1
|
Medrano-Padial C, Pérez-Novas I, Domínguez-Perles R, García-Viguera C, Medina S. Bioaccessible Phenolic Alkyl Esters of Wine Lees Decrease COX-2-Catalyzed Lipid Mediators of Oxidative Stress and Inflammation in a Time-Dependent Manner. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19016-19027. [PMID: 39145698 PMCID: PMC11363137 DOI: 10.1021/acs.jafc.4c05086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Lipophenols, phenolic compounds esterified with fatty alcohols or fatty acids, provide greater health benefits upon dietary ingestion of plant-based foods than unesterified (poly)phenols. Based on this premise, the present study aimed to demonstrate the role of gastrointestinal enzymes (pepsin, pancreatin, and pancreatic lipase) in releasing alkyl gallates and trans-caffeates from wine lees, providing bioactive compounds with enhanced capacities against oxidative stress (OS) and para-inflammation. The UHPLC-ESI-QqQ-MS/MS-based analysis revealed ethyl gallate and ethyl trans-caffeate as the most prominent compounds (1.675 and 0.872 μg/g dw, respectively), while the bioaccessibility of the derivatives of gallic and caffeic acids was dependent on the alkyl chain properties. The de novo formation of alkyl gallates during gastric and intestinal digestion resulted from intestinal enzyme activity. Moreover, the in vitro capacity of bioaccessible alkyl esters of gallic and trans-caffeic acids to reduce cyclooxygenase-2 concentration and modulate oxilipins related to OS (8-iso-PGF2α) and inflammation (PGF2α and PGE2) was demonstrated in a time-dependent manner. In conclusion, the presence of alkyl esters of gallic and trans-caffeic acids in wine lees and their subsequent formation during digestion of this byproduct emphasize their value as a source of antioxidant and anti-inflammatory compounds, encouraging the consideration of wine lees as a valuable ingredient for health-promoting coproducts.
Collapse
Affiliation(s)
- Concepción Medrano-Padial
- Laboratorio de Fitoquímica
y Alimentos Saludables (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo
25, 30100 Espinardo, Murcia, Spain
| | - Irene Pérez-Novas
- Laboratorio de Fitoquímica
y Alimentos Saludables (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo
25, 30100 Espinardo, Murcia, Spain
| | - Raúl Domínguez-Perles
- Laboratorio de Fitoquímica
y Alimentos Saludables (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo
25, 30100 Espinardo, Murcia, Spain
| | - Cristina García-Viguera
- Laboratorio de Fitoquímica
y Alimentos Saludables (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo
25, 30100 Espinardo, Murcia, Spain
| | - Sonia Medina
- Laboratorio de Fitoquímica
y Alimentos Saludables (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo
25, 30100 Espinardo, Murcia, Spain
| |
Collapse
|
2
|
Bhat P, Patil VS, Anand A, Bijjaragi S, Hegde GR, Hegde HV, Roy S. Ethyl gallate isolated from phenol-enriched fraction of Caesalpinia mimosoides Lam. Promotes cutaneous wound healing: a scientific validation through bioassay-guided fractionation. Front Pharmacol 2023; 14:1214220. [PMID: 37397484 PMCID: PMC10311562 DOI: 10.3389/fphar.2023.1214220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
The tender shoots of Caesalpinia mimosoides Lam. are used ethnomedically by the traditional healers of Uttara Kannada district, Karnataka (India) for the treatment of wounds. The current study was aimed at exploring phenol-enriched fraction (PEF) of crude ethanol extract of tender shoots to isolate and characterize the most active bio-constituent through bioassay-guided fractionation procedure. The successive fractionation and sub-fractionation of PEF, followed by in vitro scratch wound, antimicrobial, and antioxidant activities, yielded a highly active natural antioxidant compound ethyl gallate (EG). In vitro wound healing potentiality of EG was evidenced by a significantly higher percentage of cell migration in L929 fibroblast cells (97.98 ± 0.46% at 3.81 μg/ml concentration) compared to a positive control group (98.44 ± 0.36%) at the 48th hour of incubation. A significantly higher rate of wound contraction (98.72 ± 0.41%), an elevated tensile strength of the incised wound (1,154.60 ± 1.42 g/mm2), and increased quantity of connective tissue elements were observed in the granulation tissues of the 1% EG ointment treated animal group on the 15th post-wounding day. The accelerated wound healing activity of 1% EG was also exhibited by histopathological examinations through Hematoxylin and Eosin, Masson's trichome, and Toluidine blue-stained sections. Significant up-regulation of enzymatic and non-enzymatic antioxidant contents (reduced glutathione, superoxide dismutase, and catalase) and down-regulation of oxidative stress marker (lipid peroxidation) clearly indicates the effective granular antioxidant activity of 1% EG in preventing oxidative damage to the skin tissues. Further, in vitro antimicrobial and antioxidant activities of EG supports the positive correlation with its enhanced wound-healing activity. Moreover, molecular docking and dynamics for 100 ns revealed the stable binding of EG with cyclooxygenase-2 (-6.2 kcal/mol) and matrix metalloproteinase-9 (-4.6 kcal/mol) and unstable binding with tumor necrosis factor-α (-7.2 kcal/mol), suggesting the potential applicability of EG in inflammation and wound treatment.
Collapse
Affiliation(s)
- Pradeep Bhat
- Indian Council of Medical Research-National Institute of Traditional Medicine, Belagavi, India
- Post Graduate Department of Studies in Botany, Karnatak University, Dharwad, India
| | - Vishal S. Patil
- Indian Council of Medical Research-National Institute of Traditional Medicine, Belagavi, India
| | - Ashish Anand
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bengaluru, India
| | - Subhas Bijjaragi
- KLE’s SCP Arts, Science and D. D. Shirol Commerce College, Bagalkot, India
| | - Ganesh R. Hegde
- Post Graduate Department of Studies in Botany, Karnatak University, Dharwad, India
| | - Harsha V. Hegde
- Indian Council of Medical Research-National Institute of Traditional Medicine, Belagavi, India
| | - Subarna Roy
- Indian Council of Medical Research-National Institute of Traditional Medicine, Belagavi, India
| |
Collapse
|
3
|
Fan S, Feng X, Li K, Li B, Diao Y. Protective Mechanism of Ethyl Gallate against Intestinal Ischemia-Reperfusion Injury in Mice by in Vivo and in Vitro Studies Based on Transcriptomics. Chem Biodivers 2023; 20:e202200643. [PMID: 36513607 DOI: 10.1002/cbdv.202200643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Intestinal ischemia-reperfusion injury (IIRI) is a common clinical disease that can be life-threatening in severe cases. This study aimed to investigate the effects of ethyl gallate (EG) on IIRI and its underlying mechanisms. A mouse model was established to mimic human IIRI by clamping the superior mesenteric artery. Transcriptomics techniques were used in conjunction with experiments to explore the potential mechanisms of EG action. Intestinal histomorphological damage, including intestinal villi damage and mucosal hemorrhage, was significantly reversed by EG. EG also alleviated the oxidative stress, inflammation, and intestinal epithelial apoptosis caused by IIRI. 2592 up-regulated genes and 2754 down-regulated genes were identified after EG treatment, and these differential genes were enriched in signaling pathways, including fat digestion and absorption, and extracellular matrix (ECM) receptor interactions. In IIRI mouse intestinal tissue, expression of the differential protein matrix metalloproteinase 9 (MMP9), as well as its co-protein NF-κB-p65, was significantly increased, while EG inhibited the expression of MMP9 and NF-κB-p65. In Caco-2 cells in an established oxygen-glucose deprivation/reperfusion model (OGD/R), EG significantly reversed the decrease in intestinal barrier trans-epithelial electrical resistance (TEER). However, in the presence of MMP9 inhibitors, EG did not reverse the decreasing trend in TEER. This study illustrates the protective effect and mechanism of action of EG on IIRI and, combined with in vivo and in vitro experiments, it reveals that MMP9 may be the main target of EG action. This study provides new scientific information on the therapeutic effects of EG on IIRI.
Collapse
Affiliation(s)
- Shuyuan Fan
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Xiaoyan Feng
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Kun Li
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Bin Li
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China.,Dalian Anti-Infective Traditional Chinese Medicine, Development Engineering Technology Research Center, China
| |
Collapse
|
4
|
Sun G, Jiao M, Cui Y, Liang X, Liang X, Zhang S, Guo C. Identifying the mechanisms and molecular targets of Hongjingtian injection on treatment of TGFβ1-induced HK-2 cells: coupling network pharmacology with experimental verification. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1329. [PMID: 36660701 PMCID: PMC9843345 DOI: 10.21037/atm-22-5035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/02/2022] [Indexed: 01/01/2023]
Abstract
Background The study was designed to investigate the mechanism of Hongjingtian injection (HJT) in treating tubulointerstitial fibrosis (TIF) in chronic kidney diseases (CKD) based on network pharmacology and experimental verification. Methods First, active ingredients of HJT obtained from literature were screened using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and putative targets of active ingredients were predicted using the Chemmapper, SEA and Swiss Target Prediction database. Subsequently, the "compound-target" network for HJT was established. In addition, TIF disease targets were obtained from the GEO gene chips (accession number GSE20247). The intersecting targets of HJT and TIF obtained through Venny 2.1.0. The key targets and signaling pathways were determined by protein-protein interaction (PPI) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Finally, quantitative polymerase chain reaction (qPCR) and Western blot (WB) were used to validate the predicted five key genes targets (GAD1, SPHK1, P4HA2, AKR1B1, PTGES). And immunofluorescence, wound healing assay and transwell assay were used to verify the anti-fibrosis effect of HJT on TGFβ1-induced HK-2 cells. Results The network pharmacology analysis results showed that there are 36 active compounds and 1,044 putative target genes in HJT. HJT may exert its inhibitory effects against TIF by acting on 79 key targets. Besides, KEGG analysis indicated that the anti-TIF effect of HJT was mediated by multiple pathways, such as the metabolic pathway, pathways in cancer and gap junction. Among them, GAD1, SPHK1, P4HA2, AKR1B1 and PTGES are enriched in the metabolic pathway. In vitro induced cell model experiments, the immunofluorescence experience showed that HJT could restore EMT of HK-2 cells. In addition, the qPCR and WB results showed that HJT significantly restored the expression of the SPHK1 in HK-2 cells induced by TGF-β1. Conclusions This study comprehensively illuminated the active compounds, potential targets, and molecular mechanism of HJT against TIF. HJT treatment of TIF may reverse EMT caused by TGF-β1 by targeting SPHK1.
Collapse
Affiliation(s)
- Guanghui Sun
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yuying Cui
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuezhen Liang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China;,Orthopaedic Microsurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaodong Liang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shanshan Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Congcong Guo
- Department of Endocrinology and Metabology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China;,Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
5
|
The role of lactoferrin in atherosclerosis. Biometals 2022; 36:509-519. [PMID: 36053470 DOI: 10.1007/s10534-022-00441-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/24/2022] [Indexed: 11/02/2022]
Abstract
Atherosclerosis (AS) is a common pathological basis for many cardiovascular diseases (CVDs) and result in high mortality and immense health and economic burdens worldwide. Early prevention, diagnosis, and treatment are promising approaches for stemming the development and progression of AS. Lactoferrin (Lf) is an iron-binding glycoprotein belonging to the transferrin family. It is widely found in body fluids such as digestive tract fluids, tears, and milk. Lf possesses anti-inflammatory, antibacterial, immunoregulatory, antioxidant and many other physiological functions. The serum Lf level is reportedly associated with the risk of AS and AS-related CVDs. Lf administration is closely involved in several mechanisms, including cholesterol metabolism, foam cell formation, ICAM-1 expression, homocysteine and leptin levels, anti-inflammatory and antioxidant function. Moreover, Lf has also been applied in the sythesis of magnetic resonance imaging (MRI) contrast agents to detect AS. Lf plays an important role in AS and may therefore be used in its diagnosis and treatment. Thus, this article aims to review the association between Lf and the risk of AS and AS-related CVDs, the mechanisms of Lf administration on AS, and its potential application in AS diagnosis.
Collapse
|
6
|
Andrade JKS, Barros RGC, Pereira UC, Gualberto NC, de Oliveira CS, Shanmugam S, Narain N. α-Amylase inhibition, cytotoxicity and influence of the in vitro gastrointestinal digestion on the bioaccessibility of phenolic compounds in the peel and seed of Theobroma grandiflorum. Food Chem 2021; 373:131494. [PMID: 34753077 DOI: 10.1016/j.foodchem.2021.131494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 11/18/2022]
Abstract
The aim of this work was to evaluate the bioaccessibility, cytotoxicity, antioxidant and antidiabetic potential of peel and seeds of cupuassu (Theobroma grandiflorum). Thus, the extracts of cupuassu were evaluated for inhibition of α-amylase, cytotoxicity, and bioaccessibility after gastrointestinal digestion and probiotic fermentation (Lactobacillus delbrueckii, Lactobacillus jhonsoni, Lactobacillus rhamus and Bifidobacterium longum). Digestion increased concentrations of phenolics, showing bioaccessibility of up to 274.13% (total phenolics) and 1105.15% (ORAC). β-carotene, quinic, tartaric, malic, citric, epicatechin, ethyl gallate, epigallocatechin gallate, gallic acid, pyrocatechol, vanillin, ramnetine were the main compounds while the epicatechin, ethyl gallate, gallic acid and pyrocatechol were the major effective phenolic compounds. The extracts did not show toxic effects and the cupuassu seeds showed 97% inhibition of α-amylase and 47.91% bioaccessibility of pyrocatechol. This study suggests that cupuassu extracts are sources of natural antioxidants with promising antidiabetic potential, and probiotics are able to increase phenolic compounds, responsible for health benefits.
Collapse
Affiliation(s)
| | - Romy Gleyse Chagas Barros
- Laboratory of Flavor and Chromatographic Analysis, PROCTA, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Ubatã Corrêa Pereira
- Laboratory of Flavor and Chromatographic Analysis, PROCTA, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Nayjara Carvalho Gualberto
- Laboratory of Flavor and Chromatographic Analysis, PROCTA, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Christean Santos de Oliveira
- Laboratory of Flavor and Chromatographic Analysis, PROCTA, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Saravanan Shanmugam
- Department of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, CEP 49100-000, São Cristóvão, SE, Brazil
| | - Narendra Narain
- Laboratory of Flavor and Chromatographic Analysis, PROCTA, Federal University of Sergipe, São Cristóvão, SE, Brazil.
| |
Collapse
|
7
|
Yang Y, Wang D, Zhang C, Yang W, Li C, Gao Z, Pei K, Li Y. Piezo1 mediates endothelial atherogenic inflammatory responses via regulation of YAP/TAZ activation. Hum Cell 2021; 35:51-62. [PMID: 34606042 DOI: 10.1007/s13577-021-00600-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
The vascular endothelium plays a key role in the pathobiology of atherosclerotic cardiovascular disease. Endothelial cell Piezo1 mediates blood vessel formation, angiogenesis and regulation of blood pressure. However, changes of Piezo1 expression in atherosclerosis (AS) and the role of Piezo1 in the progression of atherosclerotic diseases remains obscure. Thus, the current study is to elucidate the role and mechanism of which Piezo1 mediates vascular inflammation in atherosclerotic mice and vascular endothelial inflammation induced by oxidized low density lipoprotein (ox-LDL) in vitro. Here, we have shown that the expression of Piezo1 was significantly increased in the stenotic carotid artery of ApoE-/- mice fed by high-fat diet (HFD). Pharmacological inhibition of Piezo1 (GsMTx-4) attenuated plaque formation, decreased the level of inflammation related factors (JNK, TNF-α, NF-κB, VCAM-1) of carotid plaque in atherosclerotic mice. Meanwhile, ox-LDL also upregulates Piezo1 and inflammation proteins (NF-κB, JNK and TNF-α) in endothelium cells (ECs). YAP/TAZ is activated accompanied by the enhanced Piezo1 activity in ECs induced by ox-LDL. Interference by siRNA of Piezo1 abolished the expression of YAP/TAZ and inflammation proteins (JNK, NF-κB and TNF-α). In addition, Ca2+ influx in ECs induced by ox-LDL was increased than control group, Piezo1 siRNA can reduce the calcium content. Piezo1 agonist Yoda1 increased Ca2+ influx and promote YAP nucleus translocation in ECs, genetic deletion of Piezo1 reversed it. Our results indicate that Piezo1 could mediate endothelial atherogenic inflammatory responses via regulation of YAP/TAZ activation and nuclear localization. Piezo1 may be a potential therapeutic target for atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Ying Yang
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China.,Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Danyang Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China.,Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Chunxiao Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China
| | - Wenqing Yang
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China
| | - Chao Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China
| | - Zichen Gao
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China
| | - Ke Pei
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China
| | - Yunlun Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250003, People's Republic of China.
| |
Collapse
|
8
|
Chen W, Zhong Y, Feng N, Guo Z, Wang S, Xing D. New horizons in the roles and associations of COX-2 and novel natural inhibitors in cardiovascular diseases. Mol Med 2021; 27:123. [PMID: 34592918 PMCID: PMC8482621 DOI: 10.1186/s10020-021-00358-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 01/03/2023] Open
Abstract
Age-related cardiovascular disease is the leading cause of death in elderly populations. Coxibs, including celecoxib, valdecoxib, etoricoxib, parecoxib, lumiracoxib, and rofecoxib, are selective cyclooxygenase-2 (COX-2) inhibitors used to treat osteoarthritis and rheumatoid arthritis. However, many coxibs have been discontinued due to adverse cardiovascular events. COX-2 contains cyclooxygenase (COX) and peroxidase (POX) sites. COX-2 inhibitors block COX activity without affecting POX activity. Recently, quercetin-like flavonoid compounds with OH groups in their B-rings have been found to serve as activators of COX-2 by binding the POX site. Galangin-like flavonol compounds serve as inhibitors of COX-2. Interestingly, nabumetone, flurbiprofen axetil, piketoprofen-amide, and nepafenac are ester prodrugs that inhibit COX-2. The combination of galangin-like flavonol compounds with these prodrug metabolites may lead to the development of novel COX-2 inhibitors. This review focuses on the most compelling evidence regarding the role and mechanism of COX-2 in cardiovascular diseases and demonstrates that quercetin-like compounds exert potential cardioprotective effects by serving as cofactors of COX-2.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Yingjie Zhong
- Cancer Institute, Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Nuan Feng
- Department of Nutrition, Qingdao Women and Children's Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Zhu Guo
- Cancer Institute, Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Shuai Wang
- School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, 261053, Shandong, China.
| | - Dongming Xing
- Cancer Institute, Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China. .,School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
9
|
Yang C, Li P, Wang P, Zhu BT. Mechanism of reactivation of the peroxidase catalytic activity of human cyclooxygenases by reducing cosubstrate quercetin. J Mol Graph Model 2021; 107:107941. [PMID: 34091174 DOI: 10.1016/j.jmgm.2021.107941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/22/2021] [Accepted: 05/11/2021] [Indexed: 11/19/2022]
Abstract
Our earlier studies show that the peroxidase activity of cyclooxygenase 1 and 2 (COX-1 and COX-2) can be reactivated in vitro and in vivo by the presence of certain naturally-occurring flavonoids such as quercetin and myricetin, which serve as reducing cosubstrates. These compounds can activate COX at nanomolar concentrations. In the present study, quercetin is used as a representative model compound to investigate the chemical mechanism by which the peroxidase activity of human COX-1 and COX-2 is reactivated after each catalytic cycle. Molecular docking and quantum mechanics calculations are carried out to probe the interactions of quercetin with the peroxidase sites of COX-1/2 and the reactivation mechanism. It is found that some of the partially-ionized states of quercetin can bind tightly and closely inside the peroxidase active sites of the COX enzymes and directly interact with heme Fe ion. While quercetin contains several phenolic hydroxyl groups, it is found that only the C-3'-OH group can effectively donate an electron for the reduction of heme because it not only can bind closely and tightly inside the peroxidase sites of COX-1/2, but it can also facilely donate an electron to heme Fe ion. This investigation provides a mechanistic explanation for the chemical process by which quercetin reactivates COX-1/2 peroxidases. This knowledge would aid in the rational design of drugs that can selectively target the peroxidase sites of COX-1/2 either as activators or inhibitors.
Collapse
Affiliation(s)
- Chengxi Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Peng Li
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Shenzhen Bay Laboratory, Shenzhen 518055, China.
| |
Collapse
|
10
|
Bai HW, Yang C, Wang P, Rao S, Zhu BT. Inhibition of cyclooxygenase by blocking the reducing cosubstrate at the peroxidase site: Discovery of galangin as a novel cyclooxygenase inhibitor. Eur J Pharmacol 2021; 899:174036. [PMID: 33737009 DOI: 10.1016/j.ejphar.2021.174036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/01/2022]
Abstract
Earlier we have shown that certain flavonoids (e.g., quercetin) are high-affinity reducing cosubstrates for cyclooxygenase (COX) 1 and 2. These compounds can bind inside the peroxidase active sites of COXs and donate an electron from one of their B-ring hydroxyl groups to hematin. Based on these earlier findings, it is postulated that some of the natural flavonoids such as galangin that are structural analogs of quercetin but lack the proper B-ring hydroxyl groups might function as novel inhibitors of COXs by blocking the effect of the reducing cosubstrates. This idea is tested in the present study. Computational docking analysis together with quantum chemistry calculation shows that galangin can bind inside the peroxidase active sites of COX-1 and COX-2 in a similar manner as quercetin, but it has little ability to effectively donate its electrons, thereby blocking the effect of the reducing cosubstrates like quercetin. Further experimental studies confirm that galangin can inhibit, both in vitro and in vivo, quercetin-mediated activation of the peroxidase activity of the COX-1/2 enzymes. The results of the present study demonstrate that galangin is a novel naturally-occurring inhibitor of COX-1 and COX-2, acting by blocking the function of the reducing cosubstrates at the peroxidase sites.
Collapse
Affiliation(s)
- Hyoung-Woo Bai
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; Present Address: Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - Chengxi Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Shun Rao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
11
|
Lan T, Xue X, Dunmall LC, Miao J, Wang Y. Patient-derived xenograft: a developing tool for screening biomarkers and potential therapeutic targets for human esophageal cancers. Aging (Albany NY) 2021; 13:12273-12293. [PMID: 33903283 PMCID: PMC8109069 DOI: 10.18632/aging.202934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/23/2021] [Indexed: 04/15/2023]
Abstract
Esophageal cancer (EC) represents a human malignancy, diagnosed often at the advanced stage of cancer and resulting in high morbidity and mortality. The development of precision medicine allows for the identification of more personalized therapeutic strategies to improve cancer treatment. By implanting primary cancer tissues into immunodeficient mice for expansion, patient-derived xenograft (PDX) models largely maintain similar histological and genetic representations naturally found in patients' tumor cells. PDX models of EC (EC-PDX) provide fine platforms to investigate the tumor microenvironment, tumor genomic heterogeneity, and tumor response to chemoradiotherapy, which are necessary for new drug discovery to combat EC in addition to optimization of current therapeutic strategies for EC. In this review, we summarize the methods used for establishing EC-PDX models and investigate the utilities of EC-PDX in screening predictive biomarkers and potential therapeutic targets. The challenge of this promising research tool is also discussed.
Collapse
Affiliation(s)
- Tianfeng Lan
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xia Xue
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- The Academy of Medical Science, Precision Medicine Center of the Second Affiliated Hospital of Zhengzhou University, Zhengzhou University, Henan, P.R. China
| | - Louisa Chard Dunmall
- Centre for Cancer Biomarkers and Biotherapeuitcs, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jinxin Miao
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, Henan, P.R. China
| | - Yaohe Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- Centre for Cancer Biomarkers and Biotherapeuitcs, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
12
|
Liu W, Liu J, Xing S, Pan X, Wei S, Zhou M, Li Z, Wang L, Bielicki JK. The benzoate plant metabolite ethyl gallate prevents cellular- and vascular-lipid accumulation in experimental models of atherosclerosis. Biochem Biophys Res Commun 2021; 556:65-71. [PMID: 33839416 DOI: 10.1016/j.bbrc.2021.03.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 11/18/2022]
Abstract
Ethyl gallate (EG) is a well-known constituent of medicinal plants, but its effects on atherosclerosis development are not clear. In the present study, the anti-atherosclerosis effects of EG and the underlying mechanisms were explored using macrophage cultures, zebrafish and apolipoprotein (apo) E deficient mice. Treatment of macrophages with EG (20 μM) enhanced cellular cholesterol efflux to HDL, and reduced net lipid accumulation in response to oxidized LDL. Secretion of monocyte chemotactic protein-1 (MCP-1) and interleukin-6 (IL-6) from activated macrophages was also blunted by EG. Fluorescence imaging techniques revealed EG feeding of zebrafish reduced vascular lipid accumulation and inflammatory responses in vivo. Similar results were obtained in apoE-/- mice 6.5 months of age, where plaque lesions and monocyte infiltration into the artery wall were reduced by 70% and 42%, respectively, after just 6 weeks of injections with EG (20 mg/kg). HDL-cholesterol increased 2-fold, serum cholesterol efflux capacity increased by ∼30%, and the levels of MCP-1 and IL-6 were reduced with EG treatment of mice. These results suggest EG impedes early atherosclerosis development by reducing the lipid and macrophage-content of plaque. Underlying mechanisms appeared to involve HDL cholesterol efflux mechanisms and suppression of pro-inflammatory cytokine secretion.
Collapse
Affiliation(s)
- Wenjie Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Jianmin Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Shu Xing
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Xuefang Pan
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Sheng Wei
- Behavioral Phenotyping Core Facility, Shandong University of Traditional Chinese Medicine, Jinan 250353, China.
| | - Mingyang Zhou
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Zifa Li
- Behavioral Phenotyping Core Facility, Shandong University of Traditional Chinese Medicine, Jinan 250353, China
| | - Ling Wang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - John Kevin Bielicki
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| |
Collapse
|
13
|
Milinčić DD, Stanisavljević NS, Kostić AŽ, Soković Bajić S, Kojić MO, Gašić UM, Barać MB, Stanojević SP, Lj Tešić Ž, Pešić MB. Phenolic compounds and biopotential of grape pomace extracts from Prokupac red grape variety. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2020.110739] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
14
|
Yang C, Li P, Ding X, Sui HC, Rao S, Hsu CH, Leung WP, Cheng GJ, Wang P, Zhu BT. Mechanism for the reactivation of the peroxidase activity of human cyclooxygenases: investigation using phenol as a reducing cosubstrate. Sci Rep 2020; 10:15187. [PMID: 32938962 PMCID: PMC7494923 DOI: 10.1038/s41598-020-71237-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
It has been known for many years that the peroxidase activity of cyclooxygenase 1 and 2 (COX-1 and COX-2) can be reactivated in vitro by the presence of phenol, which serves as a reducing compound, but the underlying mechanism is still poorly understood. In the present study, we use phenol as a model compound to investigate the mechanism by which the peroxidase activity of human COXs is reactivated after each catalytic cycle. Molecular docking and quantum mechanics calculations are carried out to probe the interaction of phenol with the peroxidase site of COXs and the reactivation mechanism. It is found that the oxygen atom associated with the Fe ion in the heme group (i.e., the complex of Fe ion and porphyrin) of COXs can be removed by addition of two protons. Following its removal, phenol can readily bind inside the peroxidase active sites of the COX enzymes, and directly interact with Fe in heme to facilitate electron transfer from phenol to heme. This investigation provides theoretical evidence for several intermediates formed in the COX peroxidase reactivation cycle, thereby unveiling mechanistic details that would aid in future rational design of drugs that target the peroxidase site.
Collapse
Affiliation(s)
- Chengxi Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Peng Li
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Xiaoli Ding
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Hao Chen Sui
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Shun Rao
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Chia-Hsiang Hsu
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Wing-Por Leung
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Gui-Juan Cheng
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China. .,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China. .,Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China. .,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China. .,Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
15
|
Chen W, Wang S, Wu Y, Shen X, Xu S, Guo Z, Zhang R, Xing D. The Physiologic Activity and Mechanism of Quercetin-Like Natural Plant Flavonoids. Curr Pharm Biotechnol 2020; 21:654-658. [PMID: 32048963 DOI: 10.2174/1389201021666200212093130] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/29/2019] [Accepted: 01/18/2020] [Indexed: 01/01/2023]
Abstract
The term "vitamin P" is an old but interesting concept. Most substances in this category belong to the family of flavonoids. "Vitamin P" has also been used to define the activity of some flavonoids, including quercetin, myricetin, and rutin. According to experimental studies, the "quercetin-like natural plant flavonoids" are beneficial to the body due to their various physiological and pharmacological activities in large doses (5 μM in vitro, 50 mg/kg in mice and 100 mg/kg in rats). The physiologically achievable concentration is 10 to 100 nM, which is quite high and hard to achieve from a normal diet. Thus, the physiologic activity and mechanism of "vitamin P" are still not clear. It should be noted that the quercetin-like natural plant flavonoids are physiological co-factors of cyclooxygenases (COXs), which are the rate-limiting key enzymes of prostaglandins. These quercetin-like natural plant flavonoids can strongly stimulate prostaglandin levels at lower doses (10 nM in vitro and in 0.1 mg/kg in vivo in rats). Although these "vitamin P" substances are not original substances in the body, their physiological functions affect the body. This review is focused on the most compelling evidence regarding the physiologic role and mechanism of quercetin-like natural plant flavonoids, which may be useful in understanding the physiological functions of "vitamin P", with the goal of focusing on the role of flavonoids in human physiological health.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China,Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong, 261041, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China
| | - Xin Shen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China
| | - Shutan Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China
| | - Zhu Guo
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China,Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China
| | - Renshuai Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China,Cancer Institute, Qingdao University, Qingdao, Shandong, 266071, China,Qingdao Cancer Institute, Qingdao, Shandong, 266071, China,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|