1
|
Saraf P, Bhardwaj B, Verma A, Siddiqui MA, Verma H, Kumar P, Srivastava S, Krishnamurthy S, Srikrishna S, Shrivastava SK. Design, synthesis, and evaluation of benzhydrylpiperazine-based novel dual COX-2/5-LOX inhibitors with anti-inflammatory and anti-cancer activity. RSC Med Chem 2024:d4md00471j. [PMID: 39430948 PMCID: PMC11487423 DOI: 10.1039/d4md00471j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Piperazine derivatives were screened using the ChEMBL database, paving the way for the design, synthesis, and evaluation of a novel series of dual COX-2/5-LOX inhibitors and identifying their role in mitigating cancer cell proliferation. Compound 9d with 4-Cl substitution at the terminal phenyl ring showed promising inhibition of COX-2 (IC50 = 0.25 ± 0.03 μM) and 5-LOX (IC50 = 7.87 ± 0.33 μM), outperforming the standards celecoxib (IC50 = 0.36 ± 0.023 μM) and zileuton (IC50 = 14.29 ± 0.173 μM), respectively. The two most active derivatives 9d and 9g indicated a significant anti-inflammatory response in a paw edema model by inhibiting PGE2, IL-6, and TNF-α and an increase in IL-10 concentrations. Interestingly, 9d effectively reduced pain by 55.78%, closely comparable to the 59.09% exhibited by the standard indomethacin, and was also devoid of GI, liver, kidney, and cardiac toxicity. Furthermore, 9d demonstrated anti-cancer potential against in vitro A549, COLO-205, and MIA-PA-CA-2 human cancer cell lines and an in vivo Drosophila cancer model. The pharmacokinetic investigations revealed that 9d has good oral absorption characteristics.
Collapse
Affiliation(s)
- Poorvi Saraf
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Bhagwati Bhardwaj
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Akash Verma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Mohammad Aquib Siddiqui
- Pharmacology Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India
| | - Himanshu Verma
- Pharmacology Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India
| | - Pradeep Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University Varanasi-221005 India
| | - Samridhi Srivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Sairam Krishnamurthy
- Pharmacology Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India
| | - Saripella Srikrishna
- Department of Biochemistry, Institute of Science, Banaras Hindu University Varanasi-221005 India
| | - Sushant Kumar Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| |
Collapse
|
2
|
Cao Y, Lai KM, Fu KC, Kuo CL, Tan YJ, Yu L(L, Huang D. Dual Functionality of Papaya Leaf Extracts: Anti-Coronavirus Activity and Anti-Inflammation Mechanism. Foods 2024; 13:3274. [PMID: 39456336 PMCID: PMC11506937 DOI: 10.3390/foods13203274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Papaya leaves have been used as food and traditional herbs for the treatment of cancer, diabetes, asthma, and virus infections, but the active principle has not been understood. We hypothesized that the anti-inflammatory activity could be the predominant underlying principle. To test this, we extracted papaya leaf juice with different organic solvents and found that the ethyl acetate (EA) fraction showed the most outstanding anti-inflammatory activity by suppressing the production of nitric oxide (NO, IC50 = 24.94 ± 2.4 μg/mL) and the expression of pro-inflammatory enzymes, such as inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX-2), and cytokines including interleukins (IL-1β and IL-6), and a tumor necrosis factor (TNF-α) in lipopolysaccharide (LPS)-induced RAW 264.7 cells. Transcriptomic analysis and Western blot results revealed its anti-inflammatory mechanisms were through the MAPK signaling pathway by inhibiting the phosphorylation of ERK1/2, JNKs, and p38 and the prevention of the cell surface expression of TLR4. Furthermore, we discovered that the EA fraction could inhibit the replication of alpha-coronavirus (HCoV-229E) and beta-coronavirus (HCoV-OC43 and SARS-CoV-2) and might be able to prevent cytokine storms caused by the coronavirus infection. From HPLC-QTOF-MS data, we found that the predominant phytochemicals that existed in the EA fraction were quercetin and kaempferol glycosides and carpaine. Counter-intuitively, further fractionation resulted in a loss of activity, suggesting that the synergistic effect of different components in the EA fraction contribute to the overall potent activity. Taken together, our results provide preliminary evidence for papaya leaf as a potential anti-inflammatory and anti-coronavirus agent, warranting further study for its use for human health promotion.
Collapse
Affiliation(s)
- Yujia Cao
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore;
| | - Kah-Man Lai
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; (K.-M.L.); (Y.-J.T.)
| | - Kuo-Chang Fu
- AgriGADA Biotech Pte Ltd., 8 Eu Tong Sen Street #17–82, The Central, Singapore 059818, Singapore; (K.-C.F.); (C.-L.K.)
| | - Chien-Liang Kuo
- AgriGADA Biotech Pte Ltd., 8 Eu Tong Sen Street #17–82, The Central, Singapore 059818, Singapore; (K.-C.F.); (C.-L.K.)
- Ph.D. Program for Aging, College of Medicine, China Medical University, Taichung 333, Taiwan
| | - Yee-Joo Tan
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; (K.-M.L.); (Y.-J.T.)
| | - Liangli (Lucy) Yu
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA;
| | - Dejian Huang
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore;
- Biomedical and Health Technology Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China
| |
Collapse
|
3
|
D'Orazio S, Mattoscio D. Dysregulation of the Arachidonic Acid Pathway in Cystic Fibrosis: Implications for Chronic Inflammation and Disease Progression. Pharmaceuticals (Basel) 2024; 17:1185. [PMID: 39338347 PMCID: PMC11434829 DOI: 10.3390/ph17091185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
Cystic fibrosis (CF) is the most common fatal genetic disease among Caucasian people, with over 2000 mutations in the CFTR gene. Although highly effective modulators have been developed to rescue the mutant CFTR protein, unresolved inflammation and persistent infections still threaten the lives of patients. While the central role of arachidonic acid (AA) and its metabolites in the inflammatory response is widely recognized, less is known about their impact on immunomodulation and metabolic implications in CF. To this end, here we provided a comprehensive analysis of the AA metabolism in CF. In this context, CFTR dysfunction appeared to complexly disrupt normal lipid processing, worsening the chronic airway inflammation, and compromising the immune responses to bacterial infections. As such, potential strategies targeting AA and its inflammatory mediators are being investigated as a promising approach to balance the inflammatory response while mitigating disease progression. Thus, a deeper understanding of the AA pathway dysfunction in CF may open innovative avenues for designing more effective therapeutic interventions.
Collapse
Affiliation(s)
- Simona D'Orazio
- Department of Medical, Oral and Biotechnology Sciences, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Domenico Mattoscio
- Department of Medical, Oral and Biotechnology Sciences, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
4
|
Liang S, Wang X, Zhu X. Insights from pharmacovigilance and pharmacodynamics on cardiovascular safety signals of NSAIDs. Front Pharmacol 2024; 15:1455212. [PMID: 39295938 PMCID: PMC11408209 DOI: 10.3389/fphar.2024.1455212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
Background and Aim Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used to treat fever, pain, and inflammation. Concerns regarding their cardiovascular safety have been raised. However, the underlying mechanism behind these events remains unknown. We aim to investigate the cardiovascular safety signals and receptor mechanisms of NSAIDs, employing a comprehensive approach that integrates pharmacovigilance and pharmacodynamics. Methods This study utilized a pharmacovigilance-pharmacodynamic approach to evaluate the cardiovascular safety of NSAIDs and explore potential receptor mechanisms involved. Data were analyzed using the OpenVigil 2.1 web application, which grants access to the FDA Adverse Event Reporting System (FAERS) database, in conjunction with the BindingDB database, which provides target information on the pharmacodynamic properties of NSAIDs. Disproportionality analysis employing the Empirical Bayes Geometric Mean (EBGM) and Reporting Odds Ratio (ROR) methods was conducted to identify signals for reporting cardiovascular-related adverse drug events (ADEs) associated with 13 NSAIDs. This analysis encompassed three System Organ Classes (SOCs) associated with the cardiovascular system: blood and lymphatic system disorders, cardiac disorders, and vascular disorders. The primary targets were identified through the receptor-NSAID interaction network. Ordinary least squares (OLS) regression models explored the relationship between pharmacovigilance signals and receptor occupancy rate. Results A total of 201,231 reports of cardiovascular-related ADEs were identified among the 13 NSAIDs. Dizziness, anemia, and hypertension were the most frequently reported Preferred Terms (PTs). Overall, nimesulide and parecoxib exhibited the strongest signal strengths of ADEs at SOC levels related to the cardiovascular system. On the other hand, our data presented naproxen and diclofenac as drugs of comparatively low signal strength. Cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) were identified as central targets. OLS regression analysis revealed that the normalized occupancy rate for either COX-1 or COX-2 was significantly inversely correlated with the log-transformed signal measures for blood and lymphatic system disorders and vascular disorders, and positively correlated with cardiac disorders and vascular disorders, respectively. This suggests that higher COX-2 receptor occupancy is associated with an increased cardiovascular risk from NSAIDs. Conclusion Cardiovascular safety of NSAIDs may depend on pharmacodynamic properties, specifically, the percentage of the occupied cyclooxygenase isoenzymes. More studies are needed to explore these relations and improve the prescription process.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Xianying Wang
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Xiuqing Zhu
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Ait-Oufella H, Libby P. Inflammation and Atherosclerosis: Prospects for Clinical Trials. Arterioscler Thromb Vasc Biol 2024; 44:1899-1905. [PMID: 39167675 PMCID: PMC11343092 DOI: 10.1161/atvbaha.124.320155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Affiliation(s)
- Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Sorbonne Université, Paris, France
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Zuo C, Cai L, Li Y, Ding C, Liu G, Zhang C, Wang H, Zhang Y, Ji M. The Molecular Mechanism of Radix Paeoniae Rubra.-Cortex Moutan. Herb Pair in the Treatment of Atherosclerosis: A Work Based on Network Pharmacology and In Vitro Experiments. Cardiovasc Toxicol 2024; 24:800-817. [PMID: 38951468 DOI: 10.1007/s12012-024-09881-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 07/03/2024]
Abstract
Radix Paeoniae Rubra. (Chishao, RPR) and Cortex Moutan. (Mudanpi, CM) are a pair of traditional Chinese medicines that play an important role in the treatment of atherosclerosis (AS). The main objective of this study was to identify potential synergetic function and underlying mechanisms of RPR-CM in the treatment of AS. The main active ingredients, targets of RPR-CM and AS-related genes were obtained from public databases. A Venn diagram was utilized to screen the common targets of RPR-CM in treating AS. The protein-protein interaction network was established based on STRING database. Biological functions and pathways of potential targets were analyzed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. Cytoscape was used to construct the drug-compound-target-signal pathway network. Molecular docking was performed to verify the binding ability of the bioactive ingredients and the target proteins. The endothelial inflammation model was constructed with human umbilical vein endothelial cells stimulated with ox-LDL, and the function of RPR-CM in treating AS was verified by CCK-8 assay, enzyme-linked immunosorbent assay, and qPCR. In this study, 12 active components and 401 potential target genes of RPR-CM were identified, among which quercetin, kaempferol and baicalein were considered to be the main active components. A total of 1903 AS-related genes were identified through public databases and four GEO datasets (GSE57691, GSE72633, GSE6088 and GSE199819). There are 113 common target genes of RPR-CM in treating AS. PPI network analysis identified 17 genes in cluster 1 as the core targets. Bioinformatics analysis showed that RPR-CM in AS treatment was associated with multiple downstream biological processes and signal pathways. PTGS2, JUN, CASP3, TNF, IL1B, IL6, FOS, STAT1 were identified as the core targets of RPR-CM, and molecular docking showed that the main bioactive components of RPR-CM had good binding ability with the core targets. RPR-CM extract significantly inhibited the levels of inflammatory factors TNF-α, IL-6, IL-1β, MCP-1, VCAM-1 and ICAM-1 in HUVECs, and inhibited endothelial inflammation. This study revealed the active ingredients of RPR-CM, and identified the key downstream targets and signaling pathways in the treatment of AS, providing theoretical basis for the application of RPR-CM in prevention and treatment of AS.
Collapse
Affiliation(s)
- Caojian Zuo
- Department of Cardiology, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Lianshui County, No 6, Hongri East Avenue, Huai'an, 223400, Jiangsu, China.
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China.
| | - Lidong Cai
- Department of Cardiology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Ya Li
- Department of Cardiology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Chencheng Ding
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Guiying Liu
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Changmei Zhang
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Hexiang Wang
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Yang Zhang
- Department of Cardiology, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Mingyue Ji
- Department of Cardiology, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Lianshui County, No 6, Hongri East Avenue, Huai'an, 223400, Jiangsu, China
| |
Collapse
|
7
|
Parchem K, Letsiou S, Petan T, Oskolkova O, Medina I, Kuda O, O'Donnell VB, Nicolaou A, Fedorova M, Bochkov V, Gladine C. Oxylipin profiling for clinical research: Current status and future perspectives. Prog Lipid Res 2024; 95:101276. [PMID: 38697517 DOI: 10.1016/j.plipres.2024.101276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Oxylipins are potent lipid mediators with increasing interest in clinical research. They are usually measured in systemic circulation and can provide a wealth of information regarding key biological processes such as inflammation, vascular tone, or blood coagulation. Although procedures still require harmonization to generate comparable oxylipin datasets, performing comprehensive profiling of circulating oxylipins in large studies is feasible and no longer restricted by technical barriers. However, it is essential to improve and facilitate the biological interpretation of complex oxylipin profiles to truly leverage their potential in clinical research. This requires regular updating of our knowledge about the metabolism and the mode of action of oxylipins, and consideration of all factors that may influence circulating oxylipin profiles independently of the studied disease or condition. This review aims to provide the readers with updated and necessary information regarding oxylipin metabolism, their different forms in systemic circulation, the current limitations in deducing oxylipin cellular effects from in vitro bioactivity studies, the biological and technical confounding factors needed to consider for a proper interpretation of oxylipin profiles.
Collapse
Affiliation(s)
- Karol Parchem
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Gabriela Narutowicza St., 80-233 Gdańsk, Poland; Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 53210 Pardubice, Czech Republic.
| | - Sophia Letsiou
- Department of Biomedical Sciences, University of West Attica, Ag. Spiridonos St. Egaleo, 12243 Athens, Greece.
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia.
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Isabel Medina
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain.
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| | - Anna Nicolaou
- School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, 01307 Dresden, Germany.
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France.
| |
Collapse
|
8
|
Qian C, Wang Q, Qiao Y, Xu Z, Zhang L, Xiao H, Lin Z, Wu M, Xia W, Yang H, Bai J, Geng D. Arachidonic acid in aging: New roles for old players. J Adv Res 2024:S2090-1232(24)00180-2. [PMID: 38710468 DOI: 10.1016/j.jare.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Arachidonic acid (AA), one of the most ubiquitous polyunsaturated fatty acids (PUFAs), provides fluidity to mammalian cell membranes. It is derived from linoleic acid (LA) and can be transformed into various bioactive metabolites, including prostaglandins (PGs), thromboxanes (TXs), lipoxins (LXs), hydroxy-eicosatetraenoic acids (HETEs), leukotrienes (LTs), and epoxyeicosatrienoic acids (EETs), by different pathways. All these processes are involved in AA metabolism. Currently, in the context of an increasingly visible aging world population, several scholars have revealed the essential role of AA metabolism in osteoporosis, chronic obstructive pulmonary disease, and many other aging diseases. AIM OF REVIEW Although there are some reviews describing the role of AA in some specific diseases, there seems to be no or little information on the role of AA metabolism in aging tissues or organs. This review scrutinizes and highlights the role of AA metabolism in aging and provides a new idea for strategies for treating aging-related diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW As a member of lipid metabolism, AA metabolism regulates the important lipids that interfere with the aging in several ways. We present a comprehensivereviewofthe role ofAA metabolism in aging, with the aim of relieving the extreme suffering of families and the heavy economic burden on society caused by age-related diseases. We also collected and summarized data on anti-aging therapies associated with AA metabolism, with the expectation of identifying a novel and efficient way to protect against aging.
Collapse
Affiliation(s)
- Chen Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Ze Xu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Linlin Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Haixiang Xiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Zhixiang Lin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Mingzhou Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
9
|
Ricciotti E, Haines PG, Chai W, FitzGerald GA. Prostanoids in Cardiac and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:558-583. [PMID: 38269585 PMCID: PMC10922399 DOI: 10.1161/atvbaha.123.320045] [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: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Prostanoids are biologically active lipids generated from arachidonic acid by the action of the COX (cyclooxygenase) isozymes. NSAIDs, which reduce the biosynthesis of prostanoids by inhibiting COX activity, are effective anti-inflammatory, antipyretic, and analgesic drugs. However, their use is limited by cardiovascular adverse effects, including myocardial infarction, stroke, hypertension, and heart failure. While it is well established that NSAIDs increase the risk of atherothrombotic events and hypertension by suppressing vasoprotective prostanoids, less is known about the link between NSAIDs and heart failure risk. Current evidence indicates that NSAIDs may increase the risk for heart failure by promoting adverse myocardial and vascular remodeling. Indeed, prostanoids play an important role in modulating structural and functional changes occurring in the myocardium and in the vasculature in response to physiological and pathological stimuli. This review will summarize current knowledge of the role of the different prostanoids in myocardial and vascular remodeling and explore how maladaptive remodeling can be counteracted by targeting specific prostanoids.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Philip G Haines
- Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence (P.G.H.)
| | - William Chai
- Health and Human Biology, Division of Biology and Medicine, Brown University, Providence, RI (W.C.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Medicine (G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
10
|
Tan GSQ, Morton JI, Wood S, Trevaskis NL, Magliano DJ, Windsor J, Shaw JE, Ilomäki J. COX2 inhibitor use and type 2 diabetes treatment intensification: A registry-based cohort study. Diabetes Res Clin Pract 2024; 207:111082. [PMID: 38160735 DOI: 10.1016/j.diabres.2023.111082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
AIM This study examined the association between cyclooxygenase-2 inhibitor (COX2i) use and diabetes progression in people with type 2 diabetes. METHODS We conducted a nation-wide cohort study using an Australian diabetes registry linked to medication dispensing data. We assessed time to diabetes treatment intensification among new users of COX2i compared to mild opioids. Inverse probability of treatment-weighted Cox regression models were used to adjust for age, sex, time since diabetes diagnosis, comorbidities, and socio-economic disadvantage. We conducted several sensitivity analyses, including per-protocol analyses and comparing use of any NSAID to mild opioids. RESULTS There were 8,071 new users of COX2i and 7,623 of mild opioids with 4,168 diabetes treatment intensifications over a median follow-up of 1.6 years. Use of COX2i was associated with decreased risk of treatment intensification when compared to mild opioids (HR 0.91, 95 %CI 0.85-0.96). The results were not significant in the per-protocol analyses. Use of any NSAID was associated with a lower risk of treatment intensification compared to mild opioids (HR 0.90, 95 %CI 0.85-0.96). CONCLUSIONS Treatment with COX2i may be associated with a modest decreased risk of diabetes treatment intensification compared to mild opioids. Future clinical studies are required to confirm whether COX2 inhibition has clinically significant benefits for glycaemic control.
Collapse
Affiliation(s)
- George S Q Tan
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | - Jedidiah I Morton
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Stephen Wood
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical, Sciences, Monash University, Melbourne, Victoria, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - John Windsor
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Jenni Ilomäki
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Wang Z, Mao Z, Yu M, Li H, Chen G, Wang Y, Yao Q. Role of aspirin in the prevention of heterotopic ossification following total hip replacement: a systematic review and meta-analysis. ANZ J Surg 2023; 93:1907-1916. [PMID: 37043691 DOI: 10.1111/ans.18447] [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: 12/06/2022] [Revised: 03/03/2023] [Accepted: 03/26/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND To systematically investigate if aspirin (ASA), used as venous thromboembolism (VTE) prophylaxis, plays a role in the prevention of heterotopic ossification (HO) following total hip arthroplasty (THA) and if ASA dosage impacted the rate of HO. METHODS Eligible studies published from January 2000 to July 2022 were identified from the computerized searching of PubMed, Scopus and Web of Science. HO was defined according to Brooker Classification. Pooled risk ratios (OR) and 95% confidence interval (CI) were estimated under a random-effect model. Additionally, combined HO incidences were compared according to ASA dosage (a regular dose of 325 bid vs. a low dose of 81 mg bid/162 mg qd). RESULTS Thirteen studies were included. ASA administered for VTE prophylaxis was significantly associated with a reduced risk of all-grade HO following THA (univariate, OR: 0.50, 95% CI: 0.34-0.74, P < 0.001; multivariate, OR: 0.60, 95% CI: 0.49-0.73, P < 0.001). Similar results could be observed for high-grade HO (univariate, OR: 0.57, 95% CI: 0.36-0.89, P = 0.015; multivariate, OR: 0.50, 95% CI: 0.27-0.92, P = 0.026). There was a non-significant trend towards a higher incidence of HO formation for low-dose ASA (31%, 95% CI: 29-34%), compared with regular-dose ASA (21%, 95% CI: 11-33%) (P = 0.069 under test of interaction). CONCLUSIONS ASA can be an effective option for HO prophylaxis. More well-designed trials with long-term follow-ups are encouraged to confirm the current findings and to investigate the effect of ASA dosage on HO reduction.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Zimu Mao
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Meng Yu
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Hongchuan Li
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Guoqiang Chen
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yang Wang
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Qi Yao
- Department of Orthopedics, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| |
Collapse
|
12
|
Ferreira P, Vaja R, Lopes-Pires M, Crescente M, Yu H, Nüsing R, Liu B, Zhou Y, Yaqoob M, Zhang A, Rickman M, Longhurst H, White WE, Knowles RB, Chan MV, Warner TD, Want E, Kirkby NS, Mitchell JA. Renal Function Underpins the Cyclooxygenase-2: Asymmetric Dimethylarginine Axis in Mouse and Man. Kidney Int Rep 2023; 8:1231-1238. [PMID: 37284684 PMCID: PMC10239776 DOI: 10.1016/j.ekir.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction Through the production of prostacyclin, cyclooxygenase (COX)-2 protects the cardiorenal system. Asymmetric dimethylarginine (ADMA), is a biomarker of cardiovascular and renal disease. Here we determined the relationship between COX-2/prostacyclin, ADMA, and renal function in mouse and human models. Methods We used plasma from COX-2 or prostacyclin synthase knockout mice and from a unique individual lacking COX-derived prostaglandins (PGs) because of a loss of function mutation in cytosolic phospholipase A2 (cPLA2), before and after receiving a cPLA2-replete transplanted donor kidney. ADMA, arginine, and citrulline were measured using ultra-high performance liquid-chromatography tandem mass spectrometry. ADMA and arginine were also measured by enzyme-linked immunosorbent assay (ELISA). Renal function was assessed by measuring cystatin C by ELISA. ADMA and prostacyclin release from organotypic kidney slices were also measured by ELISA. Results Loss of COX-2 or prostacyclin synthase in mice increased plasma levels of ADMA, citrulline, arginine, and cystatin C. ADMA, citrulline, and arginine positively correlated with cystatin C. Plasma ADMA, citrulline, and cystatin C, but not arginine, were elevated in samples from the patient lacking COX/prostacyclin capacity compared to levels in healthy volunteers. Renal function, ADMA, and citrulline were returned toward normal range when the patient received a genetically normal kidney, capable of COX/prostacyclin activity; and cystatin C positively correlated with ADMA and citrulline. Levels of ADMA and prostacyclin in conditioned media of kidney slices were not altered in tissue from COX-2 knockout mice compared to wildtype controls. Conclusion In human and mouse models, where renal function is compromised because of loss of COX-2/PGI2 signaling, ADMA levels are increased.
Collapse
Affiliation(s)
- Plinio Ferreira
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ricky Vaja
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Maria Lopes-Pires
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Marilena Crescente
- Blizard Institute, Barts, and The London School of Medicine and Dentistry, London, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - He Yu
- Deparment of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Rolf Nüsing
- Clinical Pharmacology and Pharmacotherapy Department, Goethe University, Frankfurt, Germany
| | - Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Magdi Yaqoob
- Blizard Institute, Barts, and The London School of Medicine and Dentistry, London, United Kingdom
| | - Anran Zhang
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Matthew Rickman
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Hilary Longhurst
- Department of Medicine, University of Auckland, and Department of Immunology, Auckland City Hospital, Auckland, New Zealand
| | - William E. White
- Blizard Institute, Barts, and The London School of Medicine and Dentistry, London, United Kingdom
| | - Rebecca B. Knowles
- Blizard Institute, Barts, and The London School of Medicine and Dentistry, London, United Kingdom
| | - Melissa V. Chan
- Blizard Institute, Barts, and The London School of Medicine and Dentistry, London, United Kingdom
| | - Timothy D. Warner
- Blizard Institute, Barts, and The London School of Medicine and Dentistry, London, United Kingdom
| | - Elizabeth Want
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Nicholas S. Kirkby
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Jane A. Mitchell
- National Heart and Lung Institute, Imperial College London, United Kingdom
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
Kaur M, Muzzammel Rehman H, Kaur G, Kaur A, Bansal M. Switching of newly synthesized linker-based derivatives of non-steroidal anti-inflammatory drugs toward anti-inflammatory and anticancer activity. Bioorg Chem 2023; 133:106406. [PMID: 36773455 DOI: 10.1016/j.bioorg.2023.106406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
A new series of linker-based derivatives of non-steroidal anti-inflammatory drugs were designed and synthesized. All the compounds were well characterized with the help of various spectroscopic techniques such as FT-IR, 1H NMR, 13C NMR, and HRMS. The main emphasis of this paper is to understand the switching of the most promising compounds 8 and 10 towards anti-inflammatory and anticancer activity in terms of in-silico and in-vitro studies in detail. During the molecular docking study, compounds 8 and 10 demonstrated the importance of hetero atoms as well as the perfect alignment of a compound in the binding pocket of a target site, which may affect their bioactivity. Here, the presence of 1,3‑dicarbonyl interactions with ASN 351 in compound 8 (not found in compound 10) may be responsible for its better inhibitory activity against the COX-2 target site. On the other hand, a slight increase in the potency of compound 10 towards anticancer activity may be due to the instantaneous participation of the OH group and carbonyl group to give conventional hydrogen bonds towards THR 149 amino acid residue, which was missing in compound 8. Molecular dynamics simulation was also performed for compounds 10 and 8 toward COX-2 and HER-2 protein sites. Further, compounds 8 and 10 were subjected to in-vitro COX-2 inhibition and cytotoxicity assay and the results obtained were in accordance with the in-silico study. Thus, compound 8 become more potent towards COX-2 inhibition with IC50 value of 48.51 µg/ml and compound 10 showed good bioactivity toward cytotoxic activity with IC50 value of 93.03 µg/ml.
Collapse
Affiliation(s)
- Mandeep Kaur
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India
| | - Hafiz Muzzammel Rehman
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Punjab, Pakistan
| | - Gurmeet Kaur
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India
| | - Amandeep Kaur
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India
| | - Manisha Bansal
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India.
| |
Collapse
|
15
|
Koshman YE, Bielinski AL, Bird BM, Green JR, Kowalkowski KL, Lai-Zhang J, Mahalingaiah PK, Sawicki JW, Talaty NN, Wilsey AS, Zafiratos MT, Van Vleet TR. Disconnect between COX-2 selective inhibition and cardiovascular risk in preclinical models. J Pharmacol Toxicol Methods 2023; 120:107251. [PMID: 36792039 DOI: 10.1016/j.vascn.2023.107251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/20/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Secondary pharmacology profiling is routinely applied in pharmaceutical drug discovery to investigate the pharmaceutical effects of a drug at molecular targets distinct from (off-target) the intended therapeutic molecular target (on-target). Data from a randomized, placebo-controlled clinical trial, the APPROVe (Adenomatous Polyp Prevention on VIOXX, rofecoxib) trial, raised significant concerns about COX-2 inhibition as a primary or secondary target, shaping the screening and decision-making processes of some pharmaceutical companies. COX-2 is often included in off-target screens due to cardiovascular (CV) safety concerns about secondary interactions with this target. Several potential mechanisms of COX-2-mediated myocardial infarctions have been considered including, effects on platelet stickiness/aggregation, vasal tone and blood pressure, and endothelial cell activation. In the present study, we focused on each of these mechanisms as potential effects of COX-2 inhibitors, to find evidence of mechanism using various in vitro and in vivo preclinical models. METHODS Compounds tested in the study, with a range of COX-2 selectivity, included rofecoxib, celecoxib, etodolac, and meloxicam. Compounds were screened for inhibition of COX-2 vs COX-1 enzymatic activity, ex vivo platelet aggregation (using whole blood from multiple species), ex vivo canine femoral vascular ring model, in vitro human endothelial cell activation (with and without COX-2 induction), and in vivo cardiovascular assessment (anesthetized dog). RESULTS The COX-2 binding assessment generally confirmed the COX-2 selectivity previously reported. COX-2 inhibitors did not have effects on platelet function (spontaneous aggregation or inhibition of aggregation), cardiovascular parameters (mean arterial pressure, heart rate, and left ventricular contractility), or endothelial cell activation. However, rofecoxib uniquely produced an endothelial mediated constriction response in canine femoral arteries. CONCLUSION Our data suggest that rofecoxib-related cardiovascular events in humans are not predicted by COX-2 potency or selectivity. In addition, the vascular ring model suggested possible adverse cardiovascular effects by COX-2 inhibitors, although these effects were not seen in vivo studies. These results may also suggest that COX-2 inhibition alone is not responsible for rofecoxib-mediated adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America.
| | - Aimee L Bielinski
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Brandan M Bird
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Jonathon R Green
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Kenneth L Kowalkowski
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Jie Lai-Zhang
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | | | - James W Sawicki
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Nari N Talaty
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Amanda S Wilsey
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Mark T Zafiratos
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Terry R Van Vleet
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| |
Collapse
|
16
|
Rytz CL, Dumanski SM, Sola DY, Ahmed SB. The Effect of Biological Sex on Arterial Stiffness and Renin-Angiotensin-Aldosterone System Activity in Response to Cyclooxygenase-2 (COX-2) Inhibition. CJC Open 2023; 5:112-119. [PMID: 36880076 PMCID: PMC9984894 DOI: 10.1016/j.cjco.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Background Cardiovascular disease is the leading cause of death globally. Cyclooxygenase (COX)-derived prostaglandins play an important role in cardiovascular health regulation. Animal studies suggest a greater vascular dependence on prostaglandins in female subjects, but whether this extends to humans is unknown. We aimed to assess the effect of COX-2 inhibition on blood pressure and arterial stiffness, validated markers of cardiovascular risk, in human adults. Methods Healthy premenopausal females and males were studied in high-salt balance before and after 14 days of daily oral celecoxib, 200 mg ingestion, on 2 identical study days. Blood pressure (BP) and pulse-wave velocity (PWV) were measured at baseline and in response to an Angiotensin II (AngII) challenge, a validated marker of renin-angiotensin-aldosterone system activity. Results Thirteen females (age [mean ± standard deviation], 38 ± 13 years) and 11 males (age, 34 ± 9 years) were studied. Pre-COX-2 inhibition, resting measures of systolic (S)BP (P = 0.2) and diastolic (D)BP (P = 0.1) were similar between sexes. Post-COX-2 inhibition, resting SBP (P < 0.001) and DBP (P = 0.02) were significantly lower in females than in males. COX-2 inhibition was not associated with changes in arterial parameters by sex (change in DBP: P = 0.54; change in PWV: P = 0.55; females vs males). COX-2 inhibition was associated with increased SBP (P = 0.039 vs pre-COX-2 inhibition), but no change in DBP (P = 0.16) or PWV (P = 0.52) response to AngII challenge in females. Measures did not differ in response to AngII pre- vs post-COX-2 inhibition in males (SBP: P = 0.88; DBP: P = 0.93; PWV: P = 0.97). Conclusions The effects of COX-2 inhibition on arterial function may differ by sex, but further studies are needed. Given the association between nonsteroidal anti-inflammatory drugs (NSAIDs) and cardiovascular risk, increased attention regarding sex-specific pathophysiology is warranted.
Collapse
Affiliation(s)
- Chantal L Rytz
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sandra M Dumanski
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Kidney Disease Network, Calgary, Alberta, Canada.,O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Darlene Y Sola
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sofia B Ahmed
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Kidney Disease Network, Calgary, Alberta, Canada.,O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
17
|
Gaul DS, Calatayud N, Pahla J, Bonetti NR, Wang YJ, Weber J, Ambrosini S, Liberale L, Costantino S, Mohammed SA, Kraler S, Van Tits LJ, Pasterk L, Vdovenko D, Akhmedov A, Ruschitzka F, Paneni F, Lüscher TF, Camici GG, Matter CM. Endothelial SIRT6 deficiency promotes arterial thrombosis in mice. J Mol Cell Cardiol 2023; 174:56-62. [PMID: 36414111 DOI: 10.1016/j.yjmcc.2022.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/27/2022] [Accepted: 11/17/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Arterial thrombosis may be initiated by endothelial inflammation or denudation, activation of blood-borne elements or the coagulation system. Tissue factor (TF), a central trigger of the coagulation cascade, is regulated by the pro-inflammatory NF-κB-dependent pathways. Sirtuin 6 (SIRT6) is a nuclear member of the sirtuin family of NAD+-dependent deacetylases and is known to inhibit NF-κB signaling. Its constitutive deletion in mice shows early lethality with hypoglycemia and accelerated aging. Of note, the role of SIRT6 in arterial thrombosis remains unknown. Thus, we hypothesized that endothelial SIRT6 protects from arterial thrombosis by modulating inhibition of NF-κB-associated pathways. APPROACH AND RESULTS Using a laser-induced carotid thrombosis model, in vivo arterial occlusion occurred 45% faster in 12-week-old male endothelial-specific Sirt6-/- mice as compared to Sirt6fl/fl controls (n ≥ 9 per group; p = 0.0012). Levels of procoagulant TF were increased in animals lacking endothelial SIRT6 as compared to control littermates. Similarly, in cultured human aortic endothelial cells, SIRT6 knockdown increased TF mRNA, protein and activity. Moreover, SIRT6 knockdown increased mRNA levels of NF-κB-associated genes tumor necrosis factor alpha (TNF-α), poly [ADP-ribose] polymerase 1 (PARP-1), vascular cell adhesion molecule 1 (VCAM-1), and cyclooxygenase-2 (COX-2); at the protein level, COX-2, VCAM-1, TNF-α, and cleaved PARP-1 remained increased after Sirt6 knockdown. CONCLUSIONS Endothelium-specific Sirt6 deletion promotes arterial thrombosis in mice. In cultured human aortic endothelial cells, SIRT6 silencing enhances TF expression and activates pro-inflammatory pathways including TNF-α, cleaved PARP-1, VCAM-1 and COX-2. Hence, endogenous endothelial SIRT6 exerts a protective role in experimental arterial thrombosis.
Collapse
Affiliation(s)
- Daniel S Gaul
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Natacha Calatayud
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Jürgen Pahla
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Nicole R Bonetti
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Yu-Jen Wang
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Julien Weber
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Samuele Ambrosini
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Lambertus J Van Tits
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Lisa Pasterk
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland; Department of Cardiology, University Hospital Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland; Department of Cardiology, University Hospital Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Christian M Matter
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland; Department of Cardiology, University Hospital Zurich, Switzerland.
| |
Collapse
|
18
|
Zhang J, Ji C, Zhai X, Tong H, Hu J. Frontiers and hotspots evolution in anti-inflammatory studies for coronary heart disease: A bibliometric analysis of 1990-2022. Front Cardiovasc Med 2023; 10:1038738. [PMID: 36873405 PMCID: PMC9978200 DOI: 10.3389/fcvm.2023.1038738] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/17/2023] [Indexed: 02/18/2023] Open
Abstract
Background Coronary heart disease (CHD) is characterized by forming of arterial plaques composed mainly of lipids, calcium, and inflammatory cells. These plaques narrow the lumen of the coronary artery, leading to episodic or persistent angina. Atherosclerosis is not just a lipid deposition disease but an inflammatory process with a high-specificity cellular and molecular response. Anti-inflammatory treatment for CHD is a promising therapy; several recent clinical studies (CANTOS, COCOLT, and LoDoCo2) provide therapeutic directions. However, bibliometric analysis data on anti-inflammatory conditions in CHD are lacking. This study aims to provide a comprehensive visual perspective on the anti-inflammatory research in CHD and will contribute to further research. Materials and methods All the data were collected from the Web of Science Core Collection (WoSCC) database. We used the Web of Science's systematic tool to analyze the year of countries/regions, organizations, publications, authors, and citations. CiteSpace and VOSviewer were used to construct visual bibliometric networks to reveal the current status and emerging hotspot trends for anti-inflammatory intervention in CHD. Results 5,818 papers published from 1990 to 2022 were included. The number of publications has been on the rise since 2003. Libby Peter is the most prolific author in the field. "Circulation" was ranked first in the number of journals. The United States has contributed the most to the number of publications. The Harvard University System is the most published organization. The top 5 clusters of keywords co-occurrence are inflammation, C-reactive protein, coronary heart disease, nonsteroidal anti-inflammatory, and myocardial infarction. The top 5 literature citation topics are chronic inflammatory diseases, cardiovascular risk; systematic review, statin therapy; high-density lipoprotein. In the past 2 years, the strongest keyword reference burst is "Nlrp3 inflammasome," and the strongest citation burst is "Ridker PM, 2017 (95.12)." Conclusion This study analyzes the research hotspots, frontiers, and development trends of anti-inflammatory applications in CHD, which is of great significance for future studies.
Collapse
Affiliation(s)
- Jiale Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenyang Ji
- Science and Technology College of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Xu Zhai
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongxuan Tong
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingqing Hu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Bishop RC, McCoy AM, Kemper AM, Stewart RM, Wilkins PA. Short-term administration of flunixin meglumine or firocoxib does not alter viscoelastic coagulation profiles in healthy horses. J Am Vet Med Assoc 2022; 260:1963-1966. [PMID: 36198050 DOI: 10.2460/javma.22.08.0367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the effect of the cyclooxygenase-2-selective NSAID firocoxib, compared to the nonselective NSAID flunixin meglumine on viscoelastic coagulation parameters in healthy horses. ANIMALS 12 healthy adult mixed-breed horses. PROCEDURES Following a crossover protocol, horses were administered flunixin meglumine (1.1 mg/kg, IV, q 12 h for 5 days), allowed a 6-month washout period, and then administered firocoxib (0.3 mg/kg, PO, once, then 0.1 mg/kg, PO, q 24 h for 4 days). Omeprazole (1 mg/kg, PO, q 24 h) was administered concurrently with each NSAID. Viscoelastic coagulation profiles and traditional coagulation parameters (prothrombin time, partial thromboplastin time, and fibrinogen) were measured before and after each treatment. RESULTS Viscoelastic coagulation parameters were within reference intervals before and after both treatments. There was a statistically significant difference between treatments for amplitude at 10 minutes after clot time (P = .02) and maximum clot formation (P = .02); however, the magnitude of change was not clinically significant. CLINICAL RELEVANCE Short-term administration of flunixin meglumine and firocoxib did not result in significant alteration of viscoelastic coagulation profiles in healthy horses. However, clinicians should be aware of possible coagulopathy secondary to NSAID administration with long-term use or critical illness, and further study is indicated.
Collapse
|
20
|
Frommeyer TC, Gilbert MM, Brittain GV, Wu T, Nguyen TQ, Rohan CA, Travers JB. UVB-Induced Microvesicle Particle Release and Its Effects on the Cutaneous Microenvironment. Front Immunol 2022; 13:880850. [PMID: 35603177 PMCID: PMC9120817 DOI: 10.3389/fimmu.2022.880850] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/04/2022] [Indexed: 12/14/2022] Open
Abstract
Ultraviolet B radiation (UVB) has profound effects on human skin that results in a broad spectrum of immunological local and systemic responses and is the major cause of skin carcinogenesis. One important area of study in photobiology is how UVB is translated into effector signals. As the skin is exposed to UVB light, subcellular microvesicle particles (MVP), a subtype of bioactive extracellular vesicles, are released causing a variety of local and systemic immunological effects. In this review, we highlight keratinocyte MVP release in keratinocytes in response to UVB. Specifically, Platelet-activating factor receptor agonists generated by UVB result in MVP released from keratinocytes. The downstream effects of MVP release include the ability of these subcellular particles to transport agents including the glycerophosphocholine-derived lipid mediator Platelet-activating factor (PAF). Moreover, even though UVB is only absorbed in the epidermis, it appears that PAF release from MVPs also mediates systemic immunosuppression and enhances tumor growth and metastasis. Tumor cells expressing PAF receptors can use this mechanism to evade chemotherapy responses, leading to treatment resistance for advanced cancers such as melanoma. Furthermore, novel pharmacological agents provide greater insight into the UVB-induced immune response pathway and a potential target for pharmacological intervention. This review outlines the need to more clearly elucidate the mechanism linking UVB-irradiation with the cutaneous immune response and its pathological manifestations. An improved understanding of this process can result in new insights and treatment strategies for UVB-related disorders from carcinogenesis to photosensitivity.
Collapse
Affiliation(s)
- Timothy C. Frommeyer
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
| | - Michael M. Gilbert
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
| | - Garrett V. Brittain
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
| | - Tongfan Wu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
| | - Trang Q. Nguyen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
| | - Craig A. Rohan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
- Department of Dermatology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
- Department of Medicine, Dayton Veterans Administration Medical Center, Dayton, OH, United States
| | - Jeffrey B. Travers
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
- Department of Dermatology, Boonshoft School of Medicine at Wright State University, Dayton, OH, United States
- Department of Medicine, Dayton Veterans Administration Medical Center, Dayton, OH, United States
- *Correspondence: Jeffrey B. Travers,
| |
Collapse
|
21
|
Kong D, Yu Y. Prostaglandin D2 signaling and cardiovascular homeostasis. J Mol Cell Cardiol 2022; 167:97-105. [DOI: 10.1016/j.yjmcc.2022.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/25/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
22
|
Ampomah PB, Cai B, Sukka SR, Gerlach BD, Yurdagul A, Wang X, Kuriakose G, Darville LNF, Sun Y, Sidoli S, Koomen JM, Tall AR, Tabas I. Macrophages use apoptotic cell-derived methionine and DNMT3A during efferocytosis to promote tissue resolution. Nat Metab 2022; 4:444-457. [PMID: 35361955 PMCID: PMC9050866 DOI: 10.1038/s42255-022-00551-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/11/2022] [Indexed: 12/19/2022]
Abstract
Efferocytosis, the clearance of apoptotic cells (ACs) by macrophages, is critical for tissue resolution, with defects driving many diseases. Mechanisms of efferocytosis-mediated resolution are incompletely understood. Here, we show that AC-derived methionine regulates resolution through epigenetic repression of the extracellular signal-regulated kinase 1/2 (ERK1/2) phosphatase Dusp4. We focus on two key efferocytosis-induced pro-resolving mediators, prostaglandin E2 (PGE2) and transforming growth factor beta 1 (TGF-β1), and show that efferocytosis induces prostaglandin-endoperoxide synthase 2/cyclooxygenase 2 (Ptgs2/COX2), leading to PGE2 synthesis and PGE2-mediated induction of TGF-β1. ERK1/2 phosphorylation/activation by AC-activated CD36 is necessary for Ptgs2 induction, but this is insufficient owing to an ERK-DUSP4 negative feedback pathway that lowers phospho-ERK. However, subsequent AC engulfment and phagolysosomal degradation lead to Dusp4 repression, enabling enhanced p-ERK and induction of the Ptgs2-PGE2-TGF-β1 pathway. Mechanistically, AC-derived methionine is converted to S-adenosylmethionine, which is used by DNA methyltransferase-3A (DNMT3A) to methylate Dusp4. Bone-marrow DNMT3A deletion in mice blocks COX2/PGE2, TGF-β1, and resolution in sterile peritonitis, apoptosis-induced thymus injury and atherosclerosis. Knowledge of how macrophages use AC-cargo and epigenetics to induce resolution provides mechanistic insight and therapeutic options for diseases driven by impaired resolution.
Collapse
Affiliation(s)
- Patrick B Ampomah
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Shreveport, Shreveport, LA, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lancia N F Darville
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yan Sun
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alan R Tall
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
23
|
Cano-Granda DV, Ramírez-Ramírez M, M. Gómez D, Hernandez JC. Effects of particulate matter on endothelial, epithelial and immune system cells. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.01.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Particulate Matter (PM) is an air pollutant that is classified according to its aerodynamic diameter into particles with a diameter of less than 10 µm (PM10), a diameter of less than 2.5 µm (PM2.5), and particles ultra-fine with a diameter less than 0.1 µm (PM0.1). PM10 is housed in the respiratory system, while PM2.5 and 0.1 can pass into the circulation to generate systemic alterations. Although several diseases associated with PM exposure, such as respiratory, cardiovascular, and central nervous system, have been documented to cause 4.2 million premature deaths per year worldwide. Few reviews address cellular and molecular mechanisms in the epithelial and endothelial cells of the tissues exposed to PM, which can cause these diseases, this being the objective of the present review. For this, a search was carried out in the NCBI and Google Scholar databases focused on scientific publications that addressed the expression of pro-inflammatory molecules, adhesion molecules, and oxidative radicals, among others, and their relationship with the effects caused by the PM. The main findings include the increase in pro-inflammatory cytokines and dysfunction in the components of the immune response; the formation of reactive oxygen species; changes in epithelial and endothelial function, evidenced by altered expression of adhesion molecules; and the increase in molecules involved in coagulation. Complementary studies are required to understand the molecular effects of harmful health effects and the future approach to strategies to mitigate this response.
Collapse
Affiliation(s)
- Danna V. Cano-Granda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia 2 Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| | - Mariana Ramírez-Ramírez
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia 2 Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| | - Diana M. Gómez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| |
Collapse
|
24
|
Impact of Medical Interventions and Comorbidities on Norwood Admission for Patients with Hypoplastic Left Heart Syndrome. Pediatr Cardiol 2022; 43:267-278. [PMID: 35034159 DOI: 10.1007/s00246-022-02818-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
The purpose of these analyses was to determine how specific comorbidities and medical interventions impact risk of inpatient mortality in those with hypoplastic left heart syndrome undergoing Norwood procedure. The secondary aims were to determine the impact of these on billed charges, postoperative length of stay, and risk of cardiac arrest. Admissions from 2004 to 2015 in the Pediatric Health Information System database with hypoplastic left heart syndrome and Norwood procedure were identified. Admission characteristics, patient interventions, and the presence of comorbidities were captured. A total of 5,138 admissions were identified meeting inclusion criteria. Of these 829 (16.1%) experienced inpatient mortality, and 352 (6.7%) experienced cardiac arrest. The frequency of inpatient mortality did not significantly change over the course of the study era. The frequency of cardiac arrest significantly decreased from 7.4% in 2004 to 4.3% in 2015 (p = 0.04). The frequency of pharmacologic therapies, particularly vasoactive use, decreased as the study period progressed. Regression analyses demonstrated a significant association between cardiac arrest and inpatient mortality with arrhythmias, acute kidney injury, and pulmonary hypertension. Similarly, regression analyses demonstrated a significant association between increase in billed charges and length of stay with year of surgery, presence of heart failure, syndromes, and acute kidney injury. For patients with hypoplastic left heart syndrome undergoing the Norwood procedure, the frequency of pharmacologic therapies and cardiac arrest has decreased over time. There are significant associations between acute kidney injury, arrythmias, and pulmonary hypertension with cardiac arrest and mortality.
Collapse
|
25
|
Bekhit AA, Nasralla SN, El-Agroudy EJ, Hamouda N, El-Fattah AA, Bekhit SA, Amagase K, Ibrahim TM. Investigation of the anti-inflammatory and analgesic activities of promising pyrazole derivative. Eur J Pharm Sci 2022; 168:106080. [PMID: 34818572 DOI: 10.1016/j.ejps.2021.106080] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 02/02/2023]
Abstract
The development of new COX-2 inhibitors with analgesic and anti-inflammatory efficacy as well as minimal gastrointestinal, renal and cardiovascular toxicity, is of vital importance to patients suffering from chronic course pain and inflammatory conditions. This study aims at evaluating the therapeutic activity and adverse drug reactions associated with the use of the newly synthesized pyrazole derivative, compound AD732, E-4-[3-(4-methylphenyl)-5-hydroxyliminomethyl-1H-pyrazol-1-yl]benzenesulfonamide, as compared to indomethacin and celecoxib as standard agents. Anti-inflammatory activity was assessed using carrageenan-induced rat paw edema and cotton pellet granuloma tests; formalin-induced hyperalgesia and hot plate tests were done to study analgesic activity. In vitro tests to determine COX-1/COX-2 selectivity and assessment of renal and gastric toxicity upon acute exposure to AD732 were also conducted. Compound AD732 exhibited promising results; higher anti-inflammatory and analgesic effects compared to standard agents, coupled with the absence of ulcerogenic effects and minimal detrimental effects on renal function. Additionally, compound AD732 was a less potent inhibitor of COX-2 in vitro than celecoxib, which may indicate lower potential cardiovascular toxicity. It may be concluded that compound AD732 appears to be a safer and more effective molecule with promising potential for the management of pain and inflammation.
Collapse
Affiliation(s)
- Adnan A Bekhit
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Pharmacy Program, Pharmacology stream, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, Kingdom of Bahrain.
| | - Sherry N Nasralla
- Pharmacy Program, Pharmacology stream, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, Kingdom of Bahrain
| | - Eman J El-Agroudy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Nahla Hamouda
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ahmed Abd El-Fattah
- Department of Materials Science, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt; Department of Chemistry, College of Science, University of Bahrain, Sakhir P.O. Box. 32038, Kingdom of Bahrain
| | - Salma A Bekhit
- High Institute of Public Health, Alexandria University, Alexandria 21568, Egypt
| | - Kikuko Amagase
- Laboratory of Pharmacology & Pharmacotherapeutics, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Tamer M Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| |
Collapse
|
26
|
Nellaiappan K, Preeti K, Khatri DK, Singh SB. Diabetic Complications: An Update on Pathobiology and Therapeutic Strategies. Curr Diabetes Rev 2022; 18:e030821192146. [PMID: 33745424 DOI: 10.2174/1573399817666210309104203] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/28/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022]
Abstract
Despite the advent of novel therapies which manage and control diabetes well, the increased risk of morbidity and mortality in diabetic subjects is associated with the devastating secondary complications it produces. Long-standing diabetes majorly drives cellular and molecular alterations, which eventually damage both small and large blood vessels. The complications are prevalent both in type I and type II diabetic subjects. The microvascular complications include diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, while the macrovascular complications include diabetic heart disease and stroke. The current therapeutic strategy alleviates the complications to some extent but does not cure or prevent them. Also, the recent clinical trial outcomes in this field are disappointing. Success in the drug discovery of diabetic complications may be achieved by a better understanding of the underlying pathophysiology and by recognising the crucial factors contributing to the development and progression of the disease. In this review, we discuss the well-studied cellular mechanisms leading to the development and progression of diabetic complications. In addition, we also highlight the various therapeutic paradigms currently in clinical practice.
Collapse
Affiliation(s)
- Karthika Nellaiappan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| | - Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| | - Shashi Bala Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| |
Collapse
|
27
|
Sadeghi M, Miroliaei M, Fateminasab F, Moradi M. Screening cyclooxygenase-2 inhibitors from Allium sativum L. compounds: in silico approach. J Mol Model 2021; 28:24. [PMID: 34970708 DOI: 10.1007/s00894-021-05016-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 12/23/2021] [Indexed: 12/31/2022]
Abstract
Inflammation is a natural protective response toward various simulators, including tissue damage or pathogens. The cyclooxygenase-2 (COX-2) is a very important protein in triggering pain and inflammation. Previous studies have claimed that Allium sativum offers a wide range of anti-inflammatory therapeutics for human consumption. Drug discovery is a complicated process, though in silico methods can make this procedure simpler and more cost-effective. At the current study, we performed the virtual screening of eight Allium sativum-derived compounds via molecular docking with COX-2 enzyme and confirmed the binding energy by docking score estimate followed by ADMET and drug-likeness investigation. The resulting highest-docking scored compound was exposed to molecular dynamics simulation (MDS) for evaluating stability of the docked enzyme-ligand complex and to gauge the oscillation and conformational alterations for the time of enzyme-ligand interaction. The factors of RMSD, RMSF, hydrogen bond interactions, and Rg after 100 ns of MDS proved the stability of alliin in the active site of COX-2 in comparison with celecoxib (CEL) as the control. Moreover, we investigated the binding affinity analysis of all compounds via MM/PBSA method. The results from this study suggest that alliin (a sulfuric compound) exhibits a higher binding affinity for the COX-2 enzyme compared to the other compounds and CEL. Alliin showed to be a possible anti-inflammatory therapeutic candidate for managing the inflammatory conditions.
Collapse
Affiliation(s)
- Morteza Sadeghi
- Faculty of Biological Science and Technology, Department of Cell and Molecular Biology & Microbiology, University of Isfahan, Isfahan, Iran
| | - Mehran Miroliaei
- Faculty of Biological Science and Technology, Department of Cell and Molecular Biology & Microbiology, University of Isfahan, Isfahan, Iran.
| | | | - Mohammad Moradi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
28
|
Felipe JL, Cassamale TB, Lourenço LD, Carvalho DB, das Neves AR, Duarte RCF, Carvalho MG, Toffoli-Kadri MC, Baroni ACM. Anti-inflammatory, ulcerogenic and platelet activation evaluation of novel 1,4-diaryl-1,2,3-triazole neolignan-celecoxib hybrids. Bioorg Chem 2021; 119:105485. [PMID: 34959176 DOI: 10.1016/j.bioorg.2021.105485] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/12/2021] [Accepted: 11/08/2021] [Indexed: 01/06/2023]
Abstract
This study reports the synthesis of novel neolignans-celecoxib hybrids and the evaluation of their biological activity. Analogs8-13(L13-L18) exhibited anti-inflammatory activity, inhibited glycoprotein expression (P-selectin) related to platelet activation, and were considered non- ulcerogenic in the animal model, even with the administration of 10 times higher than the dose used in reference therapy. In silico drug-likeness showed that the analogs are compliant with Lipinski's rule of five. A molecular docking study showed that the hybrids8-13(L13-L18) fitted similarly with celecoxib in the COX-2 active site. According to this data, it is possible to infer that extra hydrophobic interactions and the hydrogen interactions with the triazole core may improve the selectivity towards the COX-2 active site. Furthermore, the molecular docking study with P-selectin showed the binding affinity of the analogs in the active site, performing important interactions with amino acid residues such as Tyr 48. Whereas the P-selectin is a promising target to the design of new anti-inflammatory drugs with antithrombotic properties, a distinct butterfly-like structure of 1,4-diaryl-1,2,3-triazole neolignan-celecoxib hybrids synthesized in this work may be a safer alternative to the traditional COX-2 inhibitors.
Collapse
Affiliation(s)
- Josyelen L Felipe
- Laboratório de Farmacologia e Inflamação (LABFAR), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil
| | - Tatiana B Cassamale
- Laboratório de Síntese e Química Medicinal (LASQUIM), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil
| | - Leticia D Lourenço
- Laboratório de Farmacologia e Inflamação (LABFAR), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil
| | - Diego B Carvalho
- Laboratório de Síntese e Química Medicinal (LASQUIM), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil
| | - Amarith R das Neves
- Laboratório de Síntese e Química Medicinal (LASQUIM), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil
| | - Rita C F Duarte
- Departamento de Análises Clínicas e Toxicológicas da Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, MG, Brazil
| | - Maria G Carvalho
- Departamento de Análises Clínicas e Toxicológicas da Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, MG, Brazil
| | - Monica C Toffoli-Kadri
- Laboratório de Farmacologia e Inflamação (LABFAR), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil.
| | - Adriano C M Baroni
- Laboratório de Síntese e Química Medicinal (LASQUIM), FACFAN - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil.
| |
Collapse
|
29
|
Gerges SH, El-Kadi AOS. Sex differences in eicosanoid formation and metabolism: A possible mediator of sex discrepancies in cardiovascular diseases. Pharmacol Ther 2021; 234:108046. [PMID: 34808133 DOI: 10.1016/j.pharmthera.2021.108046] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Arachidonic acid is metabolized by cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes to produce prostaglandins, leukotrienes, epoxyeicosatrienoic acids (EETs), and hydroxyeicosatetraenoic acids (HETEs), along with other eicosanoids. Eicosanoids have important physiological and pathological roles in the body, including the cardiovascular system. Evidence from several experimental and clinical studies indicates differences in eicosanoid levels, as well as in the activity or expression levels of their synthesizing and metabolizing enzymes between males and females. In addition, there is a clear state of gender specificity in cardiovascular diseases (CVD), which tend to be more common in men compared to women, and their risk increases significantly in postmenopausal women compared to younger women. This could be largely attributed to sex hormones, as androgens exert detrimental effects on the heart and blood vessels, whereas estrogen exhibits cardioprotective effects. Many of androgen and estrogen effects on the cardiovascular system are mediated by eicosanoids. For example, androgens increase the levels of cardiotoxic eicosanoids like 20-HETE, while estrogens increase the levels of cardioprotective EETs. Thus, sex differences in eicosanoid levels in the cardiovascular system could be an important underlying mechanism for the different effects of sex hormones and the differences in CVD between males and females. Understanding the role of eicosanoids in these differences can help improve the management of CVD.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
30
|
Jang G, Lee S, Hong J, Park B, Kim D, Kim C. Anti-Inflammatory Effect of 4,5-Dicaffeoylquinic Acid on RAW264.7 Cells and a Rat Model of Inflammation. Nutrients 2021; 13:3537. [PMID: 34684537 PMCID: PMC8538716 DOI: 10.3390/nu13103537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Anti-inflammatory agents that are safer and more effective than the currently used non-steroidal anti-inflammatory drugs are urgently needed. The dicaffeoylquinic acid (diCQA) isomer 4,5-diCQA exhibits antioxidant activity and various other health-promoting benefits; however, its anti-inflammatory properties require further investigation. This study was conducted to evaluate the anti-inflammatory properties of 4,5-diCQA in vitro and in vivo using RAW264.7 cells and a carrageenan-induced inflammation model, respectively. In RAW264.7 cells, 4,5-diCQA pretreatment significantly inhibited lipopolysaccharide-induced expression of nitric oxide, prostaglandin E2, nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-α, interleukin-1β, and interleukin-6, without inducing cytotoxicity. The inhibitory effects of 4,5-diCQA were mediated by the suppression of nuclear factor-κB nuclear translocation and mitogen-activated protein kinase (MAPK) phosphorylation. Oral administration of 4,5-diCQA at doses of 5, 10, and 20 mg/kg of the body weight suppressed carrageenan-induced edema and the expression of nitric oxide synthase, cyclooxygenase-2, and tumor necrosis factor-α in a dose-dependent manner. Collectively, our results suggest that 4,5-diCQA exerts anti-inflammatory effects by suppressing activation of the nuclear factor-κB and MAPK pathways in vitro and reducing carrageenan-induced edema in vivo. Therefore, 4,5-diCQA shows potential as a natural alternative to non-steroidal anti-inflammatory drugs.
Collapse
Affiliation(s)
- Goeun Jang
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Korea; (G.J.); (S.L.)
| | - Seulah Lee
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Korea; (G.J.); (S.L.)
| | - Joonho Hong
- Nano Bio Research Center, Jeonnam Bioindustry Foundation, Jangseong-si 57248, Jeollanam-do, Korea;
| | - Boram Park
- Department of Dental Hygiene, College of Health and Welfare, Kyungwoon University, Gumi-si 39160, Gyeongsangbuk-do, Korea;
| | - Dokyung Kim
- Oral Biology Research Institute, College of Dentistry, Chosun University, Gwangju 61452, Korea;
| | - Chunsung Kim
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Korea; (G.J.); (S.L.)
| |
Collapse
|
31
|
Murkar A, De Koninck J, Merali Z. Cannabinoids: Revealing their complexity and role in central networks of fear and anxiety. Neurosci Biobehav Rev 2021; 131:30-46. [PMID: 34487746 DOI: 10.1016/j.neubiorev.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022]
Abstract
The first aim of the present review is to provide an in-depth description of the cannabinoids and their known effects at various neuronal receptors. It reveals that cannabinoids are highly diverse, and recent work has highlighted that their effects on the central nervous system (CNS) are surprisingly more complex than previously recognized. Cannabinoid-sensitive receptors are widely distributed throughout the CNS where they act as primary modulators of neurotransmission. Secondly, we examine the role of cannabinoid receptors at key brain sites in the control of fear and anxiety. While our understanding of how cannabinoids specifically modulate these networks is mired by their complex interactions and diversity, a plausible framework(s) for their effects is proposed. Finally, we highlight some important knowledge gaps in our understanding of the mechanism(s) responsible for their effects on fear and anxiety in animal models and their use as therapeutic targets in humans. This is particularly important for our understanding of the phytocannabinoids used as novel clinical interventions.
Collapse
Affiliation(s)
- Anthony Murkar
- University of Ottawa Institute of Mental Health Research (IMHR), Ottawa, ON, Canada; School of Psychology, University of Ottawa, Ottawa, ON, Canada.
| | - Joseph De Koninck
- University of Ottawa Institute of Mental Health Research (IMHR), Ottawa, ON, Canada; School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Zul Merali
- School of Psychology, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Institute, Aga Khan University, Nairobi, Kenya; Carleton University, Neuroscience Department, Ottawa, ON, Canada
| |
Collapse
|
32
|
Eicosanoid receptors as therapeutic targets for asthma. Clin Sci (Lond) 2021; 135:1945-1980. [PMID: 34401905 DOI: 10.1042/cs20190657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022]
Abstract
Eicosanoids comprise a group of oxidation products of arachidonic and 5,8,11,14,17-eicosapentaenoic acids formed by oxygenases and downstream enzymes. The two major pathways for eicosanoid formation are initiated by the actions of 5-lipoxygenase (5-LO), leading to leukotrienes (LTs) and 5-oxo-6,8,11,14-eicosatetraenoic acid (5-oxo-ETE), and cyclooxygenase (COX), leading to prostaglandins (PGs) and thromboxane (TX). A third group (specialized pro-resolving mediators; SPMs), including lipoxin A4 (LXA4) and resolvins (Rvs), are formed by the combined actions of different oxygenases. The actions of the above eicosanoids are mediated by approximately 20 G protein-coupled receptors, resulting in a variety of both detrimental and beneficial effects on airway smooth muscle and inflammatory cells that are strongly implicated in asthma pathophysiology. Drugs targeting proinflammatory eicosanoid receptors, including CysLT1, the receptor for LTD4 (montelukast) and TP, the receptor for TXA2 (seratrodast) are currently in use, whereas antagonists of a number of other receptors, including DP2 (PGD2), BLT1 (LTB4), and OXE (5-oxo-ETE) are under investigation. Agonists targeting anti-inflammatory/pro-resolving eicosanoid receptors such as EP2/4 (PGE2), IP (PGI2), ALX/FPR2 (LXA4), and Chemerin1 (RvE1/2) are also being examined. This review summarizes the contributions of eicosanoid receptors to the pathophysiology of asthma and the potential therapeutic benefits of drugs that target these receptors. Because of the multifactorial nature of asthma and the diverse pathways affected by eicosanoid receptors, it will be important to identify subgroups of asthmatics that are likely to respond to any given therapy.
Collapse
|
33
|
Mustafa G, Cai CL, Bodkin D, Aranda JV, Beharry KD. Antioxidants and/or fish oil reduce intermittent hypoxia-induced inflammation in the neonatal rat terminal ileum. Prostaglandins Other Lipid Mediat 2021; 155:106565. [PMID: 34051366 DOI: 10.1016/j.prostaglandins.2021.106565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022]
Abstract
Intermittent hypoxia (IH) is associated with the pathogenesis of necrotizing enterocolitis (NEC). We tested the hypothesis that early supplementation with antioxidants and/or fish oil protects the terminal ileum from oxidative injury induced by neonatal IH. Newborn rats were exposed to neonatal IH from birth (P0) until P14 during which they received daily fish oil, coenzyme Q10 (CoQ10), glutathione nanoparticles (nGSH), fish oil + CoQ10, or olive oil. Pups were then placed in room air from P14 to P21 with no further supplementation. Terminal ileum was assessed for IH-induced injury and inflammatory biomarkers. Neonatal IH induced severe damage consistent with NEC, and was associated with oxidative stress and elevations in PGE2, PGF2α, TxB2, NOS-2 and TLR-4, effects that were ameliorated with nGSH and combination CoQ10+fish oil. Early postnatal supplementation with antioxidants and/or fish oil during neonatal IH may be favorable for preserving gut integrity and reducing oxidative injury.
Collapse
Affiliation(s)
- Ghassan Mustafa
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Darren Bodkin
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA; Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA; Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
| |
Collapse
|
34
|
O'Brien JB, Roman DL. Novel treatments for chronic pain: moving beyond opioids. Transl Res 2021; 234:1-19. [PMID: 33727192 DOI: 10.1016/j.trsl.2021.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023]
Abstract
It is essential that safe and effective treatment options be available to patients suffering from chronic pain. The emergence of an opioid epidemic has shaped public opinions and created stigmas surrounding the use of opioids for the management of pain. This reality, coupled with high risk of adverse effects from chronic opioid use, has led chronic pain patients and their healthcare providers to utilize nonopioid treatment approaches. In this review, we will explore a number of cellular reorganizations that are associated with the development and progression of chronic pain. We will also discuss the safety and efficacy of opioid and nonopioid treatment options for chronic pain. Finally, we will review the evidence for adenylyl cyclase type 1 (AC1) as a novel target for the treatment of chronic pain.
Collapse
Affiliation(s)
- Joseph B O'Brien
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa; Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
35
|
Antioxidant Serine-(NSAID) Hybrids with Anti-Inflammatory and Hypolipidemic Potency. Molecules 2021; 26:molecules26134060. [PMID: 34279399 PMCID: PMC8272148 DOI: 10.3390/molecules26134060] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
A series of L-serine amides of antioxidant acids, such as Trolox, (E)-3-(3,5-di-tert-butyl-4-hydroxyphenyl)acrylic acid (phenolic derivative of cinnamic acid) and 3,5-di-tert-butyl-4-hydroxybenzoic acid (structurally similar to butylated hydroxytoluene), was synthesized. The hydroxy group of serine was esterified with two classical NSAIDs, ibuprofen and ketoprofen. The Trolox derivatives with ibuprofen (7) and ketoprofen (10) were the most potent inhibitors of lipid peroxidation (IC50 3.4 μΜ and 2.8 μΜ), several times more potent than the reference Trolox (IC50 25 μΜ). Most of the compounds decreased carrageenan-induced rat paw edema (37–67% at 150 μmol/kg). They were moderate inhibitors of soybean lipoxygenase, with the exception of ibuprofen derivative 8 (IC50 13 μΜ). The most active anti-inflammatory compounds exhibited a significant decrease in lipidemic indices in the plasma of Triton-induced hyperlipidemic rats, e.g., the most active compound 9 decreased triglycerides, total cholesterol and low-density lipoprotein cholesterol by 52%, 61% and 70%, respectively, at 150 μmol/kg (i.p.), similar to that of simvastatin, a well-known hypocholesterolemic drug. Since the designed compounds seem to exhibit multiple pharmacological actions, they may be of use for the development of agents against inflammatory and degenerative conditions.
Collapse
|
36
|
Centuori SM, Caulin C, Bauman JE. Precision and Immunoprevention Strategies for Tobacco-Related Head and Neck Cancer Chemoprevention. Curr Treat Options Oncol 2021; 22:52. [PMID: 33991232 PMCID: PMC8122210 DOI: 10.1007/s11864-021-00848-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 12/02/2022]
Abstract
OPINION STATEMENT To date, there is no FDA-approved chemoprevention approach for tobacco-related HNSCC. Effective chemoprevention approaches validated in sufficiently powered randomized trials are needed to reduce the incidence and improve survival. In this review, we recap the challenges encountered in past chemoprevention trials and discuss emerging approaches, with major focus on green chemoprevention, precision prevention, and immunoprevention. As our current depth of knowledge expands in the arena of cancer immunotherapy, the field of immunoprevention is primed for new discoveries and successes in cancer prevention.
Collapse
Affiliation(s)
- Sara M. Centuori
- Department of Medicine, University of Arizona, 1515 N. Campbell Ave, PO Box 245024, Tucson, AZ 85724-5024 USA
- University of Arizona Cancer Center, 1515 N. Campbell Ave, Tucson, AZ 85724 USA
| | - Carlos Caulin
- University of Arizona Cancer Center, 1515 N. Campbell Ave, Tucson, AZ 85724 USA
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona, 1515 N. Campbell Ave, Tucson, AZ 85724 USA
| | - Julie E. Bauman
- Department of Medicine, University of Arizona, 1515 N. Campbell Ave, PO Box 245024, Tucson, AZ 85724-5024 USA
- University of Arizona Cancer Center, 1515 N. Campbell Ave, Tucson, AZ 85724 USA
| |
Collapse
|
37
|
Figueroa EG, Gonzaléz-Candia A, Villanueva CA, Ebensperger G, Reyes RV, Llanos AJ, Herrera EA. Beneficial effects of melatonin on prostanoids pathways in pulmonary hypertensive neonates. Vascul Pharmacol 2021; 138:106853. [PMID: 33766627 DOI: 10.1016/j.vph.2021.106853] [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: 09/28/2020] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/04/2023]
Abstract
Pulmonary arterial hypertension of the newborn (PAHN) is a syndrome caused by chronic hypoxia, characterized by decreased vasodilator function, a marked vasoconstrictor activity, proliferation of smooth muscle cells (SMC) and thickening of the extracellular matrix in the pulmonary circulation, among other characteristics. Prostaglandins are derived from the arachidonic acid (AA) metabolism and are important regulators of pulmonary vascular tone. Since hypoxia induces oxidative stress and has been related to PAHN, a postnatal treatment with melatonin has been proposed due to its antioxidant properties. Here, we determined the effects of melatonin on pulmonary vascular homeostasis given by prostanoids. Ten PAHN newborn lambs were divided in two groups and treated either with vehicle or melatonin. After 1 week of treatment, we assessed pulmonary vascular prostanoids function and expression by wire myography, RT-PCR, Western Blot and immunohistochemistry. Melatonin improved in vivo and ex vivo pulmonary vasodilation. This was associated with an increased function and expression of vasodilator prostanoids at the expense of vasoconstrictor prostanoids. Our study demonstrates for the first time that melatonin may enhance the vasodilator prostanoid pathway in PAHN.
Collapse
Affiliation(s)
- Esteban G Figueroa
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Alejandro Gonzaléz-Candia
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile; Institute of Health Sciences, University of O'Higgins, Rancagua, Chile
| | - Cristian A Villanueva
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Germán Ebensperger
- Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Roberto V Reyes
- Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Aníbal J Llanos
- Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Chile
| | - Emilio A Herrera
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile; Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Chile.
| |
Collapse
|
38
|
Eicosanoid blood vessel regulation in physiological and pathological states. Clin Sci (Lond) 2021; 134:2707-2727. [PMID: 33095237 DOI: 10.1042/cs20191209] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/26/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
Arachidonic acid can be metabolized in blood vessels by three primary enzymatic pathways; cyclooxygenase (COX), lipoxygenase (LO), and cytochrome P450 (CYP). These eicosanoid metabolites can influence endothelial and vascular smooth muscle cell function. COX metabolites can cause endothelium-dependent dilation or constriction. Prostaglandin I2 (PGI2) and thromboxane (TXA2) act on their respective receptors exerting opposing actions with regard to vascular tone and platelet aggregation. LO metabolites also influence vascular tone. The 12-LO metabolite 12S-hydroxyeicosatrienoic acid (12S-HETE) is a vasoconstrictor whereas the 15-LO metabolite 11,12,15-trihydroxyeicosatrienoic acid (11,12,15-THETA) is an endothelial-dependent hyperpolarizing factor (EDHF). CYP enzymes produce two types of eicosanoid products: EDHF vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-HETE. The less-studied cross-metabolites generated from arachidonic acid metabolism by multiple pathways can also impact vascular function. Likewise, COX, LO, and CYP vascular eicosanoids interact with paracrine and hormonal factors such as the renin-angiotensin system and endothelin-1 (ET-1) to maintain vascular homeostasis. Imbalances in endothelial and vascular smooth muscle cell COX, LO, and CYP metabolites in metabolic and cardiovascular diseases result in vascular dysfunction. Restoring the vascular balance of eicosanoids by genetic or pharmacological means can improve vascular function in metabolic and cardiovascular diseases. Nevertheless, future research is necessary to achieve a more complete understanding of how COX, LO, CYP, and cross-metabolites regulate vascular function in physiological and pathological states.
Collapse
|
39
|
Hadwen B, Stranges S, Barra L. Risk factors for hypertension in rheumatoid arthritis patients-A systematic review. Autoimmun Rev 2021; 20:102786. [PMID: 33609791 DOI: 10.1016/j.autrev.2021.102786] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Rheumatoid arthritis is frequently associated with hypertension, which has been shown to increase the risk of cardiovascular disease in these patients. The aim of this systematic review was to explore demographic, behavioural or clinical factors including medication use, associated with incident hypertension in rheumatoid arthritis. METHODS MEDLINE and Scopus were searched for eligible studies that longitudinally investigated incident hypertension or changes in blood pressure (BP) in rheumatoid arthritis patients. Publications were screened by two reviewers according to predetermined inclusion and exclusion criteria. The quality of included studies was assessed via the Newcastle Ottawa Scale and Cochrane Risk of Bias Tool. RESULTS Fourteen studies were deemed eligible and included in this review. The proportion of female subjects ranged from 12 to 87% and the mean age ranged from 47 to 61 years. Regular exercise was associated with a decrease in systolic BP, p = 0.021. Methotrexate was associated with decreased risk of hypertension in two studies. LEF was associated with increased BP in two studies. COX-2 inhibitors were associated with systolic BP and diastolic BP variability (p = 0.009, 0.039, respectively) in one study. Prednisone was found to increase BP and risk of hypertension in three studies. The risk of hypertension in patients taking biologic disease modifying anti-rheumatic drugs (DMARDs) is unclear as some studies report increased BP while others report no difference for biologic compared to conventional DMARDs. CONCLUSION Despite limited longitudinal studies exploring this topic, methotrexate and exercise were shown to protect against risk of hypertension in RA patients, while prednisone and COX-2 inhibitors may increase risk of hypertension.
Collapse
Affiliation(s)
- Brook Hadwen
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, Canada
| | - Saverio Stranges
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, Canada; Department of Family Medicine, The University of Western Ontario, London, Canada; Lawson Health Research Institute, London, Canada; Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Lillian Barra
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, Canada; Lawson Health Research Institute, London, Canada; Department of Medicine, Division of Rheumatology, The University of Western Ontario, London, Canada.
| |
Collapse
|
40
|
Mitchell JA, Kirkby NS, Ahmetaj-Shala B, Armstrong PC, Crescente M, Ferreira P, Lopes Pires ME, Vaja R, Warner TD. Cyclooxygenases and the cardiovascular system. Pharmacol Ther 2021; 217:107624. [DOI: 10.1016/j.pharmthera.2020.107624] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
|
41
|
Mena M, Dalbah L, Levi L, Padilla M, Enciso R. Efficacy of topical interventions for temporomandibular disorders compared to placebo or control therapy: a systematic review with meta-analysis. J Dent Anesth Pain Med 2020; 20:337-356. [PMID: 33409363 PMCID: PMC7783375 DOI: 10.17245/jdapm.2020.20.6.337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/23/2020] [Accepted: 11/23/2020] [Indexed: 01/19/2023] Open
Abstract
This systematic review focused on the efficacy of topical products in reducing temporomandibular joint disorder (TMD)-associated pain, in comparison to placebo or control interventions. The EMBASE, Web of Science, Cochrane Library, and MEDLINE via PubMed databases were searched for randomized controlled trials (RCTs) using topical interventions in adults diagnosed with TMD. The pain intensity was the primary outcome, and other clinical findings were the secondary outcomes. The risk of bias was evaluated according to the Cochrane's handbook. The search up to February 7, 2020 identified a total of 496 unduplicated references. Nine RCTs with 355 adult patients diagnosed with TMD were included. The meta-analysis did not show a significant reduction in baseline pain intensity in the nonsteroidal anti-inflammatory drug (NSAIDs) group, when compared to the placebo group (P = 0.288). One study demonstrated a statistically significant pain score decrease for Theraflex-TMJ compared to placebo after 10 d of treatment (P = 0.003) and follow-up, 5 d after the last application (P = 0.027). Ping On reduced pain at 4 weeks of application (P < 0.001) but not after 7 d of application (P = 0.136). In one study, cannabidiol (CBD) significantly improved the pain intensity compared to placebo (P < 0.001). However, no differences were found with capsaicin in the two studies (P = 0.465). Evidence was of low quality because the studies were considered as having an unclear or a high risk of bias and a small number of studies were analyzed. The evidence is not sufficient to support the use of topical NSAIDs and capsaicin, and limited evidence was found for Threraflex-TMJ, bee venom, Ping On, and CBD, with only one study reporting for each. Additional studies are recommended to validate these results.
Collapse
Affiliation(s)
- Mariam Mena
- Master of Science Program in Orofacial Pain and Oral Medicine, Herman Ostrow School of Dentistry of USC, Los Angeles, California, USA
| | - Lana Dalbah
- Master of Science Program in Orofacial Pain and Oral Medicine, Herman Ostrow School of Dentistry of USC, Los Angeles, California, USA
- Post-graduate Orthodontic Department, European University College, Dubai, United Arab Emirates
| | - Lauren Levi
- Master of Science Program in Orofacial Pain and Oral Medicine, Herman Ostrow School of Dentistry of USC, Los Angeles, California, USA
- New York University School of Dentistry, New York, New York, USA
| | - Mariela Padilla
- Assistant Director of Online Programs, Herman Ostrow School of Dentistry of USC, Los Angeles, California, USA
| | - Reyes Enciso
- Division of Dental Public Health and Pediatric Dentistry, Herman Ostrow School of Dentistry of University of Southern California, Los Angeles, California, USA
| |
Collapse
|
42
|
Eastman CL, D'Ambrosio R, Ganesh T. Modulating neuroinflammation and oxidative stress to prevent epilepsy and improve outcomes after traumatic brain injury. Neuropharmacology 2020; 172:107907. [PMID: 31837825 PMCID: PMC7274911 DOI: 10.1016/j.neuropharm.2019.107907] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in young adults worldwide. TBI survival is associated with persistent neuropsychiatric and neurological impairments, including posttraumatic epilepsy (PTE). To date, no pharmaceutical treatment has been found to prevent PTE or ameliorate neurological/neuropsychiatric deficits after TBI. Brain trauma results in immediate mechanical damage to brain cells and blood vessels that may never be fully restored given the limited regenerative capacity of brain tissue. This primary insult unleashes cascades of events, prominently including neuroinflammation and massive oxidative stress that evolve over time, expanding the brain injury, but also clearing cellular debris and establishing homeostasis in the region of damage. Accumulating evidence suggests that oxidative stress and neuroinflammatory sequelae of TBI contribute to posttraumatic epileptogenesis. This review will focus on possible roles of reactive oxygen species (ROS), their interactions with neuroinflammation in posttraumatic epileptogenesis, and emerging therapeutic strategies after TBI. We propose that inhibitors of the professional ROS-generating enzymes, the NADPH oxygenases and myeloperoxidase alone, or combined with selective inhibition of cyclooxygenase mediated signaling may have promise for the treatment or prevention of PTE and other sequelae of TBI. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Clifford L Eastman
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA.
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA; Regional Epilepsy Center, University of Washington, 325 Ninth Ave., Seattle, WA, 98104, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, Georgia.
| |
Collapse
|
43
|
Rouzer CA, Marnett LJ. Structural and Chemical Biology of the Interaction of Cyclooxygenase with Substrates and Non-Steroidal Anti-Inflammatory Drugs. Chem Rev 2020; 120:7592-7641. [PMID: 32609495 PMCID: PMC8253488 DOI: 10.1021/acs.chemrev.0c00215] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclooxgenases are key enzymes of lipid signaling. They carry out the first step in the production of prostaglandins, important mediators of inflammation, pain, cardiovascular disease, and cancer, and they are the molecular targets for nonsteroidal anti-inflammatory drugs, which are among the oldest and most chemically diverse set of drugs known. Homodimeric proteins that behave as allosterically modulated, functional heterodimers, the cyclooxygenases exhibit complex kinetic behavior, requiring peroxide-dependent activation and undergoing suicide inactivation. Due to their important physiological and pathophysiological roles and keen interest on the part of the pharmaceutical industry, the cyclooxygenases have been the focus of a vast array of structural studies, leading to the publication of over 80 crystal structures of the enzymes in complex with substrates or inhibitors supported by a wealth of functional data generated by site-directed mutation experiments. In this review, we explore the chemical biology of the cyclooxygenases through the lens of this wealth of structural and functional information. We identify key structural features of the cyclooxygenases, break down their active site into regional binding pockets to facilitate comparisons between structures, and explore similarities and differences in the binding modes of the wide variety of ligands (both substrates and inhibitors) that have been characterized in complex with the enzymes. Throughout, we correlate structure with function whenever possible. Finally, we summarize what can and cannot be learned from the currently available structural data and discuss the critical intriguing questions that remain despite the wealth of information that has been amassed in this field.
Collapse
Affiliation(s)
- Carol A Rouzer
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J Marnett
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
44
|
Dillon KM, Carrazzone RJ, Matson JB, Kashfi K. The evolving landscape for cellular nitric oxide and hydrogen sulfide delivery systems: A new era of customized medications. Biochem Pharmacol 2020; 176:113931. [PMID: 32224139 PMCID: PMC7263970 DOI: 10.1016/j.bcp.2020.113931] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/20/2020] [Indexed: 02/09/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are industrial toxins or pollutants; however, both are produced endogenously and have important biological roles in most mammalian tissues. The recognition that these gasotransmitters have a role in physiological and pathophysiological processes has presented opportunities to harness their intracellular effects either through inhibition of their production; or more commonly, through inducing their levels and or delivering them by various modalities. In this review article, we have focused on an array of NO and H2S donors, their hybrids with other established classes of drugs, and the various engineered delivery platforms such a fibers, polymers, nanoparticles, hydrogels, and others. In each case, we have reviewed the rationale for their development.
Collapse
Affiliation(s)
- Kearsley M Dillon
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, USA; Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA
| | - Ryan J Carrazzone
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, USA; Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA
| | - John B Matson
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, USA; Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, 160 Convent Avenue, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, NY, USA.
| |
Collapse
|
45
|
Javaherforooshzadeh F, Abdalbeygi H, Janatmakan F, Gholizadeh B. Comparing the effects of ketorolac and Paracetamol on postoperative pain relief after coronary artery bypass graft surgery. A randomized clinical trial. J Cardiothorac Surg 2020; 15:80. [PMID: 32393370 PMCID: PMC7216617 DOI: 10.1186/s13019-020-01125-y] [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: 02/05/2020] [Accepted: 04/28/2020] [Indexed: 11/30/2022] Open
Abstract
Introduction Pain management after coronary artery bypass graft (CABG) surgery remains challenging. Objective This study aimed to compare the effects of Ketorolac and Paracetamol on postoperative CABG pain relief. Method This double-blind randomized clinical trial study was conducted in Ahvaz, Iran, from September 2018–December 2019. Two consecutive groups of 60 patients undergoing elective on-pump coronary artery bypass graft surgery. Intervention The patients were divided into 0.5 mg/kg of ketorolac mg/dl and 10 mg/kg of Paracetamol after surgery for pain management. Primary outcomes were: visual analog pain scale (VAS) at the time point immediately after extubation (baseline) and at 6, 12, 24 and 48 h and the total dose of morphine consumption. Secondary outcomes included the hemodynamic variables, weaning time, chest tube derange, in-hospital mortality and myocardial infarction. Statistical analysis: The data were analyzed using SPSS version 22(SPSS, Chicago, IL). The Mann-Whitney U-test was used to compare demographic data, VAS scores, vital signs, and side effects. Repeated measurements were tested within groups using Friedman’s ANOVA and the Wilcoxon rank-sum test. Values were expressed as means ± standard deviations. Statistical significance was defined as a p-value < 0.05. Results Compared with baseline scores, there were significant declines in VAS scores in both groups throughout the time sequence (P< 0.05). The statistical VAS score was slightly higher in the Paracetamol group at most time points, except for the time of 6 h. However, at 24 and 48 h, the VAS score in group Paracetamol was significantly higher than in group Ketorolac. There were no significant differences between groups about hemodynamic variables. Conclusion The efficacy of ketorolac is comparable to that of Paracetamol in postoperative CABG pain relief. Trial registry IRCT20150216021098N5. Registered at 2019-09-12.
Collapse
Affiliation(s)
- Fatemeh Javaherforooshzadeh
- Department of Cardiac Anesthesia, Ahvaz Anesthesiology and Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Hasan Abdalbeygi
- Department of Anesthesia, Ahvaz Anesthesiology and Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farahzad Janatmakan
- Department of Anesthesia, Ahvaz Anesthesiology and Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Behnam Gholizadeh
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
46
|
Arora M, Choudhary S, Singh PK, Sapra B, Silakari O. Structural investigation on the selective COX-2 inhibitors mediated cardiotoxicity: A review. Life Sci 2020; 251:117631. [PMID: 32251635 DOI: 10.1016/j.lfs.2020.117631] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/31/2020] [Indexed: 01/30/2023]
Abstract
Initially, the selective COX-2 inhibitors were developed as safer alternatives to the conventional NSAIDs, but later on, most of them were withdrawn from the market due to the risk of heart attack and stroke. Celecoxib, the first selective COX-2 inhibitor, was approved by the Food and Drug Administration (FDA) in December 1998 and was taken back from the market in 2004. Since then, many coxibs have been discontinued one by one due to adverse cardiovascular events. United States (US), Australian and European authorities related to Therapeutic Goods Administration (TGA) implemented the requirements to carry the "Black box" warning on the labels of COX-2 drugs highlighting the risks of serious cardiovascular events. These facts encouraged the researchers to explore them well and find out the biochemical basis behind the cardiotoxicity. From the last few decades, the molecular mechanisms behind the coxibs have regained the attention, especially the specific structural features of the selective COX-2 inhibitors that are associated with cardiotoxicity. This review discusses the key structural features of the selective COX-2 inhibitors and underlying mechanisms that are responsible for the cardiotoxicity. This report also unfolds different strategies that have been reported in the last 10 years to combat the problem of selective COX-2 inhibitors mediated cardiotoxicity.
Collapse
Affiliation(s)
- Mohit Arora
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Shalki Choudhary
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Pankaj Kumar Singh
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Bharti Sapra
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Om Silakari
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India.
| |
Collapse
|
47
|
Effects of the α2/α3-subtype-selective GABAA receptor positive allosteric modulator KRM-II-81 on pain-depressed behavior in rats: comparison with ketorolac and diazepam. Behav Pharmacol 2020; 30:452-461. [PMID: 30640180 DOI: 10.1097/fbp.0000000000000464] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This study examined effects of the α2/α3-subtype-selective GABAA receptor positive allosteric modulator KRM-II-81 in an assay of pain-related behavioral depression. Adult, male Sprague-Dawley rats responded for electrical brain stimulation in a frequency-rate intracranial self-stimulation (ICSS) procedure. Intraperitoneal injection of 1.8% lactic acid served as an acute noxious stimulus to depress ICSS. Effects of KRM-II-81 were evaluated in the absence and presence of the acid noxious stimulus. The NSAID ketorolac and the benzodiazepine diazepam were tested as comparators. Neither ketorolac nor KRM-II-81 altered ICSS in the absence of the acid noxious stimulus; however, diazepam produced facilitation consistent with its abuse liability. Ketorolac blocked acid-induced depression of ICSS, and effects of 1.0 mg/kg ketorolac lasted for at least 5 h. KRM-II-81 (1.0 mg/kg) produced significant antinociception after 30 min that dissipated by 60 min. Diazepam also attenuated acid-depressed ICSS, but only at doses that facilitated ICSS when administered alone. The lack of ketorolac or KRM-II-81 effects on ICSS in the absence of the acid noxious stimulus suggests low abuse liability for both compounds. The effectiveness of ketorolac to block acid-induced ICSS depression agrees with clinical analgesic efficacy of ketorolac. KRM-II-81 produced significant but less consistent and shorter-acting antinociception than ketorolac.
Collapse
|
48
|
Shaikh NF, Sambamoorthi U. Prescription Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) and Depression among Adults with Inflammatory Chronic Conditions in the United States. Psychiatr Q 2020; 91:209-221. [PMID: 31811581 PMCID: PMC7134606 DOI: 10.1007/s11126-019-09693-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The association of prescription NSAIDs to presence of depression among adults with inflammatory chronic conditions in adults with and without depression and seeking care in routine clinical practice remains unknown. We examined the association of prescription NSAIDs to depression among adults with inflammatory chronic conditions in a nationally representative sample of the US non-institutionalized civilian population. We used a retrospective cross-sectional design. Data on 10,713 adults with inflammatory chronic conditions were derived from 2015 Medical Expenditure Panel Survey (MEPS). The dependent variable was the presence or absence of depression and the key independent variable was prescription NSAIDs use. Logistic regression models were used to examine the adjusted associations of prescription NSAIDs to depression. In these regressions, other independent variables (biological, sociocultural, socio-economic, access to healthcare services, medical conditions and treatment factors, behavioural, and environmental factors) that may affect the relationship of prescription NSAIDs to depression were also included. All analyses accounted for the complex survey design of MEPS. Overall, 18.2% reported depression. Almost 21% used prescription NSAIDs. In the unadjusted model, prescription NSAIDs use had higher odds of depression (OR = 1.59;95%CI = [1.40, 1.82]) as compared to those without NSAIDs. In the fully-adjusted logistic regression model, with controls for other independent variables, adults using prescription NSAIDs had no significant association with depression (AOR = 0.97;95%CI = [0.84, 1.13]) compared to those without NSAIDs. In this first real-world study of all adults (with and without depression) in the US, we did not observe a statistically significant association of prescription NSAIDs to the presence of depression.
Collapse
Affiliation(s)
- Nazneen Fatima Shaikh
- Department of Pharmaceutical Systems and Policy, West Virginia University School of Pharmacy, Robert C. Byrd Health Sciences Center [North], P.O. Box 9510, Morgantown, WV, 26506-9510, USA.
| | - Usha Sambamoorthi
- Department of Pharmaceutical Systems and Policy, West Virginia University School of Pharmacy, Robert C. Byrd Health Sciences Center [North], P.O. Box 9510, Morgantown, WV, 26506-9510, USA
| |
Collapse
|
49
|
Hernandez-Sanabria E, Heiremans E, Calatayud Arroyo M, Props R, Leclercq L, Snoeys J, Van de Wiele T. Short-term supplementation of celecoxib-shifted butyrate production on a simulated model of the gut microbial ecosystem and ameliorated in vitro inflammation. NPJ Biofilms Microbiomes 2020; 6:9. [PMID: 32075981 PMCID: PMC7031363 DOI: 10.1038/s41522-020-0119-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/22/2020] [Indexed: 12/25/2022] Open
Abstract
Celecoxib has been effective in the prevention and treatment of chronic inflammatory disorders through inhibition of altered cyclooxygenase-2 (COX-2) pathways. Despite the benefits, continuous administration may increase risk of cardiovascular events. Understanding microbiome-drug-host interactions is fundamental for improving drug disposition and safety responses of colon-targeted formulations, but little information is available on the bidirectional interaction between individual microbiomes and celecoxib. Here, we conducted in vitro batch incubations of human faecal microbiota to obtain a mechanistic proof-of-concept of the short-term impact of celecoxib on activity and composition of colon bacterial communities. Celecoxib-exposed microbiota shifted metabolic activity and community composition, whereas total transcriptionally active bacterial population was not significantly changed. Butyrate production decreased by 50% in a donor-dependent manner, suggesting that celecoxib impacts in vitro fermentation. Microbiota-derived acetate has been associated with inhibition of cancer markers and our results suggest uptake of acetate for bacterial functions when celecoxib was supplied, which potentially favoured bacterial competition for acetyl-CoA. We further assessed whether colon microbiota modulates anti-inflammatory efficacy of celecoxib using a simplified inflammation model, and a novel in vitro simulation of the enterohepatic metabolism. Celecoxib was responsible for only 5% of the variance in bacterial community composition but celecoxib-exposed microbiota preserved barrier function and decreased concentrations of IL-8 and CXCL16 in a donor-dependent manner in our two models simulating gut inflammatory milieu. Our results suggest that celecoxib-microbiome-host interactions may not only elicit adaptations in community composition but also in microbiota functionality, and these may need to be considered for guaranteeing efficient COX-2 inhibition.
Collapse
Affiliation(s)
- Emma Hernandez-Sanabria
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
| | - Evelien Heiremans
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Marta Calatayud Arroyo
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Ruben Props
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Laurent Leclercq
- Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, Antwerp, 2340, Belgium
| | - Jan Snoeys
- Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, Antwerp, 2340, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
| |
Collapse
|
50
|
Toda K, Ueyama M, Tanaka S, Tsukayama I, Mega T, Konoike Y, Tamenobu A, Bastian F, Akai I, Ito H, Kawakami Y, Takahashi Y, Suzuki-Yamamoto T. Ellagitannins from Punica granatum leaves suppress microsomal prostaglandin E synthase-1 expression and induce lung cancer cells to undergo apoptosis. Biosci Biotechnol Biochem 2019; 84:757-763. [PMID: 31868102 DOI: 10.1080/09168451.2019.1706442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Prostaglandin E2 (PGE2), which is a potent pro-inflammatory lipid mediator, is biosynthesized from arachidonic acid by cyclooxygenase-2 (COX-2) and microsomal PGE synthase-1 (mPGES-1). Non-steroidal anti-inflammatory drugs (NSAIDs) are used clinically as COX inhibitors, but they have gastrointestinal and cardiovascular side-effects. Thus, the terminal enzyme mPGES-1 holds promise as the next therapeutic target. In this study, we found that the ellagitannins granatin A and granatin B isolated from pomegranate leaves, and geraniin, which is their structural analog, selectively suppressed mPGES-1 expression without affecting COX-2 in non-small cell lung carcinoma A549 cells. The ellagitannins also down-regulated tumor necrosis factor α, inducible nitric oxide synthase, and anti-apoptotic factor B-cell chronic lymphocytic leukemia/lymphoma 2, and induced A549 cells to undergo apoptosis. These findings indicate that the ellagitannins have anti-inflammatory and anti-carcinogenic effects, due to their specific suppression of mPGES-1.Abbreviations: Bcl-2: B-cell chronic lymphocytic leukemia/lymphoma 2; COX: cyclooxygenase; CRE: cAMP response element; DHHDP: dehydrohexahydroxydiphenoyl; Et2O: diethyl ether; EtOAc: ethyl acetate; GAPDH: glyceraldehyde 3-phosphate dehydrogenase; iNOS: inducible nitric oxide synthase; mPGES-1: microsomal prostaglandin E synthase-1; n-BuOH: water-saturated n-butanol; NSAIDs: non-steroidal anti-inflammatory drugs; NF-κB: nuclear factor-κB; PG: prostaglandin; TNF: tumor necrosis factor; TUNEL: terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling.
Collapse
Affiliation(s)
- Keisuke Toda
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Mai Ueyama
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Shomu Tanaka
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Izumi Tsukayama
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Takuto Mega
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Yuka Konoike
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Asako Tamenobu
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Februadi Bastian
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Iria Akai
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Hideyuki Ito
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Yuki Kawakami
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | - Yoshitaka Takahashi
- Department of Nutritional Science, Okayama Prefectural University, Okayama, Japan
| | | |
Collapse
|