101
|
Perrone MG, Miciaccia M, Vitale P, Ferorelli S, Araújo CDCB, de Almeida GS, Souza Domingos TF, da Silva LCRP, de Pádula M, Cabral LM, Sathler PC, Bonaccorso C, Fortuna CG, Scilimati A. An attempt to chemically state the cross-talk between monomers of COX homodimers by double/hybrid inhibitors mofezolac-spacer-mofezolac and mofezolac-spacer-arachidonic acid. Eur J Med Chem 2020; 209:112919. [PMID: 33129592 DOI: 10.1016/j.ejmech.2020.112919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases (CVDs) account for over 17 million death globally each year, including arterial thrombosis. Platelets are key components in the pathogenesis of this disease and modulating their activity is an effective strategy to treat such thrombotic events. Cyclooxygenase-1 (COX-1) isoenzyme is involved in platelet activation and is the main target of non-steroidal anti-inflammatory drugs (NSAIDs) and new selective inhibitor research. Inhibitors of general formula mofezolac-spacer-mofezolac (mof-spacer-mof) and mofezolac-spacer-arachidonic acid (mof-spacer-AA) were projected to investigate the possible cross-talk between the two monomers (Eallo and Ecat) forming the COX-1 homodimer. Mofezolac was chosen as either one or two moieties of these molecules being the known most potent and selective COX-1 inhibitor and administrated to humans as Disopain™, then arachidonic acid (AA) was used to develop molecules bearing, in the same compound, in addition to the inhibitor moiety (mofezolac) also the natural COX substrate. Depending on the nature of the spacer, COX-1 and COX-2 activity was differently inhibited by mof-spacer-mof set with a preferential COX-1 inhibition. The highest COX-1 selectivity was exhibited by the compound in which the spacer was the benzidine [N,N'-(biphenyl-4,4'-di-yl)bis (2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamide) (15): COX-1 IC50 = 0.08 μM, COX-2 IC50 > 50 μM, Selectivity Index (SI) > 625]. In the case of mof-spacer-AA set, the COX inhibitory potency and also the isoform preference changed. (5Z, 8Z, 11Z, 14Z)-N-(4-{2-[3,4-Bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}butyl)icosa-5,8,11,14-tetraenamide (19) and (5Z, 8Z, 11Z, 14Z)-N-(4'-{2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}-[1,1'-biphenyl]-4-yl)icosa-5,8,11,14-tetraenamide (21), in which the spacer is the 1,2-diaminobutane or benzidine, respectively, selectively inhibited the COX-2, whereas when the spacer is the 1,4-phenylendiamine [(5Z, 8Z, 11Z, 14Z)-N-(4-{2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}phenyl)icosa-5,8,11,14-tetraenamide) (20) the COX preference is COX-1 (COX-1 IC50 = 0.05 μM, COX-2 IC50 > 50 μM, with a COX-1 selectivity > 1000). Molecular modelling by using FLAP algorithm shows fundamental interactions of the novel compounds at the entry channel of COX and inside its catalytic cavity. The effect of these mof-spacer-mof and mof-spacer-AA in inhibiting in vitro free arachidonic acid-induced platelet aggregation was also determined. A positive profile of hemocompatibility in relation to their influence on the blood coagulation cascade and erythrocyte toxicity was observed. Cytotoxicity and genotoxicity safety were also found for these two novel sets of compounds.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Morena Miciaccia
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Paola Vitale
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Cristina da Costa Bernardes Araújo
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Gabriella Silva de Almeida
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Thaisa Francielle Souza Domingos
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | | | - Marcelo de Pádula
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Plínio Cunha Sathler
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Carmela Bonaccorso
- Department of Chemical Science, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Cosimo G Fortuna
- Department of Chemical Science, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Antonio Scilimati
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
102
|
Bindu S, Mazumder S, Bandyopadhyay U. Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: A current perspective. Biochem Pharmacol 2020; 180:114147. [PMID: 32653589 PMCID: PMC7347500 DOI: 10.1016/j.bcp.2020.114147] [Citation(s) in RCA: 659] [Impact Index Per Article: 164.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Owing to the efficacy in reducing pain and inflammation, non-steroidal anti-inflammatory drugs (NSAIDs) are amongst the most popularly used medicines confirming their position in the WHO's Model List of Essential Medicines. With escalating musculoskeletal complications, as evident from 2016 Global Burden of Disease data, NSAID usage is evidently unavoidable. Apart from analgesic, anti-inflammatory and antipyretic efficacies, NSAIDs are further documented to offer protection against diverse critical disorders including cancer and heart attacks. However, data from multiple placebo-controlled trials and meta-analyses studies alarmingly signify the adverse effects of NSAIDs in gastrointestinal, cardiovascular, hepatic, renal, cerebral and pulmonary complications. Although extensive research has elucidated the mechanisms underlying the clinical hazards of NSAIDs, no review has extensively collated the outcomes on various multiorgan toxicities of these drugs together. In this regard, the present review provides a comprehensive insight of the existing knowledge and recent developments on NSAID-induced organ damage. It precisely encompasses the current understanding of structure, classification and mode of action of NSAIDs while reiterating on the emerging instances of NSAID drug repurposing along with pharmacophore modification aimed at safer usage of NSAIDs where toxic effects are tamed without compromising the clinical benefits. The review does not intend to vilify these 'wonder drugs'; rather provides a careful understanding of their side-effects which would be beneficial in evaluating the risk-benefit threshold while rationally using NSAIDs at safer dose and duration.
Collapse
Affiliation(s)
- Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal 736101 India
| | - Somnath Mazumder
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Uday Bandyopadhyay
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India; Division of Molecular Medicine, Bose Institute, P-1/12, CIT Rd, Scheme VIIM, Kankurgachi, Kolkata, West Bengal 700054 India.
| |
Collapse
|
103
|
Fang C, Schmaier AH. Novel anti-thrombotic mechanisms mediated by Mas receptor as result of balanced activities between the kallikrein/kinin and the renin-angiotensin systems. Pharmacol Res 2020; 160:105096. [PMID: 32712319 PMCID: PMC7378497 DOI: 10.1016/j.phrs.2020.105096] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/17/2022]
Abstract
The risk of thrombosis, a globally growing challenge and a major cause of death, is influenced by various factors in the intravascular coagulation, vessel wall, and cellular systems. Among the contributors to thrombosis, the contact activation system and the kallikrein/kinin system, two overlapping plasma proteolytic systems that are often considered as synonymous, regulate thrombosis from different aspects. On one hand, components of the contact activation system such as factor XII initiates activation of the coagulation proteins promoting thrombus formation on artificial surfaces through factor XI- and possibly prekallikrein-mediated intrinsic coagulation. On the other hand, physiological activation of plasma prekallikrein in the kallikrein/kinin system on endothelial cells liberates bradykinin from associated high-molecular-weight kininogen to stimulate the constitutive bradykinin B2 receptor to generate nitric oxide and prostacyclin to induce vasodilation and counterbalance angiotensin II signaling from the renin-angiotensin system which stimulates vasoconstriction. In addition to vascular tone regulation, this interaction between the kallikrein/kinin and renin-angiotensin systems has a thrombo-regulatory role independent of the contact pathway. At the level of the G-protein coupled receptors of these systems, defective bradykinin signaling due to attenuated bradykinin formation and/or decreased B2 receptor expression, as seen in murine prekallikrein and B2 receptor null mice, respectively, leads to compensatory overexpressed Mas, the receptor for angiotensin-(1-7) of the renin-angiotensin system. Mas stimulation and/or its increased expression contributes to maintaining a healthy vascular homeostasis by generating graded elevation of plasma prostacyclin which reduces thrombosis through two independent pathways: (1) increasing the vasoprotective transcription factor Sirtuin 1 to suppress tissue factor expression, and (2) inhibiting platelet activation. This review will summarize the recent advances in this field that support these understandings. Appreciating these subtle mechanisms help to develop novel anti-thrombotic strategies by targeting the vascular receptors in the renin-angiotensin and the kallikrein/kinin systems to maintain healthy vascular homeostasis.
Collapse
Affiliation(s)
- Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology and the Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, 430030, China.
| | - Alvin H. Schmaier
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
104
|
Kirkby NS, Raouf J, Ahmetaj-Shala B, Liu B, Mazi SI, Edin ML, Chambers MG, Korotkova M, Wang X, Wahli W, Zeldin DC, Nüsing R, Zhou Y, Jakobsson PJ, Mitchell JA. Mechanistic definition of the cardiovascular mPGES-1/COX-2/ADMA axis. Cardiovasc Res 2020; 116:1972-1980. [PMID: 31688905 PMCID: PMC7519887 DOI: 10.1093/cvr/cvz290] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/23/2019] [Accepted: 10/31/2019] [Indexed: 02/05/2023] Open
Abstract
AIMS Cardiovascular side effects caused by non-steroidal anti-inflammatory drugs (NSAIDs), which all inhibit cyclooxygenase (COX)-2, have prevented development of new drugs that target prostaglandins to treat inflammation and cancer. Microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors have efficacy in the NSAID arena but their cardiovascular safety is not known. Our previous work identified asymmetric dimethylarginine (ADMA), an inhibitor of endothelial nitric oxide synthase, as a potential biomarker of cardiovascular toxicity associated with blockade of COX-2. Here, we have used pharmacological tools and genetically modified mice to delineate mPGES-1 and COX-2 in the regulation of ADMA. METHODS AND RESULTS Inhibition of COX-2 but not mPGES-1 deletion resulted in increased plasma ADMA levels. mPGES-1 deletion but not COX-2 inhibition resulted in increased plasma prostacyclin levels. These differences were explained by distinct compartmentalization of COX-2 and mPGES-1 in the kidney. Data from prostanoid synthase/receptor knockout mice showed that the COX-2/ADMA axis is controlled by prostacyclin receptors (IP and PPARβ/δ) and the inhibitory PGE2 receptor EP4, but not other PGE2 receptors. CONCLUSION These data demonstrate that inhibition of mPGES-1 spares the renal COX-2/ADMA pathway and define mechanistically how COX-2 regulates ADMA.
Collapse
Affiliation(s)
- Nicholas S Kirkby
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| | - Joan Raouf
- Unit of Rheumatology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Blerina Ahmetaj-Shala
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| | - Bin Liu
- Cardiovascular Research Centre, Shantou University Medical College, Shantou, China
| | - Sarah I Mazi
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
- King Fahad Cardiac Center, King Saud University, Riyadh, Saudi Arabia
| | - Matthew L Edin
- National Institute for Environmental Health Sciences, Durham, NC, USA
| | | | - Marina Korotkova
- Unit of Rheumatology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xiaomeng Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology & Research, Singapore, Singapore
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, UK
- Singapore Eye Research Institute, Singapore, Singapore
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Darryl C Zeldin
- National Institute for Environmental Health Sciences, Durham, NC, USA
| | - Rolf Nüsing
- Clinical Pharmacology and Pharmacotherapy Department, Goethe University, Frankfurt, Germany
| | - Yingbi Zhou
- Cardiovascular Research Centre, Shantou University Medical College, Shantou, China
| | - Per-Johan Jakobsson
- Unit of Rheumatology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Jane A Mitchell
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
105
|
Mora-Ramiro B, Jiménez-Estrada M, Zentella-Dehesa A, Ventura-Gallegos JL, Gomez-Quiroz LE, Rosiles-Alanis W, Alarcón-Aguilar FJ, Almanza-Pérez JC. Cacalol Acetate, a Sesquiterpene from Psacalium decompositum, Exerts an Anti-inflammatory Effect through LPS/NF-KB Signaling in Raw 264.7 Macrophages. JOURNAL OF NATURAL PRODUCTS 2020; 83:2447-2455. [PMID: 32672964 DOI: 10.1021/acs.jnatprod.0c00300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Inflammatory diseases remain critical health problems worldwide. The search for anti-inflammatory drugs is a primary activity in the pharmaceutical industry. Cacalol is a sesquiterpene with anti-inflammatory potential that is isolated from Psacalium decompositum, a medicinal plant with several scientific reports supporting its anti-inflammatory activity. Cacalol acetate (CA) is the most stable form. Nevertheless, the participation of CA in the main signaling pathway associated with inflammation is unknown. Our aim was to study the anti-inflammatory effect of CA and to determine its participation in NF-κB signaling. In TPA-induced edema in mice, CA produced 70.3% inhibition. To elucidate the influence of CA on the NF-κB pathway, RAW 264.7 macrophages were pretreated with CA and then stimulated with LPS, evaluating NF-ΚB activation, IKK phosphorylation, IΚB-α, p65, cytokine expression, and COX-2 release and activity. CA inhibited NF-κB activation and its upstream signaling, decreasing phosphorylation IKB-α and p65 levels. CA also reduced expression and secretion of TNF-α, IL-1β, and IL-6. Additionally, it decreased the activity and expression of COX-2 mRNA. These data support that CA regulates the NF-κB signaling pathway, which might explain, at least in part, its anti-inflammatory effect. CA is a bioactive molecule useful for the development of anti-inflammatory agents with innovative mechanisms of action.
Collapse
Affiliation(s)
- B Mora-Ramiro
- Posgrado en Biología Experimental, Division de CBS, UAM-Iztapalapa., San Rafael Atlixco 186, Vicentina, Delegación Iztapalapa, C.P. 09340, Ciudad de México, México
| | - M Jiménez-Estrada
- Departamento de Productos Naturales, Instituto de Química, UNAM, Circuito, Mario de La Cueva s/n, C.U., 04510 Ciudad de México, México
| | - A Zentella-Dehesa
- Programa Institucional de Cáncer de Mama, Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Circuito, Mario de La Cueva s/n, C.U., 04510 Ciudad de México, México
- Unidad de Bioquímica, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, México
| | - J L Ventura-Gallegos
- Programa Institucional de Cáncer de Mama, Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Circuito, Mario de La Cueva s/n, C.U., 04510 Ciudad de México, México
- Unidad de Bioquímica, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, México
| | - L E Gomez-Quiroz
- Departamento de Ciencias de la Salud, CBS, UAM-Iztapalapa, San Rafael Atlixco 186, Vicentina, Delegación Iztapalapa, C.P. 09340, Ciudad de México, México
| | - W Rosiles-Alanis
- Posgrado en Biología Experimental, Division de CBS, UAM-Iztapalapa., San Rafael Atlixco 186, Vicentina, Delegación Iztapalapa, C.P. 09340, Ciudad de México, México
| | - F J Alarcón-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de CBS, UAM-Iztapalapa, San Rafael Atlixco 186, Vicentina, Delegación Iztapalapa, C.P. 09340, Ciudad de México, México
| | - J C Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de CBS, UAM-Iztapalapa, San Rafael Atlixco 186, Vicentina, Delegación Iztapalapa, C.P. 09340, Ciudad de México, México
| |
Collapse
|
106
|
Vaja R, Chan JSK, Ferreira P, Harky A, Rogers LJ, Gashaw HH, Kirkby NS, Mitchell JA. The COVID-19 ibuprofen controversy: A systematic review of NSAIDs in adult acute lower respiratory tract infections. Br J Clin Pharmacol 2020; 87:776-784. [PMID: 32805057 DOI: 10.1111/bcp.14514] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 12/22/2022] Open
Abstract
AIMS In light of the recent safety concerns relating to NSAID use in COVID-19, we sought to evaluate cardiovascular and respiratory complications in patients taking NSAIDs during acute lower respiratory tract infections. METHODS We carried out a systematic review of randomised controlled trials and observational studies. Studies of adult patients with short-term NSAID use during acute lower respiratory tract infections, including bacterial and viral infections, were included. Primary outcome was all-cause mortality. Secondary outcomes were cardiovascular, renal and respiratory complications. RESULTS In total, eight studies including two randomised controlled trials, three retrospective and three prospective observational studies enrolling 44 140 patients were included. Five of the studies were in patients with pneumonia, two in patients with influenza, and one in a patient with acute bronchitis. Meta-analysis was not possible due to significant heterogeneity. There was a trend towards a reduction in mortality and an increase in pleuro-pulmonary complications. However, all studies exhibited high risks of bias, primarily due to lack of adjustment for confounding variables. Cardiovascular outcomes were not reported by any of the included studies. CONCLUSION In this systematic review of NSAID use during acute lower respiratory tract infections in adults, we found that the existing evidence for mortality, pleuro-pulmonary complications and rates of mechanical ventilation or organ failure is of extremely poor quality, very low certainty and should be interpreted with caution. Mechanistic and clinical studies addressing the captioned subject are urgently needed, especially in relation to COVID-19.
Collapse
Affiliation(s)
- Ricky Vaja
- Imperial College London, National Heart & Lung Institute, London, UK.,Royal Brompton Hospital, London, UK
| | - Jeffrey Shi Kai Chan
- Division of Cardiology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong.,Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Plinio Ferreira
- Imperial College London, National Heart & Lung Institute, London, UK
| | - Amer Harky
- Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Luke J Rogers
- University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Hime H Gashaw
- Imperial College London, National Heart & Lung Institute, London, UK
| | - Nicholas S Kirkby
- Imperial College London, National Heart & Lung Institute, London, UK
| | - Jane A Mitchell
- Imperial College London, National Heart & Lung Institute, London, UK
| |
Collapse
|
107
|
Protective Effects of Polyphenols against Ischemia/Reperfusion Injury. Molecules 2020; 25:molecules25153469. [PMID: 32751587 PMCID: PMC7435883 DOI: 10.3390/molecules25153469] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is a leading cause of morbidity and mortality across the world. It manifests as an imbalance between blood demand and blood delivery in the myocardium, which leads to cardiac ischemia and myocardial necrosis. While it is not easy to identify the first pathogenic cause of MI, the consequences are characterized by ischemia, chronic inflammation, and tissue degeneration. A poor MI prognosis is associated with extensive cardiac remodeling. A loss of viable cardiomyocytes is replaced with fibrosis, which reduces heart contractility and heart function. Recent advances have given rise to the concept of natural polyphenols. These bioactive compounds have been studied for their pharmacological properties and have proven successful in the treatment of cardiovascular diseases. Studies have focused on their various bioactivities, such as their antioxidant and anti-inflammatory effects and free radical scavenging. In this review, we summarized the effects and benefits of polyphenols on the cardiovascular injury, particularly on the treatment of myocardial infarction in animal and human studies.
Collapse
|
108
|
Haesen S, Cöl Ü, Schurgers W, Evens L, Verboven M, Driesen RB, Bronckaers A, Lambrichts I, Deluyker D, Bito V. Glycolaldehyde-modified proteins cause adverse functional and structural aortic remodeling leading to cardiac pressure overload. Sci Rep 2020; 10:12220. [PMID: 32699285 PMCID: PMC7376068 DOI: 10.1038/s41598-020-68974-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the role of advanced glycation end products (AGEs) in the development of diabetic vascular complications and cardiovascular diseases (CVDs). We have shown that high-molecular-weight AGEs (HMW-AGEs), present in our Western diet, impair cardiac function. Whether HMW-AGEs affect vascular function remains unknown. In this study, we aimed to investigate the impact of chronic HMW-AGEs exposure on vascular function and structure. Adult male Sprague Dawley rats were daily injected with HMW-AGEs or control solution for 6 weeks. HMW-AGEs animals showed intracardiac pressure overload, characterized by increased systolic and mean pressures. The contraction response to PE was increased in aortic rings from the HMW-AGEs group. Relaxation in response to ACh, but not SNP, was impaired by HMW-AGEs. This was associated with reduced plasma cyclic GMP levels. SOD restored ACh-induced relaxation of HMW-AGEs animals to control levels, accompanied by a reduced half-maximal effective dose (EC50). Finally, collagen deposition and intima-media thickness of the aortic vessel wall were increased with HMW-AGEs. Our data demonstrate that chronic HMW-AGEs exposure causes adverse vascular remodelling. This is characterised by disturbed vasomotor function due to increased oxidative stress and structural changes in the aorta, suggesting an important contribution of HMW-AGEs in the development of CVDs.
Collapse
Affiliation(s)
- Sibren Haesen
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Ümare Cöl
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Wouter Schurgers
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Lize Evens
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Maxim Verboven
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Ronald B Driesen
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Ivo Lambrichts
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Dorien Deluyker
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Virginie Bito
- Biomedical Research Institute (BIOMED), Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium.
| |
Collapse
|
109
|
Abstract
LINKED ARTICLES This article is part of a themed section on Eicosanoids 35 years from the 1982 Nobel: where are we now? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.8/issuetoc.
Collapse
Affiliation(s)
- Roderick J Flower
- The William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
110
|
Kang NH, Mukherjee S, Jang MH, Pham HG, Choi M, Yun JW. Ketoprofen alleviates diet-induced obesity and promotes white fat browning in mice via the activation of COX-2 through mTORC1-p38 signaling pathway. Pflugers Arch 2020; 472:583-596. [DOI: 10.1007/s00424-020-02380-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/05/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022]
|
111
|
Mason RP, Libby P, Bhatt DL. Emerging Mechanisms of Cardiovascular Protection for the Omega-3 Fatty Acid Eicosapentaenoic Acid. Arterioscler Thromb Vasc Biol 2020; 40:1135-1147. [PMID: 32212849 PMCID: PMC7176343 DOI: 10.1161/atvbaha.119.313286] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Abstract
Patients with well-controlled LDL (low-density lipoprotein) levels still have residual cardiovascular risk associated with elevated triglycerides. Epidemiological studies have shown that elevated fasting triglyceride levels associate independently with incident cardiovascular events, and abundant recent human genetic data support the causality of TGRLs (triglyceride-rich lipoproteins) in atherothrombosis. Omega-3 fatty acids, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), lower blood triglyceride concentrations but likely exert additional atheroprotective properties at higher doses. Omega-3 fatty acids modulate T-cell differentiation and give rise to various prostaglandins and specialized proresolving lipid mediators that promote resolution of tissue injury and inflammation. The REDUCE-IT (Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial) with an EPA-only formulation lowered a composite of cardiovascular events by 25% in patients with established cardiovascular disease or diabetes mellitus and other cardiovascular risk factors. This clinical benefit likely arises from multiple molecular mechanisms discussed in this review. Indeed, human plaques readily incorporate EPA, which may render them less likely to trigger clinical events. EPA and DHA differ in their effects on membrane structure, rates of lipid oxidation, inflammatory biomarkers, and endothelial function as well as tissue distributions. Trials that have evaluated DHA-containing high-dose omega-3 fatty acids have thus far not shown the benefits of EPA alone demonstrated in REDUCE-IT. This review will consider the mechanistic evidence that helps to understand the potential mechanisms of benefit of EPA.
Collapse
Affiliation(s)
- R. Preston Mason
- From the Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (R.P.M., P.L., D.L.B.)
- Elucida Research LLC, Beverly, MA (R.P.M.)
| | - Peter Libby
- From the Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (R.P.M., P.L., D.L.B.)
| | - Deepak L. Bhatt
- From the Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (R.P.M., P.L., D.L.B.)
| |
Collapse
|
112
|
Vascular Lipidomic Profiling of Potential Endogenous Fatty Acid PPAR Ligands Reveals the Coronary Artery as Major Producer of CYP450-Derived Epoxy Fatty Acids. Cells 2020; 9:cells9051096. [PMID: 32365470 PMCID: PMC7290345 DOI: 10.3390/cells9051096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 01/08/2023] Open
Abstract
A number of oxylipins have been described as endogenous PPAR ligands. The very short biological half-lives of oxylipins suggest roles as autocrine or paracrine signaling molecules. While coronary arterial atherosclerosis is the root of myocardial infarction, aortic atherosclerotic plaque formation is a common readout of in vivo atherosclerosis studies in mice. Improved understanding of the compartmentalized sources of oxylipin PPAR ligands will increase our knowledge of the roles of PPAR signaling in diverse vascular tissues. Here, we performed a targeted lipidomic analysis of ex vivo-generated oxylipins from porcine aorta, coronary artery, pulmonary artery and perivascular adipose. Cyclooxygenase (COX)-derived prostanoids were the most abundant detectable oxylipin from all tissues. By contrast, the coronary artery produced significantly higher levels of oxylipins from CYP450 pathways than other tissues. The TLR4 ligand LPS induced prostanoid formation in all vascular tissue tested. The 11-HETE, 15-HETE, and 9-HODE were also induced by LPS from the aorta and pulmonary artery but not coronary artery. Epoxy fatty acid (EpFA) formation was largely unaffected by LPS. The pig CYP2J homologue CYP2J34 was expressed in porcine vascular tissue and primary coronary artery smooth muscle cells (pCASMCs) in culture. Treatment of pCASMCs with LPS induced a robust profile of pro-inflammatory target genes: TNFα, ICAM-1, VCAM-1, MCP-1 and CD40L. The soluble epoxide hydrolase inhibitor TPPU, which prevents the breakdown of endogenous CYP-derived EpFAs, significantly suppressed LPS-induced inflammatory target genes. In conclusion, PPAR-activating oxylipins are produced and regulated in a vascular site-specific manner. The CYP450 pathway is highly active in the coronary artery and capable of providing anti-inflammatory oxylipins that prevent processes of inflammatory vascular disease progression.
Collapse
|
113
|
Gottesfeld-Hohler Memorial Foundation Risk Assessment for Early-Onset Preeclampsia in the United States: Think Tank Summary. Obstet Gynecol 2020; 135:36-45. [PMID: 31809427 DOI: 10.1097/aog.0000000000003582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Preeclampsia is responsible for significant maternal and neonatal morbidity and is associated with a substantial economic burden. Aspirin has been shown to be effective in decreasing the risk of preterm preeclampsia; however, there is no consensus on the target population for aspirin prophylaxis. In May 2018, the Gottesfeld-Hohler Memorial Foundation organized a working group meeting with the goal of identifying the optimal preeclampsia risk-assessment strategy and consequent intervention in the United States. The meeting brought together experts from the leading professional societies. We discussed available literature and trends in preeclampsia risk assessment, current professional guidelines for identifying women at risk for preeclampsia, prophylactic use of aspirin in the United States and Europe, cost-effectiveness data, and feasibility of implementation of different assessment tools and preventive strategies in the United States. We identified specific knowledge gaps and future research directions in preeclampsia risk assessment and prevention that need to be addressed before practice change.
Collapse
|
114
|
Tzoumas N, Farrah TE, Dhaun N, Webb DJ. Established and emerging therapeutic uses of PDE type 5 inhibitors in cardiovascular disease. Br J Pharmacol 2020; 177:5467-5488. [PMID: 31721165 PMCID: PMC7707100 DOI: 10.1111/bph.14920] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
PDE type 5 inhibitors (PDE5Is), such as sildenafil, tadalafil and vardenafil, are a class of drugs used to prolong the physiological effects of NO/cGMP signalling in tissues through the inhibition of cGMP degradation. Although these agents were originally developed for the treatment of hypertension and angina, unanticipated side effects led to advances in the treatment of erectile dysfunction and, later, pulmonary arterial hypertension. In the last decade, accumulating evidence suggests that PDE5Is may confer a wider range of clinical benefits than was previously recognised. This has led to a broader interest in the cardiovascular therapeutic potential of PDE5Is, in conditions such as hypertension, myocardial infarction, stroke, peripheral arterial disease, chronic kidney disease and diabetes mellitus. Here, we review the pharmacological properties and established licensed uses of this class of drug, along with emerging therapeutic developments and possible future indications.
Collapse
Affiliation(s)
- Nikolaos Tzoumas
- British Heart Foundation/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Tariq E Farrah
- British Heart Foundation/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neeraj Dhaun
- British Heart Foundation/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David J Webb
- British Heart Foundation/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
115
|
Abstract
The effect of dietary fats on cardiometabolic diseases, including cardiovascular diseases and type 2 diabetes mellitus, has generated tremendous interest. Many earlier investigations focused on total fat and conventional fat classes (such as saturated and unsaturated fats) and their influence on a limited number of risk factors. However, dietary fats comprise heterogeneous molecules with diverse structures, and growing research in the past two decades supports correspondingly complex health effects of individual dietary fats. Moreover, health effects of dietary fats might be modified by additional factors, such as accompanying nutrients and food-processing methods, emphasizing the importance of the food sources. Accordingly, the rapidly increasing scientific findings on dietary fats and cardiometabolic diseases have generated debate among scientists, caused confusion for the general public and present challenges for translation into dietary advice and policies. This Review summarizes the evidence on the effects of different dietary fats and their food sources on cell function and on risk factors and clinical events of cardiometabolic diseases. The aim is not to provide an exhaustive review but rather to focus on the most important evidence from randomized controlled trials and prospective cohort studies and to highlight current areas of controversy and the most relevant future research directions for understanding how to improve the prevention and management of cardiometabolic diseases through optimization of dietary fat intake.
Collapse
|
116
|
Ahmetaj-Shala B, Kawai R, Marei I, Nikolakopoulou Z, Shih CC, Konain B, Reed DM, Mongey R, Kirkby NS, Mitchell JA. A bioassay system of autologous human endothelial, smooth muscle cells, and leukocytes for use in drug discovery, phenotyping, and tissue engineering. FASEB J 2019; 34:1745-1754. [PMID: 31914612 PMCID: PMC6972557 DOI: 10.1096/fj.201901379rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 01/02/2023]
Abstract
Blood vessels are comprised of endothelial and smooth muscle cells. Obtaining both types of cells from vessels of living donors is not possible without invasive surgery. To address this, we have devised a strategy whereby human endothelial and smooth muscle cells derived from blood progenitors from the same donor could be cultured with autologous leukocytes to generate a same donor “vessel in a dish” bioassay. Autologous sets of blood outgrowth endothelial cells (BOECs), smooth muscle cells (BO‐SMCs), and leukocytes were obtained from four donors. Cells were treated in monoculture and cumulative coculture conditions. The endothelial specific mediator endothelin‐1 along with interleukin (IL)‐6, IL‐8, tumor necrosis factor α, and interferon gamma‐induced protein 10 were measured under control culture conditions and after stimulation with cytokines. Cocultures remained viable throughout. The profile of individual mediators released from cells was consistent with what we know of endothelial and smooth muscle cells cultured from blood vessels. For the first time, we report a proof of concept study where autologous blood outgrowth “vascular” cells and leukocytes were studied alone and in coculture. This novel bioassay has usefulness in vascular biology research, patient phenotyping, drug testing, and tissue engineering.
Collapse
Affiliation(s)
- Blerina Ahmetaj-Shala
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Ryota Kawai
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK.,Medicinal Safety Research Laboratories, Daiichi-Sankyo Co. Ltd., Tokyo, Japan
| | - Isra Marei
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK.,Qatar Foundation Research and Development Division, Doha, Qatar
| | - Zacharoula Nikolakopoulou
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK.,Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Chih-Chin Shih
- Department of Pharmacology, National Defense Medical Center, Taipei, R.O.C., Taiwan
| | - Bhatti Konain
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Daniel M Reed
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Róisín Mongey
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicholas S Kirkby
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jane A Mitchell
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
117
|
Arauna D, Furrianca M, Espinosa-Parrilla Y, Fuentes E, Alarcón M, Palomo I. Natural Bioactive Compounds As Protectors Of Mitochondrial Dysfunction In Cardiovascular Diseases And Aging. Molecules 2019; 24:molecules24234259. [PMID: 31766727 PMCID: PMC6930637 DOI: 10.3390/molecules24234259] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/05/2019] [Accepted: 09/08/2019] [Indexed: 01/04/2023] Open
Abstract
Diet, particularly the Mediterranean diet, has been considered as a protective factor against the development of cardiovascular diseases, the main cause of death in the world. Aging is one of the major risk factors for cardiovascular diseases, which have an oxidative pathophysiological component, being the mitochondria one of the key organelles in the regulation of oxidative stress. Certain natural bioactive compounds have the ability to regulate oxidative phosphorylation, the production of reactive oxygen species and the expression of mitochondrial proteins; but their efficacy within the mitochondrial physiopathology of cardiovascular diseases has not been clarified yet. The following review has the purpose of evaluating several natural compounds with evidence of mitochondrial effect in cardiovascular disease models, ascertaining the main cellular mechanisms and their potential use as functional foods for prevention of cardiovascular disease and healthy aging.
Collapse
Affiliation(s)
- Diego Arauna
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
| | - María Furrianca
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Departamento de enfermería, Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Yolanda Espinosa-Parrilla
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Laboratory of Molecular Medicine —LMM, Center for Education, Healthcare and Investigation—CADI, Universidad de Magallanes, Punta Arenas 6200000, Chile
- School of Medicine, Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Correspondence: (E.F.); (I.P.)
| | - Marcelo Alarcón
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Correspondence: (E.F.); (I.P.)
| |
Collapse
|
118
|
Pyrikova NV, Antropova ON, Osipova IV. Adverse Reactions of the Cardiovascular System when Taking Nonsteroidal Anti-inflammatory Drugs and Ways to Reduce Them. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2019. [DOI: 10.20996/1819-6446-2019-15-5-750-758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The most important issue of modern pharmacotherapy is not only efficacy, but also the safety of medicines. The use of nonsteroidal anti-inflammatory drugs (NSAIDs) is one of the main methods of treating acute and chronic pain in a wide range of diseases and pathological conditions. However, the prescription of this group of drugs requires consideration of the potential risks of complications, including from the side of the cardiovascular system. The purpose of the review was to assess the adverse reactions of the cardiovascular system when taking NSAIDs and approaches to their reduction. The article presents data on the mutual potential impact of cardiovascular diseases and musculoskeletal system, presents the results of large-scale studies of Russian and foreign authors and meta-analyzes of the NSAIDs effect on blood pressure profile, development of myocardial infarction, stroke and heart failure. The possible pathogenetic mechanisms of the side effects of NSAIDs are reviewed; the complexity of managing comorbid patients is demonstrated; it is shown that symptomatic treatment of pain and inflammatory syndrome should be carried out considering a personalized approach to the patient and rational choice of drugs.Before the NSAIDs prescription, it is necessary to consider all cardiovascular risk factors with the determination of the total risk of cardiovascular complications. In patients with a very high cardiovascular risk, the use of any NSAIDs should be avoided; with high and moderate risk, the use of NSAIDs with the most favorable cardiovascular safety profile is possible. If the patient belongs to the category of low total coronary risk, the doctor can choose any NSAIDs.
Collapse
|
119
|
Yang J, Bruun DA, Wang C, Wan D, McReynolds CB, Phu K, Inceoglu B, Lein PJ, Hammock BD. Lipidomes of brain from rats acutely intoxicated with diisopropylfluorophosphate identifies potential therapeutic targets. Toxicol Appl Pharmacol 2019; 382:114749. [PMID: 31521729 PMCID: PMC6957308 DOI: 10.1016/j.taap.2019.114749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/30/2019] [Accepted: 09/07/2019] [Indexed: 01/28/2023]
Abstract
Organophosphates (OPs), a class of phosphorus-containing chemicals that act by disrupting cholinergic transmission, include both toxic and fast-acting chemical warfare agents as well as less toxic but more easily accessible OP pesticides. The classical atropine/2-PAM antidote fails to protect against long-term symptoms following acute intoxication with OPs at levels that trigger status epilepticus. Acute OP intoxication also causes a robust neuroinflammatory response, which is implicated in the pathogenesis of long-term effects. In this study, we characterized the profiles of lipid mediators, important players in neuroinflammation, in the rat model of acute DFP intoxication. The profiles of lipid mediators were monitored in three different regions of the brain (cortex, hippocampus, and cerebellum) at 0, 1, 3, 7, 14, and 28 days post-exposure. The distribution pattern of lipid mediators was distinct in the three brain regions. In the cerebellum, the profile is dominated by LOX metabolites, while the lipid mediator profiles in cortex and hippocampus are dominated by COX metabolites followed by LOX and CYP 450 metabolites. Following acute DFP intoxication, most of the pro-inflammatory lipid mediators (e.g., PGD2 and PGE2) increased rapidly from day 1, while the concentrations of some anti-inflammatory lipid mediators (e.g. 14,15 EpETrE) decreased after DFP intoxication but recovered by day 14 post-exposure. The lipidomics results suggest two potential treatment targets: blocking the formation of prostaglandins by inhibiting COX and stabilizing the anti-inflammatory lipid mediators containing epoxides by inhibiting the enzyme soluble epoxide hydrolase (sEH).
Collapse
Affiliation(s)
- Jun Yang
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Donald A Bruun
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA
| | - Chang Wang
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA; School of Radiation Medicine and Protection, Medical College of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, PR China
| | - Debin Wan
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Cindy B McReynolds
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Kenny Phu
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Bora Inceoglu
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
120
|
Mitchell JA, Shala F, Elghazouli Y, Warner TD, Gaston-Massuet C, Crescente M, Armstrong PC, Herschman HR, Kirkby NS. Cell-Specific Gene Deletion Reveals the Antithrombotic Function of COX1 and Explains the Vascular COX1/Prostacyclin Paradox. Circ Res 2019; 125:847-854. [PMID: 31510878 PMCID: PMC6791564 DOI: 10.1161/circresaha.119.314927] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Supplemental Digital Content is available in the text. Endothelial cells (ECs) and platelets, which respectively produce antithrombotic prostacyclin and prothrombotic thromboxane A2, both express COX1 (cyclooxygenase1). Consequently, there has been no way to delineate any antithrombotic role for COX1-derived prostacyclin from the prothrombotic effects of platelet COX1. By contrast, an antithrombotic role for COX2, which is absent in platelets, is straightforward to demonstrate. This has resulted in an incomplete understanding of the relative importance of COX1 versus COX2 in prostacyclin production and antithrombotic protection in vivo.
Collapse
Affiliation(s)
- Jane A Mitchell
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., F.S., Y.E., N.S.K.)
| | - Fisnik Shala
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., F.S., Y.E., N.S.K.)
| | - Youssef Elghazouli
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., F.S., Y.E., N.S.K.)
| | - Timothy D Warner
- Blizard Institute (T.D.W., M.C., P.C.A.), Queen Mary University of London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute (C.G.-M.), Queen Mary University of London, United Kingdom
| | - Marilena Crescente
- Blizard Institute (T.D.W., M.C., P.C.A.), Queen Mary University of London, United Kingdom
| | - Paul C Armstrong
- Blizard Institute (T.D.W., M.C., P.C.A.), Queen Mary University of London, United Kingdom
| | - Harvey R Herschman
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (H.R.H.)
| | - Nicholas S Kirkby
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., F.S., Y.E., N.S.K.)
| |
Collapse
|
121
|
Parainfluenza virus infection enhances NSAIDs-induced inhibition of PGE2 generation and COX-2 expression in human airway epithelial cells. Adv Med Sci 2019; 64:338-343. [PMID: 31022559 DOI: 10.1016/j.advms.2019.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/29/2019] [Accepted: 04/12/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Respiratory viral infection and nonsteroidal anti-inflammatory drugs (NSAIDs) may affect arachidonic acid (AA) metabolism in the airway epithelium, however their joint effect has not been studied. We hypothesized, that alternations of AA metabolism in human airway epithelial cells (ECs) - induced by Parainfluenza virus type 3 (PIV3) - may be modified by concomitant treatment with NSAIDs. MATERIALS AND METHODS Nasal (RPMI 2650) and bronchial (BEAS-2B) epithelial cells were cultured into confluence and then infected with PIV3. Prostaglandin E2 (PGE2) and 15-hydroxyeicosatetraenoic acid (15-HETE) levels in cell supernatants were measured by ELISA and expression of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), 5-lipoxygenase (5-LO) and 15-lipoxygenase (15-LO) mRNA in cells was evaluated after reverse transcription with real-time polymerase chain reactions. RESULTS PGE2 generation was decreased by PIV3 infection in the upper airway epithelial cells, and increased in the lower airway epithelial cells. Both naproxen and celecoxib induced significant reduction in PGE2 release in both infected and non-infected upper and lower airway epithelial cells. However, in PIV3-infected epithelial cells celecoxib inhibited PGE2 release and COX-2 expression to significantly higher degree as compared to non-infected cells. 15-HETE generation or COX-1, 5-LO and 15-LO expression were not affected by the virus infection or by NSAIDs. CONCLUSION Virus infection in airway epithelial cells enhances inhibitory effect of NSAIDs on prostaglandin E2 generation.
Collapse
|
122
|
Wang HR, Sui HC, Zhu BT. Ellagic acid, a plant phenolic compound, activates cyclooxygenase-mediated prostaglandin production. Exp Ther Med 2019; 18:987-996. [PMID: 31316596 PMCID: PMC6601391 DOI: 10.3892/etm.2019.7667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022] Open
Abstract
In recent years, ellagic acid (EA), a naturally-occurring phenolic compound richly contained in some of the human food sources such as Longan and Litchi, was reported to have a number of biological effects. Based on our earlier 3D-QSAR/CoMFA models for cyclooxygenase (COX) I and II, we hypothesize that EA may have the potential to modulate the catalytic activity of COX enzymes, and this hypothesis is examined in the present study. The results from both in vitro and in vivo experiments show that EA is an activator of COX enzyme-catalyzed production of prostaglandin E2, a representative prostaglandin tested. Mechanistically, EA can activate the peroxidase active site of COX enzymes by serving as a co-substrate, presumably for the reduction of protoporphorin IX with FeIV inside. The effect of EA is abrogated by the co-presence of galangin, which is known to bind to COX's peroxidase active site and thereby blocks the effect of the reducing co-substrates. In view of the known physiological functions of COX enzymes in the body, it is suggested that some of the pharmacological and/or toxicological effects of EA may result from an increased production of certain prostaglandins and their related derivatives in the body.
Collapse
Affiliation(s)
- Hui Rong Wang
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P.R. China
| | - Hao Chen Sui
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Bao Ting Zhu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P.R. China
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
123
|
Marklund M, Wu JHY, Imamura F, Del Gobbo LC, Fretts A, de Goede J, Shi P, Tintle N, Wennberg M, Aslibekyan S, Chen TA, de Oliveira Otto MC, Hirakawa Y, Eriksen HH, Kröger J, Laguzzi F, Lankinen M, Murphy RA, Prem K, Samieri C, Virtanen J, Wood AC, Wong K, Yang WS, Zhou X, Baylin A, Boer JM, Brouwer IA, Campos H, Chaves PHM, Chien KL, de Faire U, Djoussé L, Eiriksdottir G, El-Abbadi N, Forouhi NG, Gaziano JM, Geleijnse JM, Gigante B, Giles G, Guallar E, Gudnason V, Harris T, Harris WS, Helmer C, Hellenius ML, Hodge A, Hu FB, Jacques PF, Jansson JH, Kalsbeek A, Khaw KT, Koh WP, Laakso M, Leander K, Hung-Ju Lin, Lind L, Luben R, Luo J, McKnight B, Mursu J, Ninomiya T, Overvad K, Psaty BM, Rimm E, Schulze MB, Siscovick D, Nielsen MS, Smith AV, Steffen BT, Steffen L, Sun Q, Sundström J, Tsai MY, Tunstall-Pedoe H, Uusitupa MIJ, van Dam RM, Veenstra J, Verschuren WM, Wareham N, Willett W, Woodward M, Yuan JM, Micha R, Lemaitre RN, Mozaffarian D. Biomarkers of Dietary Omega-6 Fatty Acids and Incident Cardiovascular Disease and Mortality. Circulation 2019; 139:2422-2436. [PMID: 30971107 PMCID: PMC6582360 DOI: 10.1161/circulationaha.118.038908] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Global dietary recommendations for and cardiovascular effects of linoleic acid, the major dietary omega-6 fatty acid, and its major metabolite, arachidonic acid, remain controversial. To address this uncertainty and inform international recommendations, we evaluated how in vivo circulating and tissue levels of linoleic acid (LA) and arachidonic acid (AA) relate to incident cardiovascular disease (CVD) across multiple international studies. METHODS We performed harmonized, de novo, individual-level analyses in a global consortium of 30 prospective observational studies from 13 countries. Multivariable-adjusted associations of circulating and adipose tissue LA and AA biomarkers with incident total CVD and subtypes (coronary heart disease, ischemic stroke, cardiovascular mortality) were investigated according to a prespecified analytic plan. Levels of LA and AA, measured as the percentage of total fatty acids, were evaluated linearly according to their interquintile range (ie, the range between the midpoint of the first and fifth quintiles), and categorically by quintiles. Study-specific results were pooled using inverse-variance-weighted meta-analysis. Heterogeneity was explored by age, sex, race, diabetes mellitus, statin use, aspirin use, omega-3 levels, and fatty acid desaturase 1 genotype (when available). RESULTS In 30 prospective studies with medians of follow-up ranging 2.5 to 31.9 years, 15 198 incident cardiovascular events occurred among 68 659 participants. Higher levels of LA were significantly associated with lower risks of total CVD, cardiovascular mortality, and ischemic stroke, with hazard ratios per interquintile range of 0.93 (95% CI, 0.88-0.99), 0.78 (0.70-0.85), and 0.88 (0.79-0.98), respectively, and nonsignificantly with lower coronary heart disease risk (0.94; 0.88-1.00). Relationships were similar for LA evaluated across quintiles. AA levels were not associated with higher risk of cardiovascular outcomes; in a comparison of extreme quintiles, higher levels were associated with lower risk of total CVD (0.92; 0.86-0.99). No consistent heterogeneity by population subgroups was identified in the observed relationships. CONCLUSIONS In pooled global analyses, higher in vivo circulating and tissue levels of LA and possibly AA were associated with lower risk of major cardiovascular events. These results support a favorable role for LA in CVD prevention.
Collapse
Affiliation(s)
- Matti Marklund
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Sweden
- The George Institute for Global Health and the Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jason HY Wu
- The George Institute for Global Health and the Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Fumiaki Imamura
- Medical Research Council Epidemiology Unit, University of Cambridge, United Kingdom
| | - Liana C. Del Gobbo
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA
| | - Amanda Fretts
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle
| | - Janette de Goede
- Division of Human Nutrition, Wageningen University, The Netherlands
| | - Peilin Shi
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Nathan Tintle
- Department of Mathematics and Statistics, Dordt College, Sioux Centre, IA
| | - Maria Wennberg
- Department of Public Health and Clinical Medicine, Nutritional Research, Umeå University, Sweden
| | | | - Tzu-An Chen
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Marcia C. de Oliveira Otto
- Division of Epidemiology, Human Genetics and Environmental Sciences, the University of Texas Health Science Center, School of Public Health, Houston
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Janine Kröger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Federica Laguzzi
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Lankinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio
| | - Rachel A. Murphy
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiesha Prem
- Saw Swee Hock School of Public Health, National University of Singapore
| | - Cécilia Samieri
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, TUMR 1219, France
| | - Jyrki Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio
| | - Alexis C. Wood
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Kerry Wong
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Australia
| | - Wei-Sin Yang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei
| | - Xia Zhou
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis
| | - Ana Baylin
- Departments of Nutritional Sciences and Epidemiology, School of Public Health, University of Michigan, Ann Arbor
| | - Jolanda M.A. Boer
- Centre for Nutrition, Prevention and Health Services, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | | | - Hannia Campos
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Paulo H. M. Chaves
- Benjamin Leon for Geriatrics Research and Education, Herbert Wertheim College of Medicine, Florida International University, Miami
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Luc Djoussé
- Brigham and Women's Hospital, Boston Veterans Affairs Healthcare System, MA
| | - Gudny Eiriksdottir
- Icelandic Heart Association, Kópavogur, Iceland; and Faculty of Medicine, University of Iceland, Reykjavik
| | - Naglaa El-Abbadi
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- USDA Jean Mayer Human Nutrition Research Center, Boston, MA
| | - Nita G. Forouhi
- Medical Research Council Epidemiology Unit, University of Cambridge, United Kingdom
| | - J. Michael Gaziano
- Brigham and Women's Hospital, Boston Veterans Affairs Healthcare System, MA
| | | | - Bruna Gigante
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Graham Giles
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Australia
| | - Eliseo Guallar
- Division of Environmental Epidemiology, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur, Iceland; and Faculty of Medicine, University of Iceland, Reykjavik
| | | | - William S. Harris
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls
- OmegaQuant Analytics, LLC, Sioux Falls, SD
| | - Catherine Helmer
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, TUMR 1219, France
| | - Mai-Lis Hellenius
- Department of Medicine, Cardiology Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Allison Hodge
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Australia
| | - Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Paul F. Jacques
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- USDA Jean Mayer Human Nutrition Research Center, Boston, MA
| | - Jan-Håkan Jansson
- Department of Public Health and Clinical Medicine, Research Unit Skellefteå, Umeå University, Umeå, Sweden
| | - Anya Kalsbeek
- Department of Mathematics and Statistics, Dordt College, Sioux Centre, IA
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge School of Clinical Medicine, United Kingdom
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore
- Duke-NUS Medical School, Singapore
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hung-Ju Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Sweden
| | - Robert Luben
- Department of Public Health and Primary Care, University of Cambridge School of Clinical Medicine, United Kingdom
| | - Juhua Luo
- Department of Epidemiology and Biostatistics, Indiana University, Bloomington
| | - Barbara McKnight
- Department of Biostatistics, School of Public Health, University of Washington, Seattle
| | - Jaakko Mursu
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Denmark
- Department of Cardiology, Aalborg University Hospital, Denmark
| | - Bruce M. Psaty
- Cardiovascular Health Study, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle
- Kaiser Permanente Washington Health Research Institute, Seattle
| | - Eric Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | | | | | - Albert V. Smith
- Icelandic Heart Association, Kópavogur, Iceland; and Faculty of Medicine, University of Iceland, Reykjavik
| | - Brian T. Steffen
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Lyn Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Hugh Tunstall-Pedoe
- Cardiovascular Epidemiology Unit, Institute of Cardiovascular Research, University of Dundee, United Kingdom
| | - Matti I. J. Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore
| | - Jenna Veenstra
- Department of Mathematics and Statistics, Dordt College, Sioux Centre, IA
| | - W.M. Monique Verschuren
- Centre for Nutrition, Prevention and Health Services, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Nick Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, United Kingdom
| | - Walter Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Mark Woodward
- The George Institute for Global Health and the Faculty of Medicine, University of New South Wales, Sydney, Australia
- Cardiovascular Epidemiology Unit, Institute of Cardiovascular Research, University of Dundee, United Kingdom
- The George Institute for Global Health, University of Oxford, United Kingdom
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer, and Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, PA
| | - Renata Micha
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | | |
Collapse
|
124
|
Mitchell JA, Kirkby NS. Eicosanoids, prostacyclin and cyclooxygenase in the cardiovascular system. Br J Pharmacol 2019; 176:1038-1050. [PMID: 29468666 PMCID: PMC6451069 DOI: 10.1111/bph.14167] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/19/2018] [Accepted: 01/29/2018] [Indexed: 12/30/2022] Open
Abstract
Eicosanoids represent a diverse family of lipid mediators with fundamental roles in physiology and disease. Within the eicosanoid superfamily are prostanoids, which are specifically derived from arachidonic acid by the enzyme cyclooxygenase (COX). COX has two isoforms; COX-1 and COX-2. COX-2 is the therapeutic target for the nonsteroidal anti-inflammatory drug (NSAID) class of pain medications. Of the prostanoids, prostacyclin, first discovered by Sir John Vane in 1976, remains amongst the best studied and retains an impressive pedigree as one of the fundamental cardiovascular protective pathways. Since this time, we have learnt much about how eicosanoids, COX enzymes and prostacyclin function in the cardiovascular system, knowledge that has allowed us, for example, to harness the power of prostacyclin as therapy to treat pulmonary arterial hypertension and peripheral vascular disease. However, there remain many unanswered questions in our basic understanding of the pathways, and how they can be used to improve human health. Perhaps, the most important and controversial outstanding question in the field remains; 'how do NSAIDs produce their much publicized cardiovascular side-effects?' This review summarizes the history, biology and cardiovascular function of key eicosanoids with particular focus on prostacyclin and other COX products and discusses how our knowledge of these pathways can applied in future drug discovery and be used to explain the cardiovascular side-effects of NSAIDs. LINKED ARTICLES: This article is part of a themed section on Eicosanoids 35 years from the 1982 Nobel: where are we now? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.8/issuetoc.
Collapse
Affiliation(s)
- Jane A Mitchell
- Cardiothoracic PharmacologyNational Heart and Lung InstituteLondonUK
| | - Nicholas S Kirkby
- Cardiothoracic PharmacologyNational Heart and Lung InstituteLondonUK
| |
Collapse
|
125
|
Upchurch C, Leitinger N. Biologically Active Lipids in Vascular Biology. FUNDAMENTALS OF VASCULAR BIOLOGY 2019. [DOI: 10.1007/978-3-030-12270-6_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
126
|
Jaén RI, Prieto P, Casado M, Martín-Sanz P, Boscá L. Post-translational modifications of prostaglandin-endoperoxide synthase 2 in colorectal cancer: An update. World J Gastroenterol 2018; 24:5454-5461. [PMID: 30622375 PMCID: PMC6319129 DOI: 10.3748/wjg.v24.i48.5454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
The biosynthesis of prostanoids is involved in both physiological and pathological processes. The expression of prostaglandin-endoperoxide synthase 2 (PTGS2; also known as COX-2) has been traditionally associated to the onset of several pathologies, from inflammation to cardiovascular, gastrointestinal and oncologic events. For this reason, the search of selective PTGS2 inhibitors has been a focus for therapeutic interventions. In addition to the classic non-steroidal anti-inflammatory drugs, selective and specific PTGS2 inhibitors, termed coxibs, have been generated and widely used. PTGS2 activity is less restrictive in terms of substrate specificity than the homeostatic counterpart PTGS1, and it accounts for the elevated prostanoid synthesis that accompanies several pathologies. The main regulation of PTGS2 occurs at the transcription level. In addition to this, the stability of the mRNA is finely regulated through the interaction with several cytoplasmic elements, ranging from specific microRNAs to proteins that control mRNA degradation. Moreover, the protein has been recognized to be the substrate for several post-translational modifications that affect both the enzyme activity and the targeting for degradation via proteasomal and non-proteasomal mechanisms. Among these modifications, phosphorylation, glycosylation and covalent modifications by reactive lipidic intermediates and by free radicals associated to the pro-inflammatory condition appear to be the main changes. Identification of these post-translational modifications is relevant to better understand the role of PTGS2 in several pathologies and to establish a correct analysis of the potential function of this protein in diseases progress. Finally, these modifications can be used as biomarkers to establish correlations with other parameters, including the immunomodulation dependent on molecular pathological epidemiology determinants, which may provide a better frame for potential therapeutic interventions.
Collapse
Affiliation(s)
- Rafael I Jaén
- Department of Metabolism and Physiopathology of Inflammatory Diseases, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid 28029, Spain
| | - Patricia Prieto
- Department of Metabolism and Physiopathology of Inflammatory Diseases, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid 28029, Spain
| | - Marta Casado
- Department of Biomedicine, Instituto de Biomedicina de Valencia (CSIC), Valencia 46010, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, y Hepáticas y Digestivas, ISCIII, Madrid 28029, Spain
| | - Paloma Martín-Sanz
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, y Hepáticas y Digestivas, ISCIII, Madrid 28029, Spain
- Unidad Asociada IIBM-ULPGC, Universidad de las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria 35001, Spain
| | - Lisardo Boscá
- Department of Metabolism and Physiopathology of Inflammatory Diseases, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, y Hepáticas y Digestivas, ISCIII, Madrid 28029, Spain
- Unidad Asociada IIBM-ULPGC, Universidad de las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria 35001, Spain
| |
Collapse
|
127
|
Sonnweber T, Pizzini A, Nairz M, Weiss G, Tancevski I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2018; 19:ijms19113285. [PMID: 30360467 PMCID: PMC6274989 DOI: 10.3390/ijms19113285] [Citation(s) in RCA: 257] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Lipid and immune pathways are crucial in the pathophysiology of metabolic and cardiovascular disease. Arachidonic acid (AA) and its derivatives link nutrient metabolism to immunity and inflammation, thus holding a key role in the emergence and progression of frequent diseases such as obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. We herein present a synopsis of AA metabolism in human health, tissue homeostasis, and immunity, and explore the role of the AA metabolome in diverse pathophysiological conditions and diseases.
Collapse
Affiliation(s)
- Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
128
|
Wang HR, Sui HC, Ding YY, Zhu BT. Stimulation of the Production of Prostaglandin E₂ by Ethyl Gallate, a Natural Phenolic Compound Richly Contained in Longan. Biomolecules 2018; 8:E91. [PMID: 30200641 PMCID: PMC6165217 DOI: 10.3390/biom8030091] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/01/2018] [Accepted: 07/03/2018] [Indexed: 12/20/2022] Open
Abstract
Ethyl gallate is a phenolic compound richly contained in Longan. In traditional Chinese medicine, Longan is widely known as a fruit with "hot" properties, with a tendency to promote inflammatory and certain other responses. The mechanism for its proinflammatory as well as health beneficial effects is poorly understood. Based on our earlier observation that certain natural phenolic compounds can serve as reducing cosubstrates for cyclooxygenases (COXs), we sought to test a hypothesis that ethyl gallate may activate the catalytic activity of the COX enzymes. Results from studies using cultured cells and animals show that ethyl gallate can activate the production of prostaglandin E₂, a representative prostaglandin tested in this study. Computational analysis indicates that ethyl gallate can activate the peroxidase active sites of COX-1 and COX-2 by serving as a reducing cosubstrate. The effect of ethyl gallate is abrogated by galangin, which is known to bind to the same peroxidase active sites of COX-1 and COX-2 as a competitive inhibitor. The findings of this study offer support for a novel hypothesis that the proinflammatory as well as health beneficial effects of Longan may partly attributable to the activation of COX-1 and COX-2 by ethyl gallate.
Collapse
Affiliation(s)
- Hui Rong Wang
- Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Hao Chen Sui
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China.
| | - Yan Yan Ding
- Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Bao Ting Zhu
- Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China.
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China.
| |
Collapse
|
129
|
Strassheim D, Karoor V, Stenmark K, Verin A, Gerasimovskaya E. A current view of G protein-coupled receptor - mediated signaling in pulmonary hypertension: finding opportunities for therapeutic intervention. ACTA ACUST UNITED AC 2018; 2. [PMID: 31380505 PMCID: PMC6677404 DOI: 10.20517/2574-1209.2018.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathological vascular remodeling is observed in various cardiovascular diseases including pulmonary hypertension (PH), a disease of unknown etiology that has been characterized by pulmonary artery vasoconstriction, right ventricular hypertrophy, vascular inflammation, and abnormal angiogenesis in pulmonary circulation. G protein-coupled receptors (GPCRs) are the largest family in the genome and widely expressed in cardiovascular system. They regulate all aspects of PH pathophysiology and represent therapeutic targets. We overview GPCRs function in vasoconstriction, vasodilation, vascular inflammation-driven remodeling and describe signaling cross talk between GPCR, inflammatory cytokines, and growth factors. Overall, the goal of this review is to emphasize the importance of GPCRs as critical signal transducers and targets for drug development in PH.
Collapse
Affiliation(s)
- Derek Strassheim
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Vijaya Karoor
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.,Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| |
Collapse
|
130
|
Estrada-Figueroa LA, Díaz-Gandarilla JA, Hernández-Ramírez VI, Arrieta-González MM, Osorio-Trujillo C, Rosales-Encina JL, Toledo-Leyva A, Talamás-Rohana P. Leishmania mexicana gp63 is the enzyme responsible for cyclooxygenase (COX) activity in this parasitic protozoa. Biochimie 2018; 151:73-84. [DOI: 10.1016/j.biochi.2018.05.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 05/28/2018] [Indexed: 10/14/2022]
|
131
|
Crescente M, Menke L, Chan MV, Armstrong PC, Warner TD. Eicosanoids in platelets and the effect of their modulation by aspirin in the cardiovascular system (and beyond). Br J Pharmacol 2018; 176:988-999. [PMID: 29512148 PMCID: PMC6451075 DOI: 10.1111/bph.14196] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/21/2022] Open
Abstract
Platelets are important players in thrombosis and haemostasis with their function being modulated by mediators in the blood and the vascular wall. Among these, eicosanoids can both stimulate and inhibit platelet reactivity. Platelet Cyclooxygenase (COX)-1-generated Thromboxane (TX)A2 is the primary prostanoid that stimulates platelet aggregation; its action is counter-balanced by prostacyclin, a product of vascular COX. Prostaglandin (PG)D2 , PGE2 and 12-hydroxyeicosatraenoic acid (HETE), or 15-HETE, are other prostanoid modulators of platelet activity, but some also play a role in carcinogenesis. Aspirin permanently inhibits platelet COX-1, underlying its anti-thrombotic and anti-cancer action. While the use of aspirin as an anti-cancer drug is increasingly encouraged, its continued use in addition to P2 Y12 receptor antagonists for the treatment of cardiovascular diseases is currently debated. Aspirin not only suppresses TXA2 but also prevents the synthesis of both known and unknown antiplatelet eicosanoid pathways, potentially lessening the efficacy of dual antiplatelet therapies. LINKED ARTICLES: This article is part of a themed section on Eicosanoids 35 years from the 1982 Nobel: where are we now? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.8/issuetoc.
Collapse
Affiliation(s)
- Marilena Crescente
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laura Menke
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Melissa V Chan
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Paul C Armstrong
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Timothy D Warner
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|