1
|
Lucas M, Freitas M, Silva AMS, Fernandes E, Ribeiro D. Styrylchromones: Biological Activities and Structure-Activity Relationship. ChemMedChem 2025; 20:e202400782. [PMID: 39480961 DOI: 10.1002/cmdc.202400782] [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: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
Styrylchromones (SC) are a group of oxygen-containing heterocyclic compounds, which are characterized by the attachment of a styryl group to the chromone core. SC can be found in nature or can be chemically synthesized in the laboratory. As their presence in nature is scarce, the synthetic origin is the most common. Two types of SC are known: 2-styrylchromones and 3-styrylchromones. However, 2-styrylchromones are the most common, being more broadly found in nature and whose chemical synthesis is more commonly described. A wide variety of SC has been described in the literature, with different substituents in different positions, the majority of which are distributed on the A- and/or B-rings. Over the years, several biological activities have been attributed to SC. This work presents a comprehensive review of the biological activities attributed to SC and their structure-activity relationship, based on a published literature search, since 1989. The following biological activities are thoroughly reviewed and discussed in this review: antioxidant, antiallergic, antiviral, antibacterial, antifungal, anti-inflammatory and antitumoral, affinity and selectivity for A3 adenosine receptors, neuroprotective, and α-glucosidase inhibition. In general, SC are composed by a promising scaffold with great potential for the development of new drugs.
Collapse
Affiliation(s)
- Mariana Lucas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - Artur M S Silva
- LAQV-REQUIMTE & QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
- Faculty of Agrarian Sciences and Environment, University of the Azores, Rua Capitão João d'Ávila-Pico da Urze, 9700-042, Angra do Heroísmo, Açores, Portugal
| |
Collapse
|
2
|
Shuai E, Xiao S, Huang J, Zeng Z, Liu S, Tan J, Zhang H, Cai W. Screening of anti-inflammatory active components in Sabia schumanniana Diels by affinity ultrafiltration and UHPLC-Q-Exactive Orbitrap mass spectrometry. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118845. [PMID: 39306211 DOI: 10.1016/j.jep.2024.118845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sabia schumanniana Diels is a traditional botanical used to treat lumbago and arthralgia. However, there has been limited research on the pharmacological effects of its chemical components. AIM OF THE STUDY This study aimed to rapidly screen for anti-inflammatory compounds in Sabia schumanniana Diels. MATERIALS AND METHODS An affinity ultrafiltration method based on UHPLC-Q-Exactive Orbitrap MS was established to rapidly screen and identify cyclooxygenase-2 (COX-2) receptor ligands. The reliability of this method was verified by molecular docking analysis and experiments with RAW264.7 cells. RESULTS Seventeen ligands were identified from Sabia schumanniana Diels using affinity ultrafiltration. Molecular docking results indicated that these ligands specifically docked with COX-2. Among them, N-nornuciferine exhibited notable anti-inflammatory activity. CONCLUSIONS The combination of affinity ultrafiltration and UHPLC-Q-Exactive Orbitrap MS is an effective and precise method for screening anti-inflammatory compounds. This study provides a foundation for further research on Sabia schumanniana Diels and offers guidance for its potential clinical applications.
Collapse
Affiliation(s)
- E Shuai
- School of Pharmacy, Weifang Medical University, Weifang, 261000, China
| | - Shunli Xiao
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China
| | - Jin Huang
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China
| | - Zihui Zeng
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China
| | - Siqiong Liu
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China
| | - Jingjing Tan
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China
| | - He Zhang
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China
| | - Wei Cai
- School of Pharmaceutical Sciences, Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, 418000, China.
| |
Collapse
|
3
|
Jain P, Satija J, Sudandiradoss C. Discovery of andrographolide hit analog as a potent cyclooxygenase-2 inhibitor through consensus MD-simulation, electrostatic potential energy simulation and ligand efficiency metrics. Sci Rep 2023; 13:8147. [PMID: 37208387 PMCID: PMC10199084 DOI: 10.1038/s41598-023-35192-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/14/2023] [Indexed: 05/21/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) is the key enzyme responsible for the conversion of arachidonic acid to prostaglandins that display pro-inflammatory properties and thus, it is a potential target protein to develop anti-inflammatory drugs. In this study, chemical and bio-informatics approaches have been employed to find a novel potent andrographolide (AGP) analog as a COX-2 inhibitor having better pharmacological properties than aspirin and rofecoxib (controls). The full amino acid sequenced human Alpha fold (AF) COX-2 protein (604AA) was selected and validated for its accuracy against the reported COX-2 protein structures (PDB ID: 5F19, 5KIR, 5F1A, 5IKQ and 1V0X) followed by multiple sequence alignment analysis to establish the sequence conservation. The systematic virtual screening of 237 AGP analogs against AF-COX-2 protein yielded 22 lead compounds based on the binding energy score (< - 8.0 kcal/mol). These were further screened out to 7 analogs by molecular docking analysis and investigated further for ADMET prediction, ligand efficiency metrics calculations, quantum mechanical analysis, MD simulation, electrostatic potential energy (EPE) docking simulation, and MM/GBSA. In-depth analysis revealed that AGP analog A3 (3-[2-[(1R,4aR,5R,6R,8aR)-6-hydroxy-5,6,8a-trimethyl-2-methylidene-3,4,4a,5,7,8-hexahydro-1H-naphthalen-1-yl]ethylidene]-4-hydroxyoxolan-2-one) forms the most stable complex with the AF-COX-2 showing the least RMSD value (0.37 ± 0.03 nm), a good number of hydrogen bonds (protein-ligand H-bond = 11, and protein H-bond = 525), minimum EPE score (- 53.81 kcal/mol), and lowest MM-GBSA before and after simulation (- 55.37 and - 56.25 kcal/mol, respectively) value compared to other analogs and controls. Thus, we suggest that the identified A3 AGP analog could be developed as a promising plant-based anti-inflammatory drug by inhibiting COX-2.
Collapse
Affiliation(s)
- Priyanka Jain
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Jitendra Satija
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - C Sudandiradoss
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
4
|
Yang C, Wang T, Zhu S, Zong Z, Luo C, Zhao Y, Liu J, Li T, Liu X, Liu C, Deng H. Nicotinamide N-Methyltransferase Remodeled Cell Metabolism and Aggravated Proinflammatory Responses by Activating STAT3/IL1β/PGE 2 Pathway. ACS OMEGA 2022; 7:37509-37519. [PMID: 36312432 PMCID: PMC9607676 DOI: 10.1021/acsomega.2c04286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
Nicotinamide N-methyltransferase (NNMT) is a cytosolic methyltransferase, catalyzing N-methylation of nicotinamide (NAM) to form 1-methylnicotinamide (1-MNAM), in which S-adenosyl-l-methionine (SAM) is the methyl donor. It has been well documented that NNMT is elevated in multiple cancers and promotes tumor aggressiveness. In the present study, we investigated the effects of NNMT overexpression on cellular metabolism and proinflammatory responses. We found that NNMT overexpression reduced NAD+ and SAM levels, and activated the STAT3 signaling pathway. Consequently, STAT3 activation upregulated interleukin 1β (IL1β) and cyclooxygenase-2 (COX2), leading to prostaglandin E2 (PGE2) accumulation. On the other hand, NNMT downregulated 15-hydroxyprostaglandin dehydrogenase (15-PGDH) which catalyzes PGE2 into inactive molecules. Moreover, secretomic data indicated that NNMT promoted secretion of collagens, pro-inflammatory cytokines, and extracellular matrix proteins, confirming NNMT aggravated inflammatory responses to promote cell growth, migration, epithelial-mesenchymal transition (EMT), and chemoresistance. Taken together, we showed that NNMT played a pro-inflammatory role in cancer cells by activating the STAT3/IL1β/PGE2 axis and proposed that NNMT was a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Changmei Yang
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Tianxiang Wang
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Songbiao Zhu
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Zhaoyun Zong
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Chengting Luo
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Yujiao Zhao
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Jing Liu
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Ting Li
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Xiaohui Liu
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| | - Chongdong Liu
- Chao
Yang Hospital of Capital Medical University, Beijing 100020, P. R. China
| | - Haiteng Deng
- MOE
Key Laboratory of Bioinformatics, Center for Synthetic and Systematic
Biology, School of Life Sciences, Tsinghua
University, Beijing 100084, P. R. China
| |
Collapse
|
5
|
Yang YS, Man RJ, Xu JF, Wang CY, Wang X, Li DD, Zhu HL. Discovery of novel 1,3-diaryl pyrazolyl ester derivatives as COX-2 inhibitory candidates with anti-tumor effect. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
6
|
Zhou P, Wu S, Huang D, Wang K, Su X, Yang R, Shao C, Wu J. Oral exposure to DEHP may stimulate prostatic hyperplasia associated with upregulation of COX-2 and L-PGDS expressions in male adult rats. Reprod Toxicol 2022; 112:160-170. [PMID: 35905844 DOI: 10.1016/j.reprotox.2022.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), a typical environmental endocrine disruptor (EED), can disrupt estrogen and androgen secretion and metabolism process, thus inducing dysfunctional reproduction such as impaired gonadal development and spermatogenesis disorder. Prostaglandin synthases (PGS) catalyze various prostaglandins biosynthesis, involved in inflammatory cascade and tumorigenesis. Yet, little is known about how PGS may impact prostatic hyperplasia development and progression. This study concentrates predominantly on the potential prostatic toxicity of DEHP exposure and the mediating role of PGS. In vivo study, adult male rats were administered via oral gavage 30 μg/kg/d, 90 μg/kg/d, 270 μg/kg/d, 810 μg/kg/d DEHP or vehicle for four weeks. The results elucidated that low-dose DEHP may cause the proliferation of the prostate with an increased PCNA/TUNEL ratio. Given the importance of estrogens and androgens in prostatic hyperplasia, our first objective was to evaluate the levels of sex hormones. DEHP improved the ratio of estradiol (E2)/testosterone (T) in a dose-dependent manner and upregulated estrogen receptor alpha (ERα) and androgen receptor (AR) expressions. Prostaglandin synthases, including cyclooxygenase-2 (COX-2) and lipocalin-type prostaglandin D synthase (L-PGDS), were significantly upregulated in the ventral prostate. COX-2 and L-PGDS might mediate the tendency of prostatic hyperplasia induced by low-dose DEHP through estradiol/androgen regulation and imbalance between proliferation and apoptosis in vivo. These findings provide the first evidence that prostaglandin synthases contribute to the tendency toward benign prostatic hyperplasia induced by DEHP. Further investigations will have to be performed to facilitate an improved understanding of the role of prostaglandin synthases in DEHP-induced prostatic lesions.
Collapse
Affiliation(s)
- Ping Zhou
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Shuangshuang Wu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Dongyan Huang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Kaiyue Wang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Xin Su
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Rongfu Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Congcong Shao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China
| | - Jianhui Wu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School ️of Pharmacy, Fudan University, China.
| |
Collapse
|
7
|
Chen B, Tao W, Yan L, Zeng M, Song L, Huang Z, Chen F. Molecular feature of arterial remodeling in the brain arteriovenous malformation revealed by arteriovenous shunt rat model and RNA sequencing. Int Immunopharmacol 2022; 107:108653. [PMID: 35247777 DOI: 10.1016/j.intimp.2022.108653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/05/2022] [Accepted: 02/20/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Morphological research suggested the feeding artery of brain arteriovenous malformation (bAVM) had vascular remodeling under the high blood flow; however, the underlying molecular mechanisms were unclear. METHODS We constructed 32 simplified AVM rat models in four groups: the control group (n = 6), 1-week high-blood-flow group (n = 9), 3-week high-blood-flow group (n = 7) and 6-week high-blood-flow group (n = 10). The circumference, blood velocity, blood flow, pressure, and wall shear of the feeding artery were measured or calculated. The arterial wall change was observed by Masson staining. RNA sequencing (RNA-seq) of feeding arteries was performed, followed by bioinformatics analysis to detect the potential molecular mechanism for bAVM artery remodeling under the high blood flow. RESULTS We observed hemodynamic injury and vascular remodeling on the feeding artery under the high blood flow. RNA-seq showed immune/inflammation infiltration and vascular smooth muscle cell (VSMC) phenotype transformation during remodeling. Weighted gene co-expression network analysis (WGCNA) and time series analysis further identified 27 key genes and pathways involved in remodeling. Upstream miRNA and molecular drugs were predicted targeting these key genes. CONCLUSIONS We depicted molecular change of bAVM arterial remodeling via RNA-seq in high-blood-flow rat models. Twenty-seven key genes may regulate immune/inflammation infiltration and VSMC phenotype transform in bAVM arterial remodeling.
Collapse
Affiliation(s)
- Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wengui Tao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Langchao Yan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Laixin Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Neurosurgery, The Second Affiliated Hospital of Mudanjiang Medical College, Mudanjiang, Heilongjiang, China; Department of Surgery, Mudanjiang Huimin Hospital, Mudanjiang, Heilongjiang, China
| | - Zheng Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fenghua Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Lucas M, Freitas M, Silva AMS, Fernandes E, Ribeiro D. Styrylchromones: Biological Activities and Structure-Activity Relationship. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2804521. [PMID: 34987699 PMCID: PMC8720608 DOI: 10.1155/2021/2804521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/14/2021] [Accepted: 10/04/2021] [Indexed: 11/24/2022]
Abstract
Styrylchromones (SC) are a group of oxygen-containing heterocyclic compounds, which are characterized by the attachment of a styryl group to the chromone core. SC can be found in nature or can be chemically synthesized in the laboratory. As their presence in nature is scarce, the synthetic origin is the most common. Two types of SC are known: 2-styrylchromones and 3-styrylchromones. However, 2-styrylchromones are the most common, being more commonly found in nature and which chemical synthesis is more commonly described. A wide variety of SC has been described in the literature, with different substituents in different positions, the majority of which are distributed on the A- and/or B-rings. Over the years, several biological activities have been attributed to SC. This work presents a comprehensive review of the biological activities attributed to SC and their structure-activity relationship, based on a published literature search, since 1989. The following biological activities are thoroughly revised and discussed in this review: antioxidant, antiallergic, antiviral, antibacterial, antifungal, anti-inflammatory, and antitumoral, affinity and selectivity for A3 adenosine receptors, neuroprotective, and α-glucosidase inhibition. In general, SC are composed by a promising scaffold with great potential for the development of new drugs.
Collapse
Affiliation(s)
- Mariana Lucas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Artur M. S. Silva
- LAQV-REQUIMTE & QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Faculty of Agrarian Sciences and Environment, University of the Azores, 9700-042 Angra do Heroísmo, Açores, Portugal
| |
Collapse
|
9
|
Kountouras J, Papaefthymiou A, Polyzos SA, Deretzi G, Vardaka E, Soteriades ES, Tzitiridou-Chatzopoulou M, Gkolfakis P, Karafyllidou K, Doulberis M. Impact of Helicobacter pylori-Related Metabolic Syndrome Parameters on Arterial Hypertension. Microorganisms 2021; 9:microorganisms9112351. [PMID: 34835476 PMCID: PMC8618184 DOI: 10.3390/microorganisms9112351] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/28/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Arterial hypertension is a risk factor for several pathologies, mainly including cardio-cerebrovascular diseases, which rank as leading causes of morbidity and mortality worldwide. Arterial hypertension also constitutes a fundamental component of the metabolic syndrome. Helicobacter pylori infection is one of the most common types of chronic infection globally and displays a plethora of both gastric and extragastric effects. Among other entities, Helicobacter pylori has been implicated in the pathogenesis of the metabolic syndrome. Within this review, we illustrate the current state-of-the-art evidence, which may link several components of the Helicobacter pylori-related metabolic syndrome, including non-alcoholic fatty liver disease and arterial hypertension. In particular, current knowledge of how Helicobacter pylori exerts its virulence through dietary, inflammatory and metabolic pathways will be discussed. Although there is still no causative link between these entities, the emerging evidence from both basic and clinical research supports the proposal that several components of the Helicobacter pylori infection-related metabolic syndrome present an important risk factor in the development of arterial hypertension. The triad of Helicobacter pylori infection, the metabolic syndrome, and hypertension represents a crucial worldwide health problem on a pandemic scale with high morbidity and mortality, like COVID-19, thereby requiring awareness and appropriate management on a global scale.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- Correspondence:
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- Department of Gastroenterology, University Hospital of Larisa, 41110 Larisa, Greece
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Georgia Deretzi
- Multiple Sclerosis Unit, Department of Neurology, Papageorgiou General Hospital, 56403 Thessaloniki, Greece;
| | - Elisabeth Vardaka
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece;
| | - Elpidoforos S. Soteriades
- Healthcare Management Program, School of Economics and Management, Open University of Cyprus, Nicosia 2252, Cyprus;
- Department of Environmental Health, Environmental and Occupational Medicine and Epidemiology (EOME), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Paraskevas Gkolfakis
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme University Hospital, 1070 Brussels, Belgium;
- Department of Medical Oncology, Institut Jules Bordet, 1000 Brussels, Belgium
| | - Kyriaki Karafyllidou
- Department of Pediatrics, University Children’s Hospital of Zurich, 8032 Zurich, Switzerland;
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
| |
Collapse
|
10
|
Eligini S, Colli S, Habib A, Aldini G, Altomare A, Banfi C. Cyclooxygenase-2 Glycosylation Is Affected by Peroxynitrite in Endothelial Cells: Impact on Enzyme Activity and Degradation. Antioxidants (Basel) 2021; 10:496. [PMID: 33806920 PMCID: PMC8005028 DOI: 10.3390/antiox10030496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
The exposure of human endothelial cells to 3-morpholinosydnonimine (SIN-1) induced the expression of cyclooxygenase-2 (COX-2) in a dose- and time-dependent manner. Interestingly, after a prolonged incubation (>8 h) several proteoforms were visualized by Western blot, corresponding to different states of glycosylation of the protein. This effect was specific for SIN-1 that generates peroxynitrite and it was not detected with other nitric oxide-donors. Metabolic labeling experiments using 35S or cycloheximide suggested that the formation of hypoglycosylated COX-2 was dependent on de novo synthesis of the protein rather than the deglycosylation of the native protein. Moreover, SIN-1 reduced the activity of the hexokinase, the enzyme responsible for the first step of glycolysis. The hypoglycosylated COX-2 induced by SIN-1 showed a reduced capacity to generate prostaglandins and the activity was only partially recovered after immunoprecipitation. Finally, hypoglycosylated COX-2 showed a more rapid rate of degradation compared to COX-2 induced by IL-1α and an alteration in the localization with an accumulation mainly detected in the nuclear membrane. Our results have important implication to understand the effect of peroxynitrite on COX-2 expression and activity, and they may help to identify new pharmacological tools direct to increase COX-2 degradation or to inhibit its activity.
Collapse
Affiliation(s)
- Sonia Eligini
- Centro Cardiologico Monzino I.R.C.C.S., 20138 Milan, Italy;
| | - Susanna Colli
- Dipartimento di Scienze Farmacologiche, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Aida Habib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
- INSERM-UMR1149, Centre de Recherche sur l’Inflammation, and Sorbonne Paris Cité, Laboratoire d’Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Université de Paris, 75018 Paris, France
| | - Giancarlo Aldini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, 20133 Milano, Italy; (G.A.); (A.A.)
| | - Alessandra Altomare
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, 20133 Milano, Italy; (G.A.); (A.A.)
| | - Cristina Banfi
- Centro Cardiologico Monzino I.R.C.C.S., 20138 Milan, Italy;
| |
Collapse
|
11
|
Camelo-Castillo A, Rivera-Caravaca JM, Orenes-Piñero E, Ramírez-Macías I, Roldán V, Lip GYH, Marín F. Gut Microbiota and the Quality of Oral Anticoagulation in Vitamin K Antagonists Users: A Review of Potential Implications. J Clin Med 2021; 10:715. [PMID: 33670220 PMCID: PMC7916955 DOI: 10.3390/jcm10040715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/08/2021] [Indexed: 12/31/2022] Open
Abstract
The efficacy and safety of vitamin K antagonists (VKAs) as oral anticoagulants (OACs) depend on the quality of anticoagulation control, as reflected by the mean time in therapeutic range (TTR). Several factors may be involved in poor TTR such as comorbidities, high inter-individual variability, interacting drugs, and non-adherence. Recent studies suggest that gut microbiota (GM) plays an important role in the pathogenesis of cardiovascular diseases, but the effect of the GM on anticoagulation control with VKAs is unknown. In the present review article, we propose different mechanisms by which the GM could have an impact on the quality of anticoagulation control in patients taking VKA therapy. We suggest that the potential effects of GM may be mediated first, by an indirect effect of metabolites produced by GM in the availability of VKAs drugs; second, by an effect of vitamin K-producing bacteria; and finally, by the structural modification of the molecules of VKAs. Future research will help confirm these hypotheses and may suggest profiles of bacterial signatures or microbial metabolites, to be used as biomarkers to predict the quality of anticoagulation. This could lead to the design of intervention strategies modulating gut microbiota, for example, by using probiotics.
Collapse
Affiliation(s)
- Anny Camelo-Castillo
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, 30120 Murcia, Spain; (A.C.-C.); (J.M.R.-C.); (I.R.-M.)
| | - José Miguel Rivera-Caravaca
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, 30120 Murcia, Spain; (A.C.-C.); (J.M.R.-C.); (I.R.-M.)
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool L7 8TX, UK;
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, 30120 Murcia, Spain;
| | - Inmaculada Ramírez-Macías
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, 30120 Murcia, Spain; (A.C.-C.); (J.M.R.-C.); (I.R.-M.)
| | - Vanessa Roldán
- Department of Hematology and Clinical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, 30008 Murcia, Spain;
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool L7 8TX, UK;
- Department of Clinical Medicine, Aalborg Thrombosis Research Unit, Aalborg University, 9000 Aalborg, Denmark
| | - Francisco Marín
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, 30120 Murcia, Spain; (A.C.-C.); (J.M.R.-C.); (I.R.-M.)
| |
Collapse
|
12
|
Discovery of novel sulfonamide-containing aminophosphonate derivatives as selective COX-2 inhibitors and anti-tumor candidates. Bioorg Chem 2020; 105:104390. [DOI: 10.1016/j.bioorg.2020.104390] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
|
13
|
Saadati S, Eduok U, Abdelrasoul A, Shoker A. A surface-enhanced Raman scattering-based approach for rapid and highly sensitive quantitative analysis of 3-carboxy-4-methyl-5-propyl-2-furanpropionate and indole-3-acetic acid in saline, human serum and uremic serum of patients with chronic kidney disease. RSC Adv 2020; 10:43489-43496. [PMID: 35519726 PMCID: PMC9058093 DOI: 10.1039/d0ra06123a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
3-Carboxy-4-methyl-5-propyl-2-furanpropionate (CMPF) and indole-3-acetic acid (IAA) are critical protein-bound uremic toxins that occur during chronic kidney disease (CKD). This study offers the first reported instance of surface-enhanced Raman scattering (SERS) coupled with an Au nanoparticle substrate for the simple quantification of CMPF and IAA in human serum samples. The detection limits of the CMPF and IAA analysis were estimated to be 0.04 nM (S/N = 3) and 0.05 μM (S/N = 3), respectively. The results demonstrate that the SERS technique is fast-acting and highly sensitive when it comes to the simultaneous and individual quantitative detection of CMPF and IAA in biological samples. We believe that this analytical tool could serve as a very useful method for practical applications during the analysis of CMPF and IAA in the serum and urine of patients at all stages of CKD and of healthy volunteers as well as in various reservoirs.
Collapse
Affiliation(s)
- Shaghayegh Saadati
- Department of Chemical and Biological Engineering, University of Saskatchewan 57 Campus Drive Saskatoon Saskatchewan S7N 5A9 Canada +306 966 4777 +306 966 2946
- Division of Biomedical Engineering, University of Saskatchewan 57 Campus Drive Saskatoon Saskatchewan S7N 5A9 Canada
| | - Ubong Eduok
- Department of Chemical and Biological Engineering, University of Saskatchewan 57 Campus Drive Saskatoon Saskatchewan S7N 5A9 Canada +306 966 4777 +306 966 2946
| | - Amira Abdelrasoul
- Department of Chemical and Biological Engineering, University of Saskatchewan 57 Campus Drive Saskatoon Saskatchewan S7N 5A9 Canada +306 966 4777 +306 966 2946
- Division of Biomedical Engineering, University of Saskatchewan 57 Campus Drive Saskatoon Saskatchewan S7N 5A9 Canada
| | - Ahmed Shoker
- Nephrology Division, College of Medicine, University of Saskatchewan 107 Wiggins Rd Saskatoon SK S7N 5E5 Canada
- Saskatchewan Transplant Programn, St. Paul's Hospital 1702 20th Street West Saskatoon Saskatchewan S7M 0Z9 Canada
| |
Collapse
|
14
|
Ledard N, Liboz A, Blondeau B, Babiak M, Moulin C, Vallin B, Guillas I, Mateo V, Jumeau C, Blirando K, Meilhac O, Limon I, Glorian M. Slug, a Cancer-Related Transcription Factor, is Involved in Vascular Smooth Muscle Cell Transdifferentiation Induced by Platelet-Derived Growth Factor-BB During Atherosclerosis. J Am Heart Assoc 2020; 9:e014276. [PMID: 31959031 PMCID: PMC7033846 DOI: 10.1161/jaha.119.014276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Heart attacks and stroke often result from occlusive thrombi following the rupture of vulnerable atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) play a pivotal role in plaque vulnerability because of their switch towards a proinflammatory/macrophage-like phenotype when in the context of atherosclerosis. The prometastatic transcription factor Slug/Snail2 is a critical regulator of cell phenotypic transition. Here, we aimed to investigate the role of Slug in the transdifferentiation process of VSMCs occurring during atherogenesis. Methods and Results In rat and human primary aortic smooth muscle cells, Slug protein expression is strongly and rapidly increased by platelet-derived growth factor-BB (PDGF-BB). PDGF-BB increases Slug protein without affecting mRNA levels indicating that this growth factor stabilizes Slug protein. Immunocytochemistry and subcellular fractionation experiments reveal that PDGF-BB triggers a rapid accumulation of Slug in VSMC nuclei. Using pharmacological tools, we show that the PDGF-BB-dependent mechanism of Slug stabilization in VSMCs involves the extracellular signal-regulated kinase 1/2 pathway. Immunohistochemistry experiments on type V and type VI atherosclerotic lesions of human carotids show smooth muscle-specific myosin heavy chain-/Slug-positive cells surrounding the prothrombotic lipid core. In VSMCs, Slug siRNAs inhibit prostaglandin E2 secretion and prevent the inhibition of cholesterol efflux gene expression mediated by PDGF-BB, known to be involved in plaque vulnerability and/or thrombogenicity. Conclusions Our results highlight, for the first time, a role of Slug in aortic smooth muscle cell transdifferentiation and enable us to consider Slug as an actor playing a role in the atherosclerotic plaque progression towards a life-threatening phenotype. This also argues for common features between acute cardiovascular events and cancer.
Collapse
Affiliation(s)
- Nahéma Ledard
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Alexandrine Liboz
- INSERM Saint-Antoine Research Center Sorbonne Université Paris France
| | - Bertrand Blondeau
- INSERM Saint-Antoine Research Center Sorbonne Université Paris France
| | - Mégane Babiak
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Célia Moulin
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Benjamin Vallin
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Isabelle Guillas
- National Institute for Health and Medical Research (INSERM) Faculté de Médecine Pitié Salpétrière UMR-S 1166 ICAN Sorbonne Université Paris France
| | - Véronique Mateo
- CIMI-Paris INSERM U1135 Faculté de Médecine Sorbonne-Université Site Pitié-Salpêtrière Sorbonne Université Paris France
| | | | - Karl Blirando
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Olivier Meilhac
- Université de La Réunion Diabète, Athérothrombose, Thérapies, Réunion, Océan Indien (UMR DéTROI U1188) - -CYROI- Sainte Clotilde La Réunion
| | - Isabelle Limon
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| | - Martine Glorian
- Institut de Biologie Paris-Seine (IBPS) Biological Adaptation and Ageing UMR 8256 Sorbonne Université Paris France
| |
Collapse
|
15
|
Taghizadeh E, Taheri F, Renani PG, Reiner Ž, Navashenaq JG, Sahebkar A. Macrophage: A Key Therapeutic Target in Atherosclerosis? Curr Pharm Des 2019; 25:3165-3174. [DOI: 10.2174/1381612825666190830153056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022]
Abstract
Background:
Atherosclerosis is a chronic inflammatory disease and a leading cause of coronary artery
disease, peripheral vascular disease and stroke. Lipid-laden macrophages are derived from circulating monocytes
and form fatty streaks as the first step of atherogenesis.
Methods:
An electronic search in major databases was performed to review new therapeutic opportunities for
influencing the inflammatory component of atherosclerosis based on monocytes/macrophages targeting.
Results:
In the past two decades, macrophages have been recognized as the main players in atherogenesis but also
in its thrombotic complications. There is a growing interest in immunometabolism and recent studies on metabolism
of macrophages have created new therapeutic options to treat atherosclerosis. Targeting recruitment, polarization,
cytokine profile extracellular matrix remodeling, cholesterol metabolism, oxidative stress, inflammatory
activity and non-coding RNAs of monocyte/macrophage have been proposed as potential therapeutic approaches
against atherosclerosis.
Conclusion:
Monocytes/macrophages have a crucial role in progression and pathogenesis of atherosclerosis.
Therefore, targeting monocyte/macrophage therapy in order to achieve anti-inflammatory effects might be a good
option for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Forough Taheri
- Sharekord Branch, Islamic Azad University, Sharekord, Iran
| | | | - Željko Reiner
- University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Department of Internal Medicine, Zagreb, Croatia
| | - Jamshid G. Navashenaq
- Immunogenetic and Cell Culture Department, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
16
|
Screening cyclooxygenase-2 inhibitors from Andrographis paniculata to treat inflammation based on bio-affinity ultrafiltration coupled with UPLC-Q-TOF-MS. Fitoterapia 2019; 137:104259. [DOI: 10.1016/j.fitote.2019.104259] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 11/19/2022]
|
17
|
Grimm H, Kretzschmar J, Cook MD, Brown MD. The Effects of Exercise, Aspirin, and Celecoxib in an Atherogenic Environment. Med Sci Sports Exerc 2019; 50:2033-2039. [PMID: 29771821 DOI: 10.1249/mss.0000000000001657] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Optimal vascular function is a hallmark of cardiovascular health. Specifically, the balance of vasoconstricting and vasodilating substances is recognized as a marker of vascular health. One of the greatest challenges to vascular health and vasodilatory balance is tumor necrosis factor alpha (TNFα)-mediated inflammation. Uncovering effective strategies that maintain a vascular environment that is more vasodilatory and antithrombotic in the face of an inflammatory challenge is favorable. PURPOSE To test the ability of various antithrombotic and provasodilatory treatments, as well as combinations thereof, to prevent unfavorable changes in markers of endothelial dysfunction in human umbilical vein endothelial cells when presented with an inflammatory challenge. METHODS Human umbilical vein endothelial cells were pretreated with exercise-like levels of laminar shear stress (LSS), aspirin, celecoxib, and their combination before a TNFα challenge. Western blot analysis as well as colorimetric assays were used to determine levels of endothelial nitric oxide synthase (eNOS) and prostacyclin (6-keto PGF1α)/thromboxane (TXB2) metabolite ratio, respectively. RESULTS Neither aspirin nor celecoxib were effective in preventing TNFα-induced reduction in eNOS. Further, aspirin was unable to maintain baseline levels of prostacyclin/thromboxane ratio in the face of the inflammatory challenge. Laminar shear stress, aspirin/LSS combination, and celecoxib/LSS combination were all able to prevent TNFα-induced alterations in eNOS levels and prostacyclin/thromboxane ratio. CONCLUSIONS Effective strategies to maintain a healthy endothelium, and therefore resistance vessel health, need to include exercise-levels of shear stress to be effective.
Collapse
Affiliation(s)
- Heather Grimm
- Kings College, Department of Sports Medicine, Wilke-Barre, PA
| | - Jan Kretzschmar
- Kings College, Department of Sports Medicine, Wilke-Barre, PA
| | - Marc D Cook
- North Carolina A&T State University, Department of Human Performance and Leisure Studies, Greensboro, NC
| | | |
Collapse
|
18
|
Gargiulo S, Rossin D, Testa G, Gamba P, Staurenghi E, Biasi F, Poli G, Leonarduzzi G. Up-regulation of COX-2 and mPGES-1 by 27-hydroxycholesterol and 4-hydroxynonenal: A crucial role in atherosclerotic plaque instability. Free Radic Biol Med 2018; 129:354-363. [PMID: 30312760 DOI: 10.1016/j.freeradbiomed.2018.09.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/26/2018] [Accepted: 09/29/2018] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is currently understood to be mainly the consequence of a complicated inflammatory process at the different stages of plaque development. Among the several inflammatory molecules involved, up-regulation of the functional cyclooxygenase 2/membrane-bound prostaglandin E synthase 1 (COX-2/mPGES-1) axis plays a key role in plaque development. Excessive production of oxidized lipids, following low-density lipoprotein (LDL) oxidation, is a characteristic feature of atherosclerosis. Among the oxidized lipids of LDLs, the oxysterol 27-hydroxycholesterol (27-OH) and the aldehyde 4-hydroxynonenal (HNE) substantially accumulate in the atherosclerotic plaque, contributing to its progression and instability through a variety of processes. This study shows that 27-OH and HNE promote up-regulation of both the inducible enzymes COX-2 and mPGES-1, leading to increased production of prostaglandin (PG) E2 and inducible nitric oxide synthase, and the subsequent release of nitric oxide in human promonocytic U937 cells. The study also examined the potential involvement of the functionally coupled COX-2/mPGES-1 in enhancing the production of certain pro-inflammatory cytokines and of matrix metalloproteinase 9 by U937 cells. This enhancement is presumably due to the induction of PGE2 synthesis, as a result of the up-regulation of the COX-2/mPGES-1, stimulated by the two oxidized lipids, 27-OH and HNE. Induction of PGE2 synthesis might thus be a mechanism of plaque instability and eventual rupture, contributing to matrix metalloproteinase production by activated macrophages.
Collapse
Affiliation(s)
- Simona Gargiulo
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy.
| |
Collapse
|
19
|
Prabhakaran J, Underwood M, Zanderigo F, Simpson NR, Cooper AR, Matthew J, Rubin-Falcone H, Parsey RV, Mann JJ, Dileep Kumar JS. Radiosynthesis and in vivo evaluation of [ 11C]MOV as a PET imaging agent for COX-2. Bioorg Med Chem Lett 2018; 28:2432-2435. [PMID: 29929881 DOI: 10.1016/j.bmcl.2018.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/08/2018] [Accepted: 06/09/2018] [Indexed: 12/15/2022]
Abstract
Radiosynthesis and in vivo evaluation of [11C]4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide (methoxy analogue of valdecoxib, [11C]MOV), a COX-2 inhibitor, was conducted in rat and baboon. Synthesis of the reference standard MOV (3), and its desmethyl precursor 2 for radiolabeling were performed using 1,2-diphenylethan-1-one as the starting material in five steps with 15% overall yield. Radiosynthesis of [11C]MOV was accomplished in 40 ± 10% yield and >99% radiochemical purity by reacting the precursor 2 in dimethyl formamide (DMF) with [11C]CH3I followed by removal of the dimethoxytrityl (DMT) protective group using trifluroacetic acid. PET studies in anesthetized baboon showed very low uptake and homogeneous distribution of [11C]MOV in brain. The radioligand underwent rapid metabolism in baboon plasma. MicroPET studies in male Sprague Dawley rats revealed [11C]MOV binding in lower thorax. The tracer binding in rats was partially blocked in heart and duodenum by the administration of 1 mg/kg oral dose of COX-2 inhibitor valdecoxib.
Collapse
Affiliation(s)
- Jaya Prabhakaran
- Department of Psychiatry, Columbia University Medical Center, New York, USA; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - Mark Underwood
- Department of Psychiatry, Columbia University Medical Center, New York, USA; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University Medical Center, New York, USA; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - Norman R Simpson
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - Anna R Cooper
- Department of Psychiatry, Columbia University Medical Center, New York, USA
| | - Jeffrey Matthew
- Department of Psychiatry, Columbia University Medical Center, New York, USA
| | - Harry Rubin-Falcone
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook Medical Center, Stony Brook, New York, USA
| | - J John Mann
- Department of Psychiatry, Columbia University Medical Center, New York, USA; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - J S Dileep Kumar
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA.
| |
Collapse
|
20
|
Liu J, Liu J, Shen F, Qin Z, Jiang M, Zhu J, Wang Z, Zhou J, Fu Y, Chen X, Huang C, Xiao W, Zheng C, Wang Y. Systems pharmacology analysis of synergy of TCM: an example using saffron formula. Sci Rep 2018; 8:380. [PMID: 29321678 PMCID: PMC5762866 DOI: 10.1038/s41598-017-18764-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/17/2017] [Indexed: 01/13/2023] Open
Abstract
Traditional Chinese medicine (TCM) follows the principle of formulae, in which the pharmacological activity of a single herb can be enhanced or potentiated by addition of other herbs. Nevertheless, the involved synergy mechanisms in formulae remain unknown. Here, a systems-based method is proposed and applied to three representative Chinese medicines in compound saffron formula (CSF): two animal spices (Moschus, Beaver Castoreum), and one herb Crocus sativus which exert synergistic effects for cardiovascular diseases (CVDs). From the formula, 42 ingredients and 66 corresponding targets are acquired based on the ADME evaluation and target fishing model. The network relationships between the compounds and targets are assembled with CVDs pathways to elucidate the synergistic therapeutic effects between the spices and the herbs. The results show that different compounds of the three medicines show similar curative activity in CVDs. Additionally, the active compounds from them shared CVDs-relevant targets (multiple compounds-one target), or functional diversity targets but with clinical relevance (multiple compounds-multiple targets-one disease). Moreover, the targets of them are largely enriched in the same CVDs pathways (multiple targets-one pathway). These results elucidate why animal spices and herbs can have pharmacologically synergistic effects on CVDs, which provides a new way for drug discovery.
Collapse
Affiliation(s)
- Jianling Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Jingjing Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Fengxia Shen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Zonghui Qin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Meng Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Jinglin Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Zhenzhong Wang
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical Co. Ltd., Lianyungang, China
| | - Jun Zhou
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical Co. Ltd., Lianyungang, China
| | - Yingxue Fu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, China
| | - Xuetong Chen
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, China
| | - Chao Huang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical Co. Ltd., Lianyungang, China.
| | - Chunli Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Yonghua Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
21
|
Mercury exposure induces proinflammatory enzymes in vascular fibroblasts. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2017; 29:231-238. [DOI: 10.1016/j.arteri.2017.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/23/2022]
|
22
|
Li J, Wang W, Han L, Feng M, Lu H, Yang L, Hu X, Shi S, Jiang S, Wang Q, Ye L. Human apolipoprotein A-I exerts a prophylactic effect on high-fat diet-induced atherosclerosis via inflammation inhibition in a rabbit model. Acta Biochim Biophys Sin (Shanghai) 2017; 49:149-158. [PMID: 28069582 DOI: 10.1093/abbs/gmw128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/29/2016] [Indexed: 01/15/2023] Open
Abstract
Apolipoprotein A-I (apoA-I) is the major functional protein fraction of high-density lipoprotein. The prophylactic effect and mechanism of human apoA-I on atherosclerosis (AS) were investigated in a high-fat diet-induced AS rabbit model. The rabbits were injected with apoA-I once a week while fed high-fat diet for 20 weeks. Our results showed that apoA-I could raise the serum level of high-density lipoprotein-cholesterol and reduce those of lipid total cholesterol, triglyceride, and low-density lipoprotein-cholesterol in AS rabbits. Decreased aortic plaque area and aortic injury degree were also observed by Oil Red O staining and HE staining in apoA-I-treated high-fat diet-induced AS rabbits. Further study elucidated that apoA-I could down-regulate the expression of some inflammatory mediators including intercellular adhesion molecule type 1, vascular adhesion molecule-1 (VCAM-1), monocyte chemoattractant protein-1, tumor necrosis factor-α, interleukin-6 (IL-6), and C-reactive protein in serum and aorta of AS rabbits. In addition, real-time quantitative RT-PCR analyses showed that the apoA-I infusions decreased the mRNA levels of two pro-inflammatory molecules, i.e. nuclear factor kappa B (NF-κB) and cyclooxygenase-2 (COX-2), in aorta of AS rabbits, which was associated with a concomitant reduction in endothelial VCAM-1 and IL-6 mRNA transcription. Together, our results support the atheroprotective and prophylactic role of apoA-I in vivo, and this activity may be correlated with its anti-inflammatory effect.
Collapse
Affiliation(s)
- Jiyang Li
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weina Wang
- Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Lei Han
- Shanghai Benemae Pharmaceutical Corporation, Shanghai International Medical Park, Shanghai 201321, China
| | - Meiqing Feng
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Hui Lu
- Shanghai RAAS Blood Products Co., Ltd, Shanghai 201401, China
| | - Li Yang
- Shanghai RAAS Blood Products Co., Ltd, Shanghai 201401, China
| | - Xiangxiang Hu
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Si Shi
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shanshan Jiang
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qian Wang
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Li Ye
- Department of Biosynthesis & Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
23
|
Pietzsch J, Laube M, Bechmann N, Pietzsch FJ, Kniess T. Protective effects of 2,3-diaryl-substituted indole-based cyclooxygenase-2 inhibitors on oxidative modification of human low density lipoproteins in vitro. Clin Hemorheol Microcirc 2017; 61:615-32. [PMID: 25547413 DOI: 10.3233/ch-141923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
It has been suggested that 2,3-diaryl-substituted indole-based cyclooxygenase-2 (COX-2) inhibitors (2,3-diaryl-indole coxibs) do not only appear as potent anti-inflammatory agents but also show the ability to scavenge reactive oxygen species (ROS). This led to the hypothesis that 2,3-diaryl-indole coxibs also may act as potent inhibitors of oxidative modification of low-density lipoprotein (LDL), which is considered a key factor in atherogenesis. The aim of this study was to explore i) the reactivity of a series of new synthesized 2,3-diaryl-indoles with several well characterized LDL oxidation systems and ii) subsequent effects on an inflammatory/atherogenic microenvironment. The results demonstrate that under the present experimental conditions 2,3-diaryl-indoles showed potent ROS scavenging activity and were able to markedly inhibit LDL oxidation. Subsequently, this led to a substantial decrease of modified LDL uptake by scavenger receptors in THP-1 macrophages in vitro and in rats in vivo. Moreover, modified LDL-mediated monocyte/neutrophil adhesion to endothelial cells, macrophage NFκB activation, as well as macrophage and endothelial cell cytokine release was diminished in vitro. The reduction of modified LDL-induced atherogenic effects by antioxidant 2,3-diaryl-indole coxibs may widen the therapeutic window of COX-2 targeted treatment.
Collapse
Affiliation(s)
- Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Dresden, Germany.,Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Dresden, Germany.,Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
| | - Nicole Bechmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Dresden, Germany.,Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
| | - Franz-Jacob Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Dresden, Germany.,Technische Universität Dresden, Medical Faculty and University Hospital, Centre for Translational Bone, Joint, and Soft Tissue Research, Dresden, Germany
| | - Torsten Kniess
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Dresden, Germany
| |
Collapse
|
24
|
Costa G, Garabito M, Jiménez-Altayó F, Onetti Y, Sabate M, Vila E, Dantas AP. Sex differences in angiotensin II responses contribute to a differential regulation of cox-mediated vascular dysfunction during aging. Exp Gerontol 2016; 85:71-80. [DOI: 10.1016/j.exger.2016.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/22/2016] [Accepted: 09/28/2016] [Indexed: 12/01/2022]
|
25
|
Jitprasertwong P, Charadram N, Kumphune S, Pongcharoen S, Sirisinha S. Female sex hormones modulate Porphyromonas gingivalis lipopolysaccharide-induced Toll-like receptor signaling in primary human monocytes. J Periodontal Res 2016; 51:395-406. [PMID: 26364725 DOI: 10.1111/jre.12320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND OBJECTIVE Female sex hormones are elevated and are potential host response modifiers during pregnancy. Modulation of immune responses by estrogen and progesterone may be responsible for periodontal inflammation. Therefore, we aimed to investigate the role of β-estradiol and progesterone in human monocyte immune responses, at cellular and molecular levels, to identify their role as a possible immunological link between pregnancy and periodontal disease. MATERIAL AND METHODS Primary human monocytes were purified from human peripheral blood mononuclear cells by adherent method. Expression of Toll-like receptor (TLR) 2, 4 and CD14 was analyzed by flow cytometry. TLR2, TLR4, cyclooxygenase-2 (COX2), nuclear factor-kappa B (NF-κB) and NF-κB inhibitor-alpha mRNA expressions were measured using real-time reverse transcriptase-polymerase chain reaction and prostaglandin E2 secretion was assayed by enzyme-linked immunosorbent assay. NF-κB expression was also examined by immunofluorescence. Western blotting was performed to determine the activation of mitogen-activated protein kinase pathway. RESULTS We report herein that both β-estradiol and progesterone significantly reduced TLR2 expression at both protein and mRNA levels but had less of an effect on TLR4 expression in primary human monocytes. We also found that the hormones decreased monocyte cell surface protein expression of CD14. Significantly, β-estradiol and progesterone dose-dependently downregulated monocyte expression of COX2 mRNA. Pretreatment monocytes with β-estradiol or progesterone reduced effects of Porphyromonas gingivalis lipopolysaccharide (LPS) on COX2 mRNA expression and decreased prostaglandin E2 secretion by the monocytes. Furthermore, we demonstrated that both β-estradiol and progesterone inhibited P. gingivalis LPS-induced NF-κB signaling pathway through the upregulation of NF-κB inhibitor-alpha expression. However, neither β-estradiol nor progesterone altered the phosphorylation of the p38, the extracellular signal-regulated kinase 1/2 and the c-Jun N-terminal activated kinase in P. gingivalis LPS-stimulated monocytes. Thus, the inhibitory effects of these hormones on the response of human monocytes to P. gingivalis LPS appear to be independent on mitogen-activated protein kinase signaling pathway. CONCLUSION The results of the present study suggest that β-estradiol and progesterone could influence the immune response of human monocytes to periodontal pathogens and this process may have a role in the clinical manifestations of periodontal disease associated with pregnancy.
Collapse
Affiliation(s)
- P Jitprasertwong
- Department of Preventive Dentistry, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - N Charadram
- Department of Restorative Dentistry, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - S Kumphune
- Faculty of Allied Health Science, Biomedical Research Unit in Cardiovascular Sciences, Naresuan University, Phitsanulok, Thailand
| | - S Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology (CEMB), Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - S Sirisinha
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
26
|
Avendaño MS, Martínez-Revelles S, Aguado A, Simões MR, González-Amor M, Palacios R, Guillem-Llobat P, Vassallo DV, Vila L, García-Puig J, Beltrán LM, Alonso MJ, Cachofeiro MV, Salaices M, Briones AM. Role of COX-2-derived PGE2 on vascular stiffness and function in hypertension. Br J Pharmacol 2016; 173:1541-55. [PMID: 26856544 DOI: 10.1111/bph.13457] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 01/08/2016] [Accepted: 01/29/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Prostanoids derived from COX-2 and EP receptors are involved in vascular remodelling in different cardiovascular pathologies. This study evaluates the contribution of COX-2 and EP1 receptors to vascular remodelling and function in hypertension. EXPERIMENTAL APPROACH Spontaneously hypertensive rats (SHR) and angiotensin II (AngII)-infused (1.44 mg · kg(-1) · day(-1), 2 weeks) mice were treated with the COX-2 inhibitor celecoxib (25 mg · kg(-1) · day(-1) i.p) or with the EP1 receptor antagonist SC19220 (10 mg · kg(-1) · day(-1) i.p.). COX-2(-/-) mice with or without AngII infusion were also used. KEY RESULTS Celecoxib and SC19220 treatment did not modify the altered lumen diameter and wall : lumen ratio in mesenteric resistance arteries from SHR-infused and/or AngII-infused animals. However, both treatments and COX-2 deficiency decreased the augmented vascular stiffness in vessels from hypertensive animals. This was accompanied by diminished vascular collagen deposition, normalization of altered elastin structure and decreased connective tissue growth factor and plasminogen activator inhibitor-1 gene expression. COX-2 deficiency and SC19220 treatment diminished the increased vasoconstrictor responses and endothelial dysfunction induced by AngII infusion. Hypertensive animals showed increased mPGES-1 expression and PGE2 production in vascular tissue, normalized by celecoxib. Celecoxib treatment also decreased AngII-induced macrophage infiltration and TNF-α expression. Macrophage conditioned media (MCM) increased COX-2 and collagen type I expression in vascular smooth muscle cells; the latter was reduced by celecoxib treatment. CONCLUSIONS AND IMPLICATIONS COX-2 and EP1 receptors participate in the increased extracellular matrix deposition and vascular stiffness, the impaired vascular function and inflammation in hypertension. Targeting PGE2 receptors might have benefits in hypertension-associated vascular damage.
Collapse
Affiliation(s)
- M S Avendaño
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - S Martínez-Revelles
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - A Aguado
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - M R Simões
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain.,Dept. Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - M González-Amor
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - R Palacios
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - P Guillem-Llobat
- Centro de Biología Molecular "Severo Ochoa", UAM-CSIC, Madrid, Spain
| | - D V Vassallo
- Dept. Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - L Vila
- Laboratorio de Angiología, Biología Vascular e Inflamación, Instituto de Investigación Biomédica (IIB Sant Pau), Barcelona, Spain
| | - J García-Puig
- Servicio de Medicina Interna, Hospital Universitario La Paz, UAM, IdiPaz, Madrid, Spain
| | - L M Beltrán
- Servicio de Medicina Interna, Hospital Universitario La Paz, UAM, IdiPaz, Madrid, Spain
| | - M J Alonso
- Dept Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - M V Cachofeiro
- Dept. Fisiología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - M Salaices
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - A M Briones
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
27
|
Sihra JK, Thumser AE, Langat MK, Crouch NR, Mulholland DA. Constituents of Bulbs of three Species of the Hyacinthaceae (Hyacinthoideae): Eucomis vandermerwei, E. zambesiaca and Resnova humifusa. Nat Prod Commun 2015. [DOI: 10.1177/1934578x1501000718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Phytochemical analyses of the bulbs of Eucomis vandermerwei and E. zambesiaca yielded homoisoflavonoids and triterpenoid derivatives. A new (17 S*,23 S*)-epoxy-3β,15β,29-trihydroxy-27-norlanost-8-en-24-one) was isolated from E. zambesiaca. Resnova humifusa yielded homoisoflavonoids, and a tetrahydropyran derivative, 2-methyl-3-(4 S*,5 R*,7 S*-trihydroxy-8 S*-hydroxymethyltetrahydro-6H-4-pyranyl)-2-propenoic acid. All compounds were assayed for COX-2 inhibition of cyclooxygenase; 3,5,7-trihydroxy-3-(4′-methoxybenzyl)-4-chromanone was found to have significant activity.
Collapse
Affiliation(s)
- Jaspreet K. Sihra
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Alfred E. Thumser
- Division of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Moses K. Langat
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Neil R. Crouch
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, 4041, South Africa
- Ethnobotnany Unit, National Botanical Institute, P.O. Box 52099, Berea Road, Durban, South Africa
| | - Dulcie A. Mulholland
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, 4041, South Africa
| |
Collapse
|
28
|
Amadio P, Baldassarre D, Tarantino E, Zacchi E, Gianellini S, Squellerio I, Amato M, Weksler BB, Tremoli E, Barbieri SS. Production of prostaglandin E2 induced by cigarette smoke modulates tissue factor expression and activity in endothelial cells. FASEB J 2015; 29:4001-10. [PMID: 26065856 DOI: 10.1096/fj.14-268383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 06/02/2015] [Indexed: 01/15/2023]
Abstract
Cigarette smoke (CS) increases the incidence of atherothrombosis, the release of prostaglandin (PG) E2, and the amount of tissue factor (TF). The link between PGE2 and TF, and the impact of this interaction on CS-induced thrombosis, is unknown. Plasma from active smokers showed higher concentration of PGE2, TF total antigen, and microparticle-associated TF (MP-TF) activity compared with never smokers. Similar results were obtained in mice and in mouse cardiac endothelial cells (MCECs) after treatment with aqueous CS extracts (CSEs) plus IL-1β [CSE (6.4 puffs/L)/IL-1β (2 μg/L)]. A significant correlation between PGE2 and TF total antigen or MP-TF activity were observed in both human and mouse plasma or tissue. Inhibition of PGE synthase reduced TF in vivo and in vitro and prevented the arterial thrombosis induced by CSE/IL-1β. Only PG E receptor 1 (EP1) receptor antagonists (SC51089:IC50 ∼ 1 μM, AH6809:IC50 ∼ 7.5 μM) restored the normal TF and sirtuin 1 (SIRT1) levels in MCECs before PGE2 (EC50 ∼ 2.5 mM) or CSE/IL-1β exposure. Similarly, SIRT1 activators (CAY10591: IC50 ∼ 10 μM, resveratrol: IC50 ∼ 5 μM) or prostacyclin analogs (IC50 ∼ 5 μM) prevented SIRT1 inhibition and reduced TF induced by CSE/IL-1β or by PGE2. In conclusion, PGE2 increases both TF expression and activity through the regulation of the EP1/SIRT1 pathway. These findings suggest that EP1 may represent a possible target to prevent prothrombotic states.
Collapse
Affiliation(s)
- Patrizia Amadio
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Damiano Baldassarre
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Eva Tarantino
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Elena Zacchi
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Sara Gianellini
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Isabella Squellerio
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Mauro Amato
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Babette B Weksler
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Elena Tremoli
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Silvia S Barbieri
- *Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Division of Hematology-Medical Oncology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
29
|
Kim MJ, Lim HS, Jin S, Jung JA, Noh YH, Kim YH, Bae KS. Pharmacokinetic, Pharmacodynamic, and Safety/Tolerability Profiles of CG100649, a Novel COX-2 Inhibitor: Results of a Phase I, Randomized, Multiple-dose Study in Healthy Korean Men and Women. Clin Ther 2015; 37:197-210. [DOI: 10.1016/j.clinthera.2014.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2014] [Indexed: 11/29/2022]
|
30
|
Fabule J, Adebajo A. Comparative evaluation of cardiovascular outcomes in patients with osteoarthritis and rheumatoid arthritis on recommended doses of nonsteroidal anti-inflammatory drugs. Ther Adv Musculoskelet Dis 2014; 6:111-30. [PMID: 25342992 DOI: 10.1177/1759720x14541668] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIMS AND OBJECTIVES We conducted an analysis to explore whether the cardiovascular outcomes associated with nonsteroidal anti-inflammatory drugs (NSAIDs), when used in licensed doses by patients with osteoarthritis or rheumatoid arthritis, was class or compound dependent. METHODS Using the Ovid technology search engine, we conducted a search of the literature for relevant studies published between 1995 and 2011. We also retrieved further studies following manual searches. The primary endpoint was major vascular events and the secondary endpoints were stroke, hypertension and congestive heart failure. A total of 19 studies were analysed. Studies conducted in the osteoarthritis and rheumatoid arthritis patients' population that reported on cardiovascular events were included in the analysis. The analysis was conducted using the software Review Manager 5.1 and Cochrane methodology. RESULTS Using the primary endpoint of major vascular events (MVE) and a prespecified cutoff point of 1.30, diclofenac (versus 1 comparator) and rofecoxib (versus 2 comparators) had increased risk for MVE [odds ratio (OR) >1.30]. Using the same criteria, diclofenac (versus 1 comparator) had an increased risk of myocardial infarction (MI). Although celecoxib had a slightly increased risk for MI (OR 1.33, versus 1 comparator), the confidence interval included 1 and was not significant. For the secondary endpoints, etoricoxib and rofecoxib were significantly worse off for HT (versus 1 comparator each) and naproxen was significantly worse off for stroke (versus 1 comparator). Although ibuprofen was worse off for HT (versus 1 comparator) the increased risk was not significant. CONCLUSION From the analysis conducted, it appears that the risk for cardiovascular events in arthritis patients on licensed doses of NSAIDs varies considerably and is likely to depend on the individual compound.
Collapse
Affiliation(s)
- John Fabule
- Astrazeneca - Global Medical Affairs, 2 Kingdom Street, London W2 6BD, UK
| | - Ade Adebajo
- Academic Rheumatology Group, Faculty of Medicine, University of Sheffield and Barnsley Hospital NHS Foundation Trust, Barnsley, UK
| |
Collapse
|
31
|
Dou L, Sallée M, Cerini C, Poitevin S, Gondouin B, Jourde-Chiche N, Fallague K, Brunet P, Calaf R, Dussol B, Mallet B, Dignat-George F, Burtey S. The cardiovascular effect of the uremic solute indole-3 acetic acid. J Am Soc Nephrol 2014; 26:876-87. [PMID: 25145928 DOI: 10.1681/asn.2013121283] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
In CKD, uremic solutes may induce endothelial dysfunction, inflammation, and oxidative stress, leading to increased cardiovascular risk. We investigated whether the uremic solute indole-3 acetic acid (IAA) predicts clinical outcomes in patients with CKD and has prooxidant and proinflammatory effects. We studied 120 patients with CKD. During the median study period of 966 days, 29 patients died and 35 experienced a major cardiovascular event. Kaplan-Meier analysis revealed that mortality and cardiovascular events were significantly higher in the higher IAA group (IAA>3.73 µM) than in the lower IAA group (IAA<3.73 µM). Multivariate Cox regression analysis demonstrated that serum IAA was a significant predictor of mortality and cardiovascular events after adjustments for age and sex; cholesterol, systolic BP, and smoking; C-reactive protein, phosphate, body mass index, and albumin; diastolic BP and history of cardiovascular disease; and uremic toxins p-cresyl sulfate and indoxyl sulfate. Notably, IAA level remained predictive of mortality when adjusted for CKD stage. IAA levels were positively correlated with markers of inflammation and oxidative stress: C-reactive protein and malondialdehyde, respectively. In cultured human endothelial cells, IAA activated an inflammatory nongenomic aryl hydrocarbon receptor (AhR)/p38MAPK/NF-κB pathway that induced the proinflammatory enzyme cyclooxygenase-2. Additionally, IAA increased production of endothelial reactive oxygen species. In conclusion, serum IAA may be an independent predictor of mortality and cardiovascular events in patients with CKD. In vitro, IAA induces endothelial inflammation and oxidative stress and activates an inflammatory AhR/p38MAPK/NF-κB pathway.
Collapse
Affiliation(s)
- Laetitia Dou
- Aix Marseille University, Inserm, UMR 1076, Marseille, France;
| | - Marion Sallée
- Aix Marseille University, Inserm, UMR 1076, Marseille, France; Nephrology Dialysis Renal Transplantation Center, APHM, CHU Conception, Marseille, France
| | - Claire Cerini
- Aix Marseille University, Inserm, UMR 1076, Marseille, France
| | | | - Bertrand Gondouin
- Aix Marseille University, Inserm, UMR 1076, Marseille, France; Nephrology Dialysis Renal Transplantation Center, APHM, CHU Conception, Marseille, France
| | - Noemie Jourde-Chiche
- Nephrology Dialysis Renal Transplantation Center, APHM, CHU Conception, Marseille, France
| | - Karim Fallague
- Aix Marseille University, Inserm, UMR 1076, Marseille, France
| | - Philippe Brunet
- Aix Marseille University, Inserm, UMR 1076, Marseille, France; Nephrology Dialysis Renal Transplantation Center, APHM, CHU Conception, Marseille, France
| | - Raymond Calaf
- Biochemistry Laboratory, Aix Marseille University, Marseille, France; and
| | - Bertrand Dussol
- Aix Marseille University, Inserm, UMR 1076, Marseille, France; Nephrology Dialysis Renal Transplantation Center, APHM, CHU Conception, Marseille, France
| | - Bernard Mallet
- Biochemistry Laboratory, APHM, CHU Timone, Marseille, France
| | | | - Stephane Burtey
- Aix Marseille University, Inserm, UMR 1076, Marseille, France; Nephrology Dialysis Renal Transplantation Center, APHM, CHU Conception, Marseille, France
| |
Collapse
|
32
|
Cyclooxygenase 2, toll-like receptor 4 and interleukin 1β mRNA expression in atherosclerotic plaques of type 2 diabetic patients. Inflamm Res 2014; 63:851-8. [PMID: 25095741 DOI: 10.1007/s00011-014-0759-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 07/08/2014] [Accepted: 07/15/2014] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES AND DESIGN Inflammation has a prominent role in the development of atherosclerosis. Type 2 diabetes could contribute to atherosclerosis development by promoting inflammation. This status might accelerate changes in intrinsic vascular wall cells and favor plaque formation. Cyclooxygenase 2 (COX-2) is highly expressed in atherosclerotic plaques. COX-2 gene expression is promoted through activation of toll-like receptor 4 (TLR4) and pro-inflammatory cytokine interleukin 1β (IL1-β). Aim of this study is to investigate whether expression profiles of pro-inflammatory genes such as COX-2, TLR4 and IL1-β in atherosclerotic plaques are altered in type 2 diabetes (T2D). METHODS Total RNA was isolated from plaques of atherosclerotic patients and expression of COX-2, TLR4, IL1-β analyzed using real-time PCR. Histological analysis was performed on sections of the plaque to establish the degree of instability. RESULTS Statistically significant differences in mRNA expression of COX-2 and IL1-β were found in plaques of T2D compared with non-T2D patients. A multi-variable linear regression model suggests that COX-2 mRNA expression is affected by T2D pathology and IL1-β mRNA expression in atherosclerotic plaques. CONCLUSIONS Our results support the hypothesis that T2D pathology contributes in vivo to increase the inflammatory process associated with the atherosclerotic plaque formation, as shown by an increment of COX-2 and IL1-β mRNA expression.
Collapse
|
33
|
Sheng S, Wang J, Wang L, Liu H, Li P, Liu M, Long C, Xie C, Xie X, Su W. Network pharmacology analyses of the antithrombotic pharmacological mechanism of Fufang Xueshuantong Capsule with experimental support using disseminated intravascular coagulation rats. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:735-744. [PMID: 24832112 DOI: 10.1016/j.jep.2014.04.048] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang Xueshuantong (FXST) Capsule is developed on a traditional Chinese medicine remedy, with a four-herb formula of Panax notoginseng, Radix astragali, Salvia miltiorrhizae and Radix scrophulariaceae. It has been used for treatment of the clinic cardiovascular disease for many years. MATERIALS AND METHODS Due to its complexity of compositions and polypharmacological effects, it often complicates understanding of the mechanisms of action. In the present work, we have constructed an integrated model of system pharmacology to investigate the polypharmacological mechanisms of FXST formulation for treatment of thrombosis disease. RESULTS The predicted results showed that 22 ingredients in FXST were closely associated with 41 protein targets related to blood coagulation, fibrinolysis and platelet aggregation. Through analysis of the compound-protein target association, significant cross-targets between each herb indicated the multiple active chemical ingredients might interact with the same target simultaneously and thus explained the synergistic mechanisms of the principle of Traditional Chinese medicines (TCMs) as ''Jun (emperor) - Chen (minister) - Zuo (adjuvant) - Shi (courier)''. To validate the polypharmacological effects predicted by our network pharmacology (NetPharm) analysis, we have carried out experimental investigation the effects of FXST on the disorders of the blood coagulation system in a lipopolysaccharide-induced disseminated intravascular coagulation (DIC) rat model. The results showed that FXST could significantly ameliorate the activation of coagulation system, which is congruent with the cross-target prediction by NetPharm approach. CONCLUSIONS The combined investigations provide more insight into better understanding of the pharmacological mechanisms of FXST, and may also offer an alternative avenue to further explore the chemical and pharmacological basis of TCMs.
Collapse
Affiliation(s)
- Shujing Sheng
- Guangzhou Quality R & D Center of Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China; Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan 523325, PR China
| | - Jinxu Wang
- Guangzhou Quality R & D Center of Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China; Visiting Scholar in Xie's laboratory at University of Pittsburgh, USA
| | - Lirong Wang
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hong Liu
- Guangzhou Quality R & D Center of Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Peibo Li
- Guangzhou Quality R & D Center of Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Menghua Liu
- Guangzhou Quality R & D Center of Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Chaofeng Long
- Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan 523325, PR China
| | - Chengshi Xie
- Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan 523325, PR China
| | - Xiangqun Xie
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Weiwei Su
- Guangzhou Quality R & D Center of Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China.
| |
Collapse
|
34
|
Tong X, Lv P, Mathew AV, Liu D, Niu C, Wang Y, Ji L, Li J, Fu Z, Pan B, Pennathur S, Zheng L, Huang Y. The compensatory enrichment of sphingosine -1- phosphate harbored on glycated high-density lipoprotein restores endothelial protective function in type 2 diabetes mellitus. Cardiovasc Diabetol 2014; 13:82. [PMID: 24751283 PMCID: PMC4021293 DOI: 10.1186/1475-2840-13-82] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/07/2014] [Indexed: 02/07/2023] Open
Abstract
Background Glycation of high-density lipoprotein (HDL) decreases its ability to induce cyclooxygenase-2 (COX-2) expression and prostacyclin I-2 (PGI-2) release in endothelial cells. Whether lipid content of HDL, especially sphingosine-1-phosphate (S1P), plays any specific role in restoring the protective function of HDL in type 2 diabetes mellitus (T2DM) is still unknown. Methods and results Immunochemical techniques demonstrated that glycated HDL loses its protective function of regulating COX-2 expression compared with diabetic HDL. We proved that the lipid content, especially phospholipid content differed between diabetic HDL and glycated HDL. Levels of HDL-c-bound S1P were increased in T2DM compared with control subjects as detected by UPLC-MS/MS (HDL-c-bound S1P in control subjects vs. T2DM: 309.1 ± 13.71 pmol/mg vs. 382.1 ± 24.45 pmol/mg, P < 0.05). Additionally, mRNA levels of S1P lyase enzymes and S1P phosphatase 1/2 were decreased in peripheral blood by real-time PCR. Antagonist of S1P receptor 1 and 3 (S1PR1/3) diminished the functional difference between apoHDL&PL (HDL containing the protein components and phospholipids) and diabetic apoHDL&PL (diabetic HDL containing the protein components and phospholipids). With different doses of S1P reconstituted on glycated HDL, its function in inducing the COX-2 expression was restored to the same level as diabetic HDL. The mechanism of S1P reconstituted HDL (rHDL) in the process of regulating COX-2 expression involved the phosphorylation of ERK/MAPK-CREB signal pathway. Conclusion/Significance S1P harbored on HDL is the main factor which restores its protective function in endothelial cells in T2DM. S1P and its receptors are potential therapeutic targets in ameliorating the vascular dysfunction in T2DM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Lemin Zheng
- Department of Neurology, Peking University First Hospital, Beijing 100034, China.
| | | |
Collapse
|
35
|
Li H, Gao S, Ye J, Feng X, Cai Y, Liu Z, Lu J, Li Q, Huang X, Chen S, Liu P. COX-2 is involved in ET-1-induced hypertrophy of neonatal rat cardiomyocytes: role of NFATc3. Mol Cell Endocrinol 2014; 382:998-1006. [PMID: 24291639 DOI: 10.1016/j.mce.2013.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 10/31/2013] [Accepted: 11/19/2013] [Indexed: 01/04/2023]
Abstract
Endothelin-1 (ET-1) is a critical molecule that involved in heart failure. It has been proved that ET-1 stimulation results in cardiac hypertrophy both in vitro and in vivo, but the mechanisms underlying remain largely unknown. In this study, we reported that cyclooxygenase-2 (COX-2) might be an important mediator of hypertrophic responses to ET-1 stimulation. In the cultured rat neonatal cardiomyocytes, ET-1 significantly upregulated the expression and activity of COX-2, which was accompanied by increase in cell surface area and BNP mRNA level. In contrast, ET-1-dependent cardiomyocyte hypertrophy was abolished by COX-2 selective inhibitors, NS-398 and celecoxib, or by COX-2 RNA interference, but the inhibitory effects could be diminished by pretreatment with PGE2. Furthermore, cyclosporin A (CsA) and knockdown of nuclear factor of activated T-cells c3 (NFATc3) inhibited the expression of COX-2 induced by ET-1, and NFATc3 could also bound to the -GGAAA- sequence in the promoter region of rat COX-2 gene, indicating that calcineurin/NFATc3 signaling participated in the transcriptional regulation of COX-2 following ET-1 treatment. These findings provided further insight into the roles of ET-1 in cardiac hypertrophy and suggested a potential therapeutic strategy against cardiac hypertrophy by inhibiting COX-2.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Calcineurin/genetics
- Calcineurin/metabolism
- Cardiomegaly/genetics
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Celecoxib
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Cyclosporine/pharmacology
- Dinoprostone/pharmacology
- Endothelin-1/metabolism
- Endothelin-1/pharmacology
- Gene Expression Regulation
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- NFATC Transcription Factors/genetics
- NFATC Transcription Factors/metabolism
- Natriuretic Peptide, Brain/genetics
- Natriuretic Peptide, Brain/metabolism
- Nitrobenzenes/pharmacology
- Primary Cell Culture
- Promoter Regions, Genetic
- Protein Binding
- Pyrazoles/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction
- Sulfonamides/pharmacology
- Transcription, Genetic
Collapse
Affiliation(s)
- Hong Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Si Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Jiantao Ye
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Xiaojun Feng
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Yi Cai
- Guangzhou Research Institute of Snake Venom, Guangzhou Medical College, Guangzhou 510182, Guangdong, PR China
| | - Zhiping Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Jing Lu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Qin Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Xiaoyang Huang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China
| | - Shaorui Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China.
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, Guangzhou 510006, Guangdong, PR China.
| |
Collapse
|
36
|
Waller CP, Thumser AE, Langat MK, Crouch NR, Mulholland DA. COX-2 inhibitory activity of homoisoflavanones and xanthones from the bulbs of the Southern African Ledebouria socialis and Ledebouria ovatifolia (Hyacinthaceae: Hyacinthoideae). PHYTOCHEMISTRY 2013; 95:284-290. [PMID: 23859260 DOI: 10.1016/j.phytochem.2013.06.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 06/13/2013] [Accepted: 06/17/2013] [Indexed: 06/02/2023]
Abstract
The bulbs of Ledebouria socialis (Hyacinthaceae) yielded the benzocyclobutene homoisoflavonoid, (R)-2',5-dihydroxy-3',4',7-trimethoxyspiro{2H-1-benzopyran-3-(4H)-9-bicyclo[4.2.0]octa[1,3,5]triene}-4-one, socialinone (1). Ledebouria ovatifolia yielded (2ε,3R)-2,5-dihydroxy-7-methoxyspiro[2H-1-benzopyran-3(4H), 5'(6'H)-cyclobuta[f][1,3]benzodioxol]-4-one (2) and the homoisoflavanone, (E)-3-(3',4'-dihydroxybenzylidene)-5,7-dihydroxychroman-4-one, ovatifolionone (5), the dihydrochalcone, 4,4'-dihydroxy-2',6'-dimethoxydihydrochalcone (3), and xanthone, 1,6-dihydroxy-2,3,5-trimethoxy-8-methyl-9H-xanthen-9-one (4) along with 21 known compounds. Structures were determined using spectroscopic techniques. The anti-inflammatory activities of the homoisoflavonoids and xanthones isolated were evaluated against cyclooxygenase-1 and -2 isoenzymes. (R)-3-(3',4'-Dihydroxybenzyl)-7-hydroxy-5-methoxychroman-4-one (7), (E)-3-(3',4'-dihydroxybenzylidene)-7-hydroxy-5-methoxychroman-4-one (10), 1,3,6-trihydroxy-2-methoxy-8-methylxanthen-9-one (6) and ovatifolionone acetate (5Ac) exhibited significant activity against cyclooxygenase-2 at <10μM.
Collapse
Affiliation(s)
- Catherine P Waller
- Natural Products Research Group, Department of Chemistry, University of Surrey, Guildford, GU2 7XH Surrey, UK
| | | | | | | | | |
Collapse
|
37
|
Virdis A, Bacca A, Colucci R, Duranti E, Fornai M, Materazzi G, Ippolito C, Bernardini N, Blandizzi C, Bernini G, Taddei S. Endothelial Dysfunction in Small Arteries of Essential Hypertensive Patients. Hypertension 2013; 62:337-44. [DOI: 10.1161/hypertensionaha.111.00995] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Essential hypertensive patients show a reduced nitric oxide availability secondary to oxidative stress generation in peripheral microcirculation. Cyclooxygenase (COX) contributes to reduce nitric oxide availability. We assessed the possible vascular sources of oxidative stress, including COX-1, COX-2, and nicotinamide adenine dinucleotide phosphate oxidase, as determinants of endothelial dysfunction in small arteries isolated from essential hypertensive patients or normotensive controls. Small arteries were dissected after subcutaneous fat biopsies and evaluated on a pressurized micromyograph. Endothelium-dependent vasodilation was assessed by acetylcholine, repeated under NG-nitro-
l
-arginine methyl ester, SC-560 (COX-1 inhibitor), DuP-697 (COX-2 inhibitor), ascorbic acid, or the nicotinamide adenine dinucleotide phosphate oxidase inhibitors apocynin or diphenylene iodonium. Vascular oxidative stress generation (fluorescent dihydroethidium), COX-1 and COX-2 expression (Western blot), and localization (immunohistochemistry) were also assessed. In controls, response to acetylcholine was blunted by NG-nitro-
l
-arginine methyl ester (
P
<0.001) and unmodified by SC-560, DuP-697, or ascorbic acid. In hypertensive patients, relaxation to acetylcholine was blunted, resistant to NG-nitro-
l
-arginine methyl ester or SC-560, and enhanced (
P
<0.01) by DuP-697, apocynin, or diphenylene iodonium (
P
<0.05). Furthermore, in hypertensive patients, response to acetylcholine was normalized by ascorbic acid or apocynin+DuP-697. Intravascular oxidative stress generation was enhanced in hypertensive patients, decreased (
P
<0.01) by DuP-697, partly attenuated by apocynin or diphenylene iodonium, and prevented by ascorbic acid. Enhanced COX-2 expression and localization in the vascular media of hypertensive patients were also detected. In small resistance arteries of essential hypertensive patients, COX-2 is overexpressed and reduces nitric oxide availability. COX-2 represents a major source of oxidative stress generation, whereas nicotinamide adenine dinucleotide phosphate oxidase plays a minor, but significant, role in promoting superoxide generation.
Collapse
Affiliation(s)
- Agostino Virdis
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Alessandra Bacca
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Rocchina Colucci
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Emiliano Duranti
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Matteo Fornai
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Gabriele Materazzi
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Nunzia Bernardini
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Corrado Blandizzi
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Giampaolo Bernini
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| | - Stefano Taddei
- From the Department of Clinical and Experimental Medicine (A.V., A.B., R.C., E.D., M.F., C.I., N.B., C.B., G.B., S.T.) and Department of Surgery (G.M.), University of Pisa, Pisa, Italy
| |
Collapse
|
38
|
Gomez I, Foudi N, Longrois D, Norel X. The role of prostaglandin E2 in human vascular inflammation. Prostaglandins Leukot Essent Fatty Acids 2013; 89:55-63. [PMID: 23756023 DOI: 10.1016/j.plefa.2013.04.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 10/26/2022]
Abstract
Prostaglandins (PG) are the product of a cascade of enzymes such as cyclooxygenases and PG synthases. Among PG, PGE2 is produced by 3 isoforms of PGE synthase (PGES) and through activation of its cognate receptors (EP1-4), this PG is involved in the pathophysiology of vascular diseases. Some anti-inflammatory drugs (e.g. glucocorticoids, nonsteroidal anti-inflammatory drugs) interfere with its metabolism or effects. Vascular cells can initiate many of the responses associated with inflammation. In human vascular tissue, PGE2 is involved in many physiological processes, such as increasing vascular permeability, cell proliferation, cell migration and control of vascular smooth muscle tone. PGE2 has been shown to contribute to the pathogenesis of atherosclerosis, abdominal aortic aneurysm but also in physiologic/adaptive processes such as angiogenesis. Understanding the roles of PGE2 and its cognate receptors in vascular diseases could help to identify diagnostic and prognostic biomarkers. In addition, from these recent studies new promising therapeutic approaches like mPGES-1 inhibition and/or EP4-antagonism should be investigated.
Collapse
Affiliation(s)
- I Gomez
- INSERM, U698, Paris F-75018, France; University Paris Nord, UMR-S698, Paris F-75018, France
| | | | | | | |
Collapse
|
39
|
Huang CC, McDermott MM, Liu K, Kuo CH, Wang SY, Tao H, Tseng YJ. Plasma metabolomic profiles predict near-term death among individuals with lower extremity peripheral arterial disease. J Vasc Surg 2013; 58:989-96.e1. [PMID: 23688629 DOI: 10.1016/j.jvs.2013.04.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/19/2013] [Accepted: 04/15/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Individuals with peripheral arterial disease (PAD) have a nearly two-fold increased risk of all-cause and cardiovascular disease mortality compared to those without PAD. This pilot study determined whether metabolomic profiling can accurately identify patients with PAD who are at increased risk of near-term mortality. METHODS We completed a case-control study using (1)H NMR metabolomic profiling of plasma from 20 decedents with PAD, without critical limb ischemia, who had blood drawn within 8 months prior to death (index blood draw) and within 10 to 28 months prior to death (preindex blood draw). Twenty-one PAD participants who survived more than 30 months after their index blood draw served as a control population. RESULTS Results showed distinct metabolomic patterns between preindex decedent, index decedent, and survivor samples. The major chemical signals contributing to the differential pattern (between survivors and decedents) arose from the fatty acyl chain protons of lipoproteins and the choline head group protons of phospholipids. Using the top 40 chemical signals for which the intensity was most distinct between survivor and preindex decedent samples, classification models predicted near-term all-cause death with overall accuracy of 78% (32/41), a sensitivity of 85% (17/20), and a specificity of 71% (15/21). When comparing survivor with index decedent samples, the overall classification accuracy was optimal at 83% (34/41) with a sensitivity of 80% (16/20) and a specificity of 86% (18/21), using as few as the top 10 to 20 chemical signals. CONCLUSIONS Our results suggest that metabolomic profiling of plasma may be useful for identifying PAD patients at increased risk for near-term death. Larger studies using more sensitive metabolomic techniques are needed to identify specific metabolic pathways associated with increased risk of near-term all-cause mortality among PAD patients.
Collapse
Affiliation(s)
- Chiang-Ching Huang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | | | | | | | | | | | | |
Collapse
|
40
|
Liu H, Wang J, Zhou W, Wang Y, Yang L. Systems approaches and polypharmacology for drug discovery from herbal medicines: an example using licorice. JOURNAL OF ETHNOPHARMACOLOGY 2013; 146:773-93. [PMID: 23415946 DOI: 10.1016/j.jep.2013.02.004] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 02/03/2013] [Accepted: 02/04/2013] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Licorice, one of the oldest and most popular herbal medicines in the world, has been widely used in traditional Chinese medicine as a cough reliever, anti-inflammatory, anti-anabrosis, immunomodulatory, anti-platelet, antiviral (hepatitis) and detoxifying agent. Licorice was used as an example to show drug discovery from herbal drugs using systems approaches and polypharmacology. AIM OF THE STUDY Herbal medicines are becoming more mainstream in clinical practice and show value in treating and preventing diseases. However, due to its extreme complexity both in chemical components and mechanisms of action, deep understanding of botanical drugs is still difficult. Thus, a comprehensive systems approach which could identify active ingredients and their targets in the crude drugs and more importantly, understand the biological basis for the pharmacological properties of herbal medicines is necessary. MATERIALS AND METHODS In this study, a novel systems pharmacology model that integrates oral bioavailability screening, drug-likeness evaluation, blood-brain barrier permeation, target identification and network analysis has been established to investigate the herbal medicines. RESULTS The comprehensive systems approach effectively identified 73 bioactive components from licorice and 91 potential targets for this medicinal herb. These 91 targets are closely associated with a series of diseases of respiratory system, cardiovascular system, and gastrointestinal system, etc. These targets are further mapped to drug-target and drug-target-disease networks to elucidate the mechanism of this herbal medicine. CONCLUSION This work provides a novel in silico strategy for investigation of the botanical drugs containing a huge number of components, which has been demonstrated by the well-studied licorice case. This attempt should be helpful for understanding definite mechanisms of action for herbal medicines and discovery of new drugs from plants.
Collapse
Affiliation(s)
- Hui Liu
- Bioinformatics Center, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | |
Collapse
|
41
|
Wang W, Shi X, Yuan Y, Zhu H, Zhou W, Huang H, Feng M. Inhibitory effect of apolipoprotein A-I on matrix metalloproteinase-2 expression in vivo and in vitro. Acta Biochim Biophys Sin (Shanghai) 2013; 45:194-202. [PMID: 23299078 DOI: 10.1093/abbs/gms121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In the present study, we investigated the effects of apolipoprotein A-I (apoA-I) on matrix metalloproteinase-2 (MMP-2) expression in vivo and in vitro. First, we detected the effects of apoA-I on aorta MMP-2, peroxisome proliferator-activated receptor α/γ (PPAR α/γ), cyclooxygenase-2 (COX-2), and nuclear factor kappa B (NF-κB) expressions in atherosclerotic rabbit models using immunohistochemical methods. The results showed that the expressions of MMP-2, COX-2, and NF-κB were decreased in aortas of atherosclerotic rabbits treated with apoA-I, while PPAR α/γ expression was increased. Then, we chose the important inflammation cells, macrophages to testify those effects in vitro. Macrophages were divided into six groups and treated with different concentrations of apoA-I, the mRNA expressions of MMP-2, PPAR α/γ, and COX-2 were then determined using reverse-transcription polymerase chain reaction, and protein expression of PPAR γ, NF-κB were detected by western blot analysis. The levels of MMP-2 and PPAR α in cultured supernatants were determined using enzyme-linked immunosorbent assays. Interestingly, the in vitro results were similar to the results of the in vivo study. After incubation with apoA-I for 24 h, the expressions of MMP-2, COX-2, and NF-κB were decreased, while PPAR α/γ expression was increased. In consideration of their particular roles in the process of making plaque stable in vivo and in vitro, we speculate that the inhibitory effect of apoA-I on MMP-2 expression may have a close relationship with the effects of apoA-I on PPAR α/γ, COX-2, and NF-κB expressions. Although further research is needed to clarify the underlying mechanisms of these effects, our findings provide a novel insight into the anti-atherosclerotic plaque rupture effects of apoA-I.
Collapse
Affiliation(s)
- Weina Wang
- Department of Pharmacology, Fudan University, Shanghai 201203, China
| | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Szklenar M, Kalkowski J, Stangl V, Lorenz M, Rühl R. Eicosanoids and Docosanoids in Plasma and Aorta of Healthy and Atherosclerotic Rabbits. J Vasc Res 2013; 50:372-82. [DOI: 10.1159/000350865] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/17/2013] [Indexed: 11/19/2022] Open
|
44
|
Karaoglu A, Tunc T, Aydemir G, Onguru O, Uysal B, Kul M, Aydinoz S, Oztas E, Sarici U. Role of cyclooxygenase 2 and endothelial nitric oxide synthetase in preclinical atherosclerosis. Fetal Pediatr Pathol 2012; 31:432-8. [PMID: 22443285 DOI: 10.3109/15513815.2012.659408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cyclooxygenase-2 and endothelial nitric oxide (NO) synthase enzymes may have a role in developing preclinical atherosclerosis. Designed groups were as follows: smoke exposed rats before and during pregnancy, only before pregnancy, and controls. Cross-sectional samples of abdominal aorta were examined immunohistochemically. Cyclooxygenase-2 and eNOS expression was evaluated semi-quantitatively through staining extent (focal, diffuse) and staining intensity. Diffuse COX-2 expression was detected in study groups. Endothelial NO synthase expression was diffuse in study groups. COX-2 and eNOS may contribute to the formation of preatherosclerotic lesions in offspring of rats exposed to cigarette smoke through inflammatory response.
Collapse
Affiliation(s)
- Abdulbaki Karaoglu
- Department of Pediatrics, Gulhane Military Medical Academy, Etlik, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Furuya H, Wada M, Shimizu Y, Yamada PM, Hannun YA, Obeid LM, Kawamori T. Effect of sphingosine kinase 1 inhibition on blood pressure. FASEB J 2012; 27:656-64. [PMID: 23109673 DOI: 10.1096/fj.12-219014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Accumulating evidence suggests that sphingosine kinase 1 (SphK1) plays a key role in carcinogenesis by regulating cyclooxygenase-2 (COX-2) expression. Recent clinical studies have revealed that COX-2 inhibitors cause adverse cardiovascular side effects, likely due to inhibition of prostacyclin (PGI(2)). In this work, we investigated the roles of SphK1 inhibition on blood pressure (BP). The results show that lack of SphK1 expression did not exacerbate angiotensin II (Ang II)-induced acute hypertension, whereas celecoxib, a COX-2 inhibitor, augmented and sustained higher BP in mice. Interestingly, SphK1-knockout mice inhibited prostaglandin E(2) (PGE(2)) but not PGI(2) production in response to Ang II, whereas celecoxib blocked both PGE(2) and PGI(2) production. Mechanistically, SphK1 down-regulation by siRNA in human umbilical vein endothelial cells decreased cytokine-induced PGE(2) production primarily through inhibition of microsomal PGE synthase-1 (mPGES-1), not COX-2. SphK1 down-regulation also decreased MKK6 expression, which phosphorylates and activates P38 MAPK, which, in turn, regulates early growth response-1 (Egr-1), a transcription factor of mPGES-1. Together, these data indicate that SphK1 regulates PGE(2) production by mPGES-1 expression via the p38 MAPK pathway, independent of COX-2 signaling, in endothelial cells, suggesting that SphK1 inhibition may be a promising strategy for cancer chemoprevention with lack of the adverse cardiovascular side effects associated with coxibs.
Collapse
Affiliation(s)
- Hideki Furuya
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Tarp S, Bartels EM, Bliddal H, Furst DE, Boers M, Danneskiold-Samsøe B, Rasmussen M, Christensen R. Effect of nonsteroidal antiinflammatory drugs on the C-reactive protein level in rheumatoid arthritis: A meta-analysis of randomized controlled trials. ACTA ACUST UNITED AC 2012; 64:3511-21. [DOI: 10.1002/art.34644] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
47
|
Gomes A, Couto D, Alves A, Dias I, Freitas M, Porto G, Duarte JA, Fernandes E. Trihydroxyflavones with antioxidant and anti-inflammatory efficacy. Biofactors 2012; 38:378-86. [PMID: 22806885 DOI: 10.1002/biof.1033] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/02/2012] [Indexed: 12/31/2022]
Abstract
The classical anti-inflammatory therapies are frequently ineffective and present numerous and severe side effects, especially in long term use, which requires the development of anti-inflammatory drugs with different scaffolds and mechanisms of action. Owing to the high antioxidant potential and anti-inflammatory activities already inferred for hydroxyflavones, we found it would be relevant to evaluate the anti-inflammatory potential of a series of trihydroxyflavones by testing their ability to scavenge reactive oxygen species (ROS) and reactive nitrogen species (RNS) in cells and cell-free systems and to inhibit the proinflammatory pathways mediated by the enzymes cyclooxygenase (COX) and 5-lipoxygenase (5-LOX), in which reactive species have a proven involvement. The tested trihydroxyflavones proved to be effective inhibitors of neutrophils' oxidative burst and were shown to scavenge different ROS and RNS in cell-free systems. The most active compound in the majority of the assays was 3,3',4'-trihydroxyflavone, which was somehow expected due to the presence of the ortho-dihydroxy in the B-ring, an important structural feature in terms of free radical scavenging activity. Additionally, the studied compounds were able to inhibit the production of leukotriene B(4) by 5-LOX in activated neutrophils. 3,5,7-Trihydroxyflavone was able to inhibit both COX-1 and COX-2, which makes it a dual inhibitor of COX and 5-LOX pathways and, therefore, a promising candidate for a new therapeutic option in the treatment of inflammatory processes.
Collapse
Affiliation(s)
- Ana Gomes
- REQUIMTE, Departamento de Ciências Químicas, Universidade do Porto, Porto, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Majed BH, Khalil RA. Molecular mechanisms regulating the vascular prostacyclin pathways and their adaptation during pregnancy and in the newborn. Pharmacol Rev 2012; 64:540-82. [PMID: 22679221 PMCID: PMC3400831 DOI: 10.1124/pr.111.004770] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Prostacyclin (PGI(2)) is a member of the prostanoid group of eicosanoids that regulate homeostasis, hemostasis, smooth muscle function and inflammation. Prostanoids are derived from arachidonic acid by the sequential actions of phospholipase A(2), cyclooxygenase (COX), and specific prostaglandin (PG) synthases. There are two major COX enzymes, COX1 and COX2, that differ in structure, tissue distribution, subcellular localization, and function. COX1 is largely constitutively expressed, whereas COX2 is induced at sites of inflammation and vascular injury. PGI(2) is produced by endothelial cells and influences many cardiovascular processes. PGI(2) acts mainly on the prostacyclin (IP) receptor, but because of receptor homology, PGI(2) analogs such as iloprost may act on other prostanoid receptors with variable affinities. PGI(2)/IP interaction stimulates G protein-coupled increase in cAMP and protein kinase A, resulting in decreased [Ca(2+)](i), and could also cause inhibition of Rho kinase, leading to vascular smooth muscle relaxation. In addition, PGI(2) intracrine signaling may target nuclear peroxisome proliferator-activated receptors and regulate gene transcription. PGI(2) counteracts the vasoconstrictor and platelet aggregation effects of thromboxane A(2) (TXA(2)), and both prostanoids create an important balance in cardiovascular homeostasis. The PGI(2)/TXA(2) balance is particularly critical in the regulation of maternal and fetal vascular function during pregnancy and in the newborn. A decrease in PGI(2)/TXA(2) ratio in the maternal, fetal, and neonatal circulation may contribute to preeclampsia, intrauterine growth restriction, and persistent pulmonary hypertension of the newborn (PPHN), respectively. On the other hand, increased PGI(2) activity may contribute to patent ductus arteriosus (PDA) and intraventricular hemorrhage in premature newborns. These observations have raised interest in the use of COX inhibitors and PGI(2) analogs in the management of pregnancy-associated and neonatal vascular disorders. The use of aspirin to decrease TXA(2) synthesis has shown little benefit in preeclampsia, whereas indomethacin and ibuprofen are used effectively to close PDA in the premature newborn. PGI(2) analogs have been used effectively in primary pulmonary hypertension in adults and have shown promise in PPHN. Careful examination of PGI(2) metabolism and the complex interplay with other prostanoids will help design specific modulators of the PGI(2)-dependent pathways for the management of pregnancy-related and neonatal vascular disorders.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemistry
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cytochrome P-450 Enzyme Inhibitors
- Cytochrome P-450 Enzyme System/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/metabolism
- Enzyme Inhibitors/chemistry
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Epoprostenol/analogs & derivatives
- Epoprostenol/biosynthesis
- Epoprostenol/pharmacology
- Female
- Humans
- Infant, Newborn
- Intramolecular Oxidoreductases/antagonists & inhibitors
- Intramolecular Oxidoreductases/metabolism
- Ligands
- Pregnancy
- Pregnancy Complications, Cardiovascular/enzymology
- Pregnancy Complications, Cardiovascular/metabolism
- Pregnancy Complications, Cardiovascular/prevention & control
- Prostaglandin-Endoperoxide Synthases/metabolism
- Receptors, Epoprostenol/agonists
- Receptors, Epoprostenol/antagonists & inhibitors
- Receptors, Epoprostenol/metabolism
- Signal Transduction
- Thromboxane-A Synthase/antagonists & inhibitors
- Thromboxane-A Synthase/metabolism
- Vascular Diseases/enzymology
- Vascular Diseases/metabolism
- Vascular Diseases/prevention & control
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Batoule H Majed
- Harvard Medical School, Brigham and Women's Hospital, Division of Vascular Surgery, 75 Francis St., Boston, MA 02115, USA
| | | |
Collapse
|
49
|
Shafi T, Parekh RS, Jaar BG, Plantinga LC, Oberai PC, Eckfeldt JH, Levey AS, Powe NR, Coresh J. Serum β-trace protein and risk of mortality in incident hemodialysis patients. Clin J Am Soc Nephrol 2012; 7:1435-45. [PMID: 22745274 DOI: 10.2215/cjn.02240312] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Residual kidney function in dialysis patients is associated with better survival, but there are no simple methods for its assessment. β-Trace protein is a novel endogenous filtration marker of kidney function that is not removed during hemodialysis and may serve as a marker for residual kidney function similar to serum creatinine in patients not on dialysis. The objective of this study was to determine the association of serum β-trace protein with mortality in incident hemodialysis patients. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Serum β-trace protein was measured in baseline samples from 503 participants of a national prospective cohort study of incident dialysis patients with enrollment during 1995-1998 and follow-up until 2004. Outcomes were all-cause and cardiovascular disease mortality analyzed using Cox regression adjusted for demographic, clinical, and treatment factors. RESULTS Serum β-trace protein levels were higher in individuals with no urine output compared with individuals with urine output (9.0±3.5 versus 7.6±3.1 mg/L; P<0.001). There were 321 deaths (159 deaths from cardiovascular disease) during follow-up (median=3.3 years). Higher β-trace protein levels were associated with higher risk of mortality. The adjusted hazard ratio and 95% confidence interval for all-cause mortality per doubling of serum β-trace protein was 1.36 (1.09-1.69). The adjusted hazard ratios (95% confidence intervals) for all-cause mortality in the middle and highest tertiles compared with the lowest tertile were 0.95 (0.69-1.32) and 1.72 (1.25-2.37). Similar results were noted for cardiovascular disease mortality. CONCLUSIONS The serum level of β-trace protein is an independent predictor of death and cardiovascular disease mortality in incident hemodialysis patients.
Collapse
Affiliation(s)
- Tariq Shafi
- Division of Nephrology, Johns Hopkins University School of Medicine, 301 Mason Lord Drive, Suite 2500, Baltimore, MD 21224-2780, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sepsis-associated changes of the arachidonic acid metabolism and their diagnostic potential in septic patients. Crit Care Med 2012; 40:1478-86. [PMID: 22511130 DOI: 10.1097/ccm.0b013e3182416f05] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Sepsis-associated changes of the arachidonic acid metabolism and the utility of arachidonic acid metabolites for the diagnosis of sepsis have been poorly investigated so far. Therefore, the primary objective of our study was to screen for differentially regulated arachidonic acid metabolites in septic patients using a lipopolysaccharide whole-blood model and to investigate their diagnostic potential. DESIGN Prospective, observational, single-center, clinical study. SETTING Intensive care unit at University Hospital Leipzig. PATIENTS Thirty-five patients (first cohort 25 patients, second cohort 10 patients) meeting the criteria for severe sepsis or septic shock were enrolled. Eighteen healthy volunteers (first cohort 15 subjects, second cohort 3 subjects) were enrolled as controls. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Arachidonic acid and its metabolites were investigated in supernatants of nonactivated (baseline) and lipopolysaccharide-activated heparinized whole blood of healthy subjects (n=15) and septic patients (n=25) by solid phase extraction and subsequent liquid chromatography-tandem mass spectrometry. Arachidonic acid, arachidonic acid analogues, and the cyclooxygenase-associated metabolites prostaglandin E2, 11-hydroxyeicosatetraenoic acid, and thromboxane B2 were identified as differentiating metabolites between septic patients and healthy subjects. Some of these compounds, including arachidonic acid, its analogues, and the cyclooxygenase metabolites prostaglandin E2 and thromboxane B2 differed at baseline. The inducibility of arachidonic acid and the cyclooxygenase metabolites 11-hydroxyeicosatetraenoic and prostaglandin E2 were reduced by 80% to 90% in septic patients. The degree of the inducibility was associated with severity of sepsis and clinical outcome. A reduced inducibility of COX-2 but preserved inducibility of mPGES-1 on gene expression level were confirmed in an independent cohort of septic patients (n=10) by quantitative reverse-transcription polymerase chain reaction compared to healthy controls (n=3). CONCLUSIONS Arachidonic acid metabolism is markedly affected in patients with sepsis. Our data suggest that the analysis of arachidonic acid metabolites in an in vitro whole blood activation model may be a promising approach for risk estimation in septic patients that has to be further evaluated in subsequent large-scale clinical studies.
Collapse
|