bioavailability properties with no indication of mutagenicity, tumorigenicity, irritability and reproductive effects. Oral absorption experiments using the human Caco-2 model showed high intestinal absorption of NSC745885 by passive transport mechanism with no intestinal efflux or active transport mechanisms. The unique molecular characterization as well as the illustrated anticancer spectra of activity and bioavailability properties warrant further development of our compounds and present a foundation brick in the pre-clinical investigations to implement such compounds in clinical practice.
Collapse
Affiliation(s)
- Ahmed Atef Ahmed Ali
- Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ru Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Tsung-Chih Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chun-Liang Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Chung Lee
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Yang Shiau
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chiao-Hsi Chiang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsu-Shan Huang
- Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
43
|
Yang H, Guan L, Li S, Jiang Y, Xiong N, Li L, Wu C, Zeng H, Liu Y. Mechanosensitive caveolin-1 activation-induced PI3K/Akt/mTOR signaling pathway promotes breast cancer motility, invadopodia formation and metastasis in vivo. Oncotarget 2016; 7:16227-47. [PMID: 26919102 PMCID: PMC4941310 DOI: 10.18632/oncotarget.7583] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/05/2016] [Indexed: 01/27/2023] Open
Abstract
Cancer cells are subjected to fluid shear stress during passage through the venous and lymphatic system. Caveolin-1 (Cav-1), a principal structural component of caveolar membrane domains, contributes to cancer development but its mechanobiological roles under low shear stress (LSS) conditions remain largely unknown. Here, we identified Cav-1 is mechanosensitive to LSS exposure, and its activation-induced PI3K/Akt/mTOR signaling promotes motility, invadopodia formation and metastasis of breast carcinoma MDA-MB-231 cells. Application of LSS (1.8 and 4.0 dynes/cm2) to MDA-MB-231 cells significantly increased the cell motility, invadopodia formation, MT1-MMP expression, ECM degradation, and also induced a sustained activation of Cav-1 and PI3K/Akt/mTOR signaling cascades. Methyl-β-cyclodextrin-caused caveolae destruction markedly decreased LSS-induced activation of both Cav-1 and PI3K/Akt/mTOR, leading to suppress MT1-MMP expression, inhibit invadopodia formation and ECM degradation, suggesting that caveolae integrity also involved in metastasis. Immunocytochemical assay showed that LSS induces the Cav-1 clustering in lipid rafts and co-localization of Cav-1 and MT1-MMP on invadopodia. Immunofluorescence confocal analysis demonstrated that Cav-1 activation were required for the acquisition of a polarized phenotype in MDA-MB-231 cells. Finally, Cav-1 knockdown significantly suppressed tumor colonization in the lungs and distant metastases in animal models. Our findings highlight the importance of Cav-1 in hematogenous metastasis, and provide new insights into the underlying mechanisms of mechanotransduction induced by LSS.
Collapse
Affiliation(s)
- Hong Yang
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
- Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Liuyuan Guan
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Ying Jiang
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Niya Xiong
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Li Li
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
- Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Hongjuan Zeng
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
- Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
- Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, P.R. China
| |
Collapse
|
44
|
Qu W, Wang Y, Wu Q, Liu J, Hao D. Emodin inhibits HMGB1-induced tumor angiogenesis in human osteosarcoma by regulating SIRT1. Int J Clin Exp Med 2015; 8:15054-15064. [PMID: 26628989 PMCID: PMC4658878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 07/17/2015] [Indexed: 06/05/2023]
Abstract
UNLABELLED The anti-cancer effects of emodin, including inhibition of proliferation, invasion, metastasis and angiogenesis, were confirmed by various previous studies. However, the specific mechanisms were not clear. In this study, we investigated emodin's anti-angiogenesis effect and focused on the mechanisms in human osteosarcoma (OS). OS cells were implanted to nude mice to form OS xenografts. Immunofluorescence assay was used to assess vWF expression in tumor tissue. MTT assay was employed to screen proper emodin concentrations unrelated with proliferation inhibition. siRNA technique was utilized to silence SIRT1 expression in OS cells. Expression levels of SIRT1 and VEGF were investigated by real-time PCR and western blotting. H4-k16Ac expression which indicated the deacetylation activity of SIRT1 was also detected by western blotting. As in results, HMGB1 treatment exacerbated OS angiogenesis both in vivo and in vitro. Emodin administration attenuated angiogenesis in both OS and HMGB1 treated OS in vivo and in vitro. After emodin treatment, the expression level and deacetylation activity of SIRT1 were dramatically enhanced. HMGB1-induced angiogenesis was more striking in SIRT1 silenced OS cells. SIRT1 silencing also impaired the anti-angiogenesis effect of emodin in OS cells. IN CONCLUSION SIRT expression and deacetylation activity elevation are involved in emodin's anti-angiogenesis effect in human OS.
Collapse
Affiliation(s)
- Wei Qu
- Department of Spine Surgery, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine No 555, Friendship Rd, Xi'an 710054, China
| | - Yufei Wang
- Department of Spine Surgery, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine No 555, Friendship Rd, Xi'an 710054, China
| | - Qining Wu
- Department of Spine Surgery, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine No 555, Friendship Rd, Xi'an 710054, China
| | - Jijun Liu
- Department of Spine Surgery, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine No 555, Friendship Rd, Xi'an 710054, China
| | - Dingjun Hao
- Department of Spine Surgery, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine No 555, Friendship Rd, Xi'an 710054, China
| |
Collapse
|
45
|
Chen GL, Zhang JJ, Kao X, Wei LW, Liu ZY. Emodin ameliorates lipopolysaccharides-induced corneal inflammation in rats. Int J Ophthalmol 2015; 8:665-9. [PMID: 26308161 DOI: 10.3980/j.issn.2222-3959.2015.04.04] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 01/05/2015] [Indexed: 01/30/2023] Open
Abstract
AIM To investigate the effect of emodin on pseudomonas aeruginosa lipopolysaccharides (LPS)-induced corneal inflammation in rats. METHODS Corneal infection was induced by pseudomonas aeruginosa LPS in Wistar rats. The inflammation induced by LPS were examined by slit lamp microscope and cytological checkup of aqueous humor. Corneal tissue structure was observed by hematoxylin and eosin (HE) staining. The activation of nuclear factor kappaB (NF-κB) was determined by Western blot. Messenger ribonucleic acid (mRNA) of tumor necrosis factor-α (TNF-α) and intercellular adhesion molecule-1 (ICAM-1) in LPS-challenged rat corneas were measured with reverse transcription-polymerase chain reaction (RT-PCR). RESULTS Typical manifestations of acute corneal inflammation were observed in LPS-induce rat model, and the corneal inflammatory response and structure were improved in rats pretreated with emodin. Treatment with emodin could improve corneal structure, reduce corneal injure by reducing corneal inflammatory response. Emodin could inhibit the decreasing lever of inhibitor of kappaB alpha (IкBα) express, and the mRNA expression of TNF-α and ICAM-1 in corneal tissues was also inhibited by emodin. The differences were statistically significant between groups treated with emodin and those without treatment (P<0.01). CONCLUSION Emodin could ameliorate LPS-induced corneal inflammation, which might via inhibiting the activation of NF-κB.
Collapse
Affiliation(s)
- Guo-Ling Chen
- Department of Ophthalmology, the Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| | - Jing-Jing Zhang
- Department of Ophthalmology, the Third People's Hospital of Jinan, Jinan 250101, Shandong Province, China
| | - Xin Kao
- Department of Ophthalmology, the Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| | - Lu-Wan Wei
- Department of Anatomy, Shandong Univeristy School of Medicine, Jinan 250012, Shandong Province, China
| | - Zhi-Yu Liu
- Department of Anatomy, Shandong Univeristy School of Medicine, Jinan 250012, Shandong Province, China
| |
Collapse
|
46
|
Chen G, Zhang J, Zhang H, Xiao Y, Kao X, Liu Y, Liu Z. Anti-inflammatory effect of emodin on lipopolysaccharide-induced keratitis in Wistar rats. Int J Clin Exp Med 2015; 8:12382-12389. [PMID: 26550148 PMCID: PMC4612833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/03/2015] [Indexed: 06/05/2023]
Abstract
Emodin, a major bioactive extract of several Chinese herbs, has been shown to have a number of biological activities including antiviral, anti-inflammatory, anti-tumor, anti-fibrosis etc. In the present study, we investigated the effects of emodin as an anti-inflammatory agent on lipopolysaccharide (LPS) induced keratitis in Wistar rats. Clinical score, slit-lamp microscope were used to determine corneal inflammatory response. Corneal structure was observed by hematoxylin-eosin staining and transmission electron microscopy. Messenger ribonucleic acid levels of tight junction protein and cytokines were determined by reverse transcription- polymerase chain reaction. The activation of nuclear factor-kappa B (NF-κB) was detected with Western blot. We found that disorganized corneal tissue and cellular structures were observed in keratitis rats and emodin could deduce inflammatory response and improve corneal structure. Pretreated with emodin could up-regulate and down-regulate the mRNA expression of occludin and Interleukin-6. The activation of NF-κB could be inhibited partly after emodin treatment. In conclusion, emodin reduced corneal inflammation in LPS-induced keratitis in Wistar rats due to its capability of inhibition in NF-κB activation.
Collapse
Affiliation(s)
- Guoling Chen
- Department of Ophthalmology, The Second Hospital of Shandong UniversityJinan, China
| | - Jingjing Zhang
- Department of Ophthalmology, The Third People’s Hospital of JinanJinan, China
| | - Han Zhang
- Department of Ophthalmology, The Second Hospital of Shandong UniversityJinan, China
| | - Ying Xiao
- Department of Ophthalmology, The Second Hospital of Shandong UniversityJinan, China
| | - Xin Kao
- Department of Ophthalmology, The Second Hospital of Shandong UniversityJinan, China
| | - Yanli Liu
- Department of Anatomy, Shandong University School of MedicineJinan, China
| | - Zhiyu Liu
- Department of Anatomy, Shandong University School of MedicineJinan, China
| |
Collapse
|
47
|
Bounda GA, Feng YU. Review of clinical studies of Polygonum multiflorum Thunb. and its isolated bioactive compounds. Pharmacognosy Res 2015; 7:225-36. [PMID: 26130933 PMCID: PMC4471648 DOI: 10.4103/0974-8490.157957] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 02/18/2015] [Accepted: 06/02/2015] [Indexed: 01/05/2023] Open
Abstract
Polygonum multiflorum Thunb. (PMT), officially listed in the Chinese Pharmacopoeia, is one of the most popular perennial Chinese traditional medicines known as He shou wu in China and East Asia, and as Fo-ti in North America. Mounting pharmacological studies have stressed out its key benefice for the treatment of various diseases and medical conditions such as liver injury, cancer, diabetes, alopecia, atherosclerosis, and neurodegenerative diseases as well. International databases such as PubMed/Medline, Science citation Index and Google Scholar were searched for clinical studies recently published on P. multiflorum. Various clinical studies published articles were retrieved, providing information relevant to pharmacokinetics-pharmacodynamics analysis, sleep disorders, dyslipidemia treatment, and neurodegenerative diseases. This review is an effort to update the clinical picture of investigations ever carried on PMT and/or its isolated bio-compounds and to enlighten its therapeutic assessment.
Collapse
Affiliation(s)
- Guy-Armel Bounda
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - YU Feng
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, China
| |
Collapse
|
48
|
Nair AR, Ebenezer PJ, Saini Y, Francis J. Angiotensin II-induced hypertensive renal inflammation is mediated through HMGB1-TLR4 signaling in rat tubulo-epithelial cells. Exp Cell Res 2015; 335:238-47. [PMID: 26033363 DOI: 10.1016/j.yexcr.2015.05.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 04/27/2015] [Accepted: 05/13/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Angiotensin II is a vaso-constrictive peptide that regulates blood pressure homeostasis. Even though the inflammatory effects of AngII in renal pathophysiology have been studied, there still exists a paucity of data with regard to the mechanism of action of AngII-mediated kidney injury. The objective of this study was to elucidate the mechanistic role of HMGB1-TLR4 signaling in AngII-induced inflammation in the kidney. EXPERIMENTAL APPROACH Rat tubular epithelial cells (NRK52E) were treated with AngII over a preset time-course. In another set of experiments, HMGB1 was neutralized and TLR4 was knocked down using small interfering RNA targeting TLR4. Cell extracts were subjected to RT-PCR, immunoblotting, flow cytometry, and ELISA. KEY RESULTS AngII-induced inflammation in NRK52E cells increased gene and protein expression of TLR4, HMGB1 and key proinflammatory cytokines (TNFα and IL1β). Pretreatment with Losartan (an AT1 receptor blocker) attenuated the AngII-induced expression of TLR4 and inflammatory cytokines. TLR4 silencing was used to elucidate the specific role played by TLR4 in AngII-induced inflammation. TLR4siRNA treatment in these cells significantly decreased the AngII-induced inflammatory effect. Consistent observations were made when the Ang II treated cells were pretreated with anti-HMGB1. Downstream activation of NFκB and rate of generation of ROS was also decreased on gene silencing of TLR4 and exposure to anti-HMGB1. CONCLUSIONS AND IMPLICATIONS These results indicate a key role for HMGB1-TLR4 signaling in AngII-mediated inflammation in the renal epithelial cells. Our data also reveal that AngII-induced effects could be alleviated by HMGB1-TLR4 inhibition, suggesting this pathway as a potential therapeutic target for hypertensive renal dysfunctions.
Collapse
Affiliation(s)
- Anand R Nair
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Philip J Ebenezer
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Yogesh Saini
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Joseph Francis
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States.
| |
Collapse
|
49
|
Lim KM, Kwon JH, Kim K, Noh JY, Kang S, Park JM, Lee MY, Bae ON, Chung JH. Emodin inhibits tonic tension through suppressing PKCδ-mediated inhibition of myosin phosphatase in rat isolated thoracic aorta. Br J Pharmacol 2015; 171:4300-10. [PMID: 24909118 DOI: 10.1111/bph.12804] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 04/29/2014] [Accepted: 05/03/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Dysregulated tonic tension and calcium sensitization in blood vessels has frequently been observed in many cardiovascular diseases. Despite a huge therapeutic potential, little is known about natural products targeting tonic tension and calcium sensitization. EXPERIMENTAL APPROACH We screened natural products for inhibitory effects on vasoconstriction using the rat isolated thoracic aorta and found that an anthraquinone derivative, emodin, attenuated tonic tension. Organ bath system, primary vascular smooth muscle cells, confocal microscopy and Western blot analysis were employed to demonstrate the suppressive effects of emodin on PKCδ-mediated myosin phosphatase inhibition. KEY RESULTS Emodin, an active ingredient of Polygonum multiflorum extract, inhibited phenylephrine-induced vasoconstriction in rat isolated thoracic aorta, and inhibited vasoconstriction induced by 5-HT and endothelin-1. It also generally suppressed vasoconstrictions mediated by voltage-operated, store-operated calcium channels and intracellular calcium store. However, emodin did not affect agonist-induced calcium increases in primary smooth muscle cells. In contrast, post-treatment with emodin following phenylephrine stimulation potently suppressed tonic tension in rat aortic rings. Western blot analysis revealed that emodin inhibited phenylephrine-induced phospho-myosin light chain (pMLC) and the phosphorylation of myosin-targeting subunit and C-kinase-activated protein phosphatase-1 inhibitor (CPI-17). This was mediated by selective inhibition of PKCδ, whereas PKCα was not involved. CONCLUSION AND IMPLICATIONS Emodin attenuates tonic tension through the blockade of PKCδ and CPI-17-mediated MLC-phosphatase inhibition. This new mode of action for the suppression of tonic tension and structural insights into PKCδ inhibition revealed by emodin may provide new information for the development of modulators of tonic tension and for the treatment of hypertension.
Collapse
Affiliation(s)
- Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nemmar A, Al-Salam S, Yuvaraju P, Beegam S, Ali BH. Emodin mitigates diesel exhaust particles-induced increase in airway resistance, inflammation and oxidative stress in mice. Respir Physiol Neurobiol 2015; 215:51-7. [PMID: 26001677 DOI: 10.1016/j.resp.2015.05.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 01/16/2023]
Abstract
Clinical and experimental studies have reported that short-term exposure to particulate air pollution is associated with inflammation, oxidative stress and impairment of lung function. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) has a strong antioxidant and anti-inflammatory actions. Therefore, in the present study, we evaluated the possible ameliorative effect of emodin on diesel exhaust particles (DEP)-induced impairment of lung function, inflammation and oxidative stress in mice. Mice were intratracheally instilled with DEP (20 μg/mouse) or saline (control). Emodin was administered intraperitoneally 1h before and 7h after pulmonary exposure to DEP. Twenty-four hours following DEP exposure, we evaluated airway resistance measured by forced oscillation technique, lung inflammation and oxidative stress. Emodin treatment abated the DEP-induced increase in airway resistance, and prevented the influx of neutrophils in bronchoalveolar lavage fluid. Similarly, lung histopathology confirmed the protective effect of emodin on DEP-induced lung inflammation. DEP induced a significant increase of proinflammatory cytokines in the lung including tumor necrosis factor α, interleukin 6 and interleukin 1β. The latter effect was significantly ameliorated by emodin. DEP caused a significant increase in lung lipid peroxidation, reactive oxygen species and a significant decrease of reduced glutathione concentration. These effects were significantly mitigated by emodin. We conclude that emodin significantly mitigated DEP-induced increase of airway resistance, lung inflammation and oxidative stress. Pending further pharmacological and toxicological studies, emodin may be considered a potentially useful pulmonary protective agent against particulate air pollution-induced lung toxicity.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates.
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Priya Yuvaraju
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine & Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Al-Khod, Oman
| |
Collapse
|